DHX36 is a multifunctional ATP-dependent helicase that primarily unwinds G-quadruplex (G4) structures in nucleic acids. It plays critical roles in genomic integrity, gene expression regulation, and functions as a sensor to initiate antiviral responses . The protein binds with high affinity to and unwinds G4 structures formed in both DNA and RNA molecules (G4-DNA and G4-RNA), making it essential for numerous cellular processes . These G4 structures consist of helical arrangements containing guanine tetrads that DHX36 specifically targets and resolves.
DHX36 participates in several crucial biological processes:
Genomic integrity maintenance: Resolves G4 structures that could otherwise lead to genomic instability
Transcriptional regulation: Resolves G4-DNA structures in gene promoters (including YY1, KIT/c-kit, and ALPL) to positively regulate their expression
Post-transcriptional regulation: Unwinds G4-RNA structures in the 3'-UTR polyadenylation sites of pre-mRNAs, such as TP53, stimulating their 3'-end processing in response to DNA damage
Telomere maintenance: Converts G4-RNA structures in telomerase RNA template components into double-stranded RNA to promote P1 helix formation
MicroRNA processing: Binds to pre-miRNA terminal loops, regulating their transport and contributing to the control of dendritic spine morphogenesis
The rabbit polyclonal DHX36 antibody (ab70269) has been validated for multiple research applications, including:
Immunoprecipitation (IP)
Western Blotting (WB)
The antibody has been cited in 14 scientific publications, demonstrating its reliability and acceptance in the research community . It has been confirmed to react with human and mouse samples, making it suitable for comparative studies across these species.
When designing experiments to study DHX36 function, researchers must account for its high specificity for G-quadruplex structures. The unique binding affinity of DHX36 for G4 structures requires careful consideration of nucleic acid sequence contexts in experimental systems.
When analyzing DHX36 interactions with potential target sequences, researchers should:
Identify putative G-quadruplex forming sequences (G-rich regions with potential to form G-tetrads)
Confirm G4 structure formation using techniques such as circular dichroism spectroscopy
Analyze DHX36 binding and unwinding activity using both G4-forming and control (non-G4) sequences
Consider the impact of different cations (particularly potassium) on G4 stability during buffer preparation
Account for potential competition between DHX36 and other G4-binding proteins in cellular contexts
This specificity also offers opportunities for targeted experiments exploring the role of G4 structures in specific genomic contexts, such as promoter regions of YY1, KIT, and ALPL genes .
Distinguishing DHX36 activity on DNA versus RNA G-quadruplexes requires specialized experimental approaches:
Substrate specificity assays: Compare DHX36 binding affinity and unwinding rates between equivalent G4-DNA and G4-RNA sequences using purified recombinant protein and synthetic oligonucleotides.
Nuclease protection assays: Utilize DNA-specific (DNase I) or RNA-specific (RNase A) nucleases to selectively degrade non-G4 structures while protecting DHX36-bound substrates.
Cross-linking immunoprecipitation (CLIP) approaches:
CLIP-seq for RNA interactions
ChIP-seq for DNA interactions
These can be performed in parallel to map genome-wide DNA versus RNA interactions.
Cellular compartment analysis: Examine DHX36 distribution between nucleus (primarily DNA interactions) and cytoplasm (primarily RNA interactions) using fractionation followed by Western blotting.
Mutational analysis: Create DHX36 mutants with selective defects in either DNA or RNA G4 binding to distinguish substrate-specific activities.
DHX36's critical function in telomerase RNA processing positions it as a potential target in both aging and cancer research:
Cancer implications: DHX36 converts G4-RNA structures in telomerase RNA template components (TERC) into double-stranded RNA to promote P1 helix formation, ensuring accurate reverse transcription . Cancer cells typically upregulate telomerase to maintain telomere length and achieve replicative immortality. DHX36 inhibition could potentially disrupt this process by preventing proper TERC processing.
Aging research: Telomere shortening is a hallmark of cellular aging. DHX36's role in TERC processing may impact the efficiency of telomere maintenance in somatic cells. Researchers studying aging should consider DHX36 expression and activity when investigating telomere dynamics in senescent cells.
Experimental considerations: Researchers examining DHX36 in these contexts should:
Monitor telomere length using q-FISH or qPCR-based methods
Assess telomerase activity via TRAP assays
Examine TERC processing and structure using RNA structure probing techniques
Analyze correlations between DHX36 expression levels and telomere maintenance in various cell types
When using DHX36 antibody (ab70269) for Western blotting, researchers should consider the following technical parameters for optimal results:
Sample preparation:
Extract proteins using RIPA buffer supplemented with protease inhibitors
Include phosphatase inhibitors if studying phosphorylation-dependent activities of DHX36
Denature samples in Laemmli buffer with DTT at 95°C for 5 minutes
Gel electrophoresis:
Use 8-10% SDS-PAGE gels (DHX36 is approximately 110 kDa)
Load 20-30 μg of total protein per lane for cell lysates
Include positive control lysates from cells known to express DHX36
Transfer conditions:
Wet transfer to PVDF membrane at 100V for 90 minutes with cooling
Alternatively, semi-dry transfer at 25V for 30 minutes for faster results
Blocking and antibody incubation:
Block with 5% non-fat dry milk in TBST for 1 hour at room temperature
Dilute primary antibody 1:1000 in blocking solution
Incubate with primary antibody overnight at 4°C with gentle agitation
Wash 3 times with TBST, 5 minutes each
Incubate with HRP-conjugated secondary antibody (1:5000) for 1 hour at room temperature
Wash 3 times with TBST, 5 minutes each
Detection:
Use enhanced chemiluminescence (ECL) substrate
Expect band at approximately 110 kDa corresponding to DHX36
For effective immunoprecipitation of DHX36 and its associated nucleic acids:
Cross-linking optimization:
For RNA interactions: Use 0.1-0.3% formaldehyde or UV cross-linking (254 nm)
For DNA interactions: Use 1% formaldehyde for 10 minutes at room temperature
Lysis conditions:
Use mild lysis buffer (25 mM Tris-HCl pH 7.5, 150 mM NaCl, 1% NP-40, 1 mM EDTA)
Include RNase inhibitors when studying RNA interactions
Add benzonase for DNA studies to reduce background
Antibody selection:
Washing and elution:
Perform stringent washes (at least 5) with lysis buffer
For DNA studies: Include high-salt wash steps (up to 500 mM NaCl)
For RNA studies: Keep salt concentration lower (150 mM NaCl maximum)
Elute bound complexes with sample buffer for protein analysis or by proteinase K digestion for nucleic acid analysis
Recovery and analysis of bound nucleic acids:
Extract RNA using TRIzol or similar reagent
Extract DNA using phenol-chloroform
Analyze by qPCR, sequencing, or structure-specific assays for G-quadruplexes
When conducting research on DHX36, several critical controls should be included:
Expression controls:
Positive control: Cell lines with confirmed high DHX36 expression (e.g., HeLa cells)
Negative control: DHX36 knockdown/knockout cells generated via siRNA or CRISPR-Cas9
Rescue control: DHX36-deficient cells with reintroduced wild-type DHX36
Functional controls:
Catalytic-dead mutant: DHX36 with mutations in the ATPase domain to eliminate helicase activity
G4-binding mutant: DHX36 with mutations in the G4-binding domain to eliminate G4 recognition
Substrate controls: G4-forming sequences versus matched sequences unable to form G4 structures
Localization controls:
Nuclear marker (e.g., HDAC1) for nuclear DHX36 functions
Cytoplasmic marker (e.g., GAPDH) for cytoplasmic DHX36 functions
Fluorescent tagged DHX36 with subcellular markers for colocalization studies
Antibody validation controls:
Secondary antibody only control to assess non-specific binding
Pre-absorption control with immunizing peptide to confirm specificity
Multiple antibodies targeting different DHX36 epitopes to confirm observations
When performing immunohistochemistry with DHX36 antibody, researchers may encounter several challenges:
High background staining:
Problem: Non-specific binding of primary or secondary antibodies
Solution: Increase blocking time (2-3 hours), use higher concentration of blocking protein (5-10% normal serum), include 0.1% Triton X-100 in blocking solution to reduce non-specific binding
Weak or absent signal:
Problem: Insufficient antigen retrieval or antibody concentration
Solution: Optimize antigen retrieval methods (try citrate buffer pH 6.0 or EDTA buffer pH 9.0), increase antibody concentration, extend primary antibody incubation to overnight at 4°C
Inconsistent staining between samples:
Problem: Variability in fixation or processing
Solution: Standardize fixation protocols (10% neutral buffered formalin for 24 hours), ensure consistent section thickness (4-5 μm), process all samples simultaneously
Nuclear versus cytoplasmic localization discrepancies:
Problem: DHX36 can localize to both compartments depending on cellular context
Solution: Use confocal microscopy with Z-stack imaging to precisely determine localization, include co-staining with nuclear and cytoplasmic markers
Tissue-specific optimization:
Problem: Different tissues may require different protocols
Solution: Perform titration experiments for each new tissue type, adjust antigen retrieval time based on tissue density
Variability in DHX36 detection across cell lines represents a common challenge that can be addressed through systematic optimization:
Expression level differences:
Problem: DHX36 expression varies significantly between cell types
Solution: Adjust loading amounts based on preliminary expression screening, use housekeeping genes with similar expression levels across cell types for normalization
Post-translational modification variations:
Problem: DHX36 may undergo different modifications affecting antibody recognition
Solution: Use phosphatase inhibitors consistently, consider using multiple antibodies recognizing different epitopes, perform immunoprecipitation followed by mass spectrometry to identify modifications
Extraction efficiency differences:
Problem: DHX36 may be differentially extracted from different cell types
Solution: Compare multiple lysis protocols (RIPA, NP-40, sonication-based), include benzonase to release chromatin-bound protein, optimize lysis time and conditions for each cell type
Interference from binding partners:
Problem: Cell-specific DHX36 complexes may mask antibody binding sites
Solution: Include brief denaturation step, test different detergent concentrations, use epitope retrieval techniques similar to those in immunohistochemistry
Cross-reactivity concerns:
Problem: Antibody may recognize related helicases in certain cell contexts
Solution: Confirm specificity using DHX36 knockout cells for each cell line, perform peptide competition assays, validate with orthogonal methods (e.g., mass spectrometry)
Based on current understanding of DHX36 function, several promising research directions warrant investigation:
Role in antiviral immunity:
DHX36 in neurodegenerative disorders:
G4 structures are prevalent in genes associated with neurodegeneration
DHX36's role in resolving these structures may impact disease progression
Investigation of DHX36 expression and function in neurodegenerative disease models
Cancer therapeutic applications:
DHX36's influence on oncogene expression through G4 resolution
Potential for synthetic lethality approaches in tumors with altered G4 metabolism
Development of small molecule DHX36 inhibitors as possible therapeutics
Roles in RNA stress granule dynamics:
G4 structures in mRNAs can influence stress granule formation
DHX36 may regulate stress response through unwinding of G4-RNAs
Investigation of DHX36 localization and function during cellular stress
Interplay with other G4 binding proteins:
Cooperative or competitive interactions with other G4 binding factors
Regulatory networks controlling G4 structure resolution
Temporal dynamics of G4 formation and resolution in different cellular contexts
Single-cell technologies offer powerful approaches to understand DHX36 function with unprecedented resolution:
Single-cell RNA sequencing applications:
Examine cell-to-cell variability in DHX36 expression
Correlate DHX36 levels with expression of G4-regulated genes
Identify cell-type specific roles in heterogeneous tissues
Single-cell ATAC-seq combined with DHX36 analysis:
Map chromatin accessibility at G4-containing promoters
Correlate with DHX36 expression levels
Identify cell states where DHX36 activity may be particularly important
In situ approaches:
Develop G4-structure specific probes for visualization in intact cells
Correlate G4 abundance with DHX36 localization at single-cell level
Track dynamic changes in G4 structures during cell cycle or differentiation
Single-molecule imaging:
Track individual DHX36 molecules in living cells
Measure dwelling time at specific genomic loci
Determine kinetics of G4 resolution in different cellular contexts
Single-cell proteomics:
Quantify DHX36 protein levels alongside interacting partners
Identify cell-specific DHX36 complexes
Correlate post-translational modifications with functional outcomes