DTX16 Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Composition: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Made-to-order (14-16 weeks)
Synonyms
DTX16 antibody; At5g52450 antibody; K24M7.20 antibody; Protein DETOXIFICATION 16 antibody; AtDTX16 antibody; Multidrug and toxic compound extrusion protein 16 antibody; MATE protein 16 antibody
Target Names
DTX16
Uniprot No.

Target Background

Database Links

KEGG: ath:AT5G52450

STRING: 3702.AT5G52450.1

UniGene: At.22618

Protein Families
Multi antimicrobial extrusion (MATE) (TC 2.A.66.1) family
Subcellular Location
Membrane; Multi-pass membrane protein.

Q&A

What is DTX16 and what cellular functions is it involved in?

DTX16/DHX16 is a DEAH-box RNA helicase that plays critical roles in pre-mRNA splicing as a component of the spliceosome. It contributes to pre-mRNA splicing after spliceosome formation and prior to the first transesterification reaction. As a component of the minor spliceosome, it is involved in the splicing of U12-type introns in pre-mRNAs. Beyond splicing, DHX16 has been identified as a pattern recognition receptor that senses splicing signals in viral RNA, playing a role in innate antiviral immune responses . Mechanistically, TRIM6 promotes the interaction between unanchored 'Lys-48'-polyubiquitin chains and DHX16, leading to DHX16 interaction with RIGI and ssRNA to amplify RIGI-dependent innate antiviral immune responses .

What applications are DHX16/DTX16 antibodies validated for?

DHX16 antibodies are typically validated for several key laboratory techniques:

  • Western Blotting (WB): For detection of DHX16 protein in cell and tissue lysates

  • Immunoprecipitation (IP): For isolation of DHX16 protein complexes

  • Immunohistochemistry (IHC): For visualization of protein localization in tissue sections

  • Immunocytochemistry/Immunofluorescence (ICC-IF): For determining subcellular localization

Most commercially available anti-DHX16 antibodies are validated for Western blot applications, with additional validations for IP and immunofluorescence depending on the specific antibody . When selecting an antibody, researchers should verify the validation status for their specific application and species of interest.

What are the optimal sample preparation conditions for DHX16 antibody in Western blotting?

For optimal Western blot results with DHX16 antibody:

  • Cell lysis: Use RIPA buffer supplemented with protease inhibitors to prevent protein degradation

  • Protein concentration: Load 20-40 μg of total protein per lane

  • Denaturation: Heat samples at 95°C for 5 minutes in reducing sample buffer

  • Gel selection: Use 8-10% SDS-PAGE gels (DHX16 has a molecular weight of approximately 110 kDa)

  • Transfer conditions: Transfer to PVDF membrane at 100V for 90 minutes in cold transfer buffer containing 20% methanol

  • Blocking: 5% non-fat dry milk or BSA in TBST for 1 hour at room temperature

  • Primary antibody dilution: Typically 1:1000-1:2000 in blocking buffer (optimize based on specific antibody)

  • Incubation: Overnight at 4°C with gentle rocking

  • Detection: Use appropriate HRP-conjugated secondary antibody and ECL detection system

Additionally, reducing conditions are typically recommended for optimal band detection, as demonstrated in experimental data from antibody validation studies .

How do I determine the appropriate antibody concentration for immunofluorescence staining of DHX16?

Determining the optimal antibody concentration for immunofluorescence requires a systematic titration approach:

  • Begin with a concentration range based on manufacturer recommendations (typically 1-10 μg/mL for most DHX16 antibodies)

  • Prepare a dilution series (e.g., 1, 2, 5, and 10 μg/mL) in antibody dilution buffer

  • Include proper controls:

    • Negative control: Secondary antibody only

    • Positive control: A cell line known to express DHX16 (e.g., K562 or SW13 cell lines)

    • Specificity control: Cells with DHX16 knockdown

  • Analyze signal-to-noise ratio for each concentration

  • Select the lowest concentration that provides clear specific staining with minimal background

For DHX16, cytoplasmic and nuclear staining patterns are expected based on its known subcellular localization. Based on protocols used in validation studies, a concentration of approximately 5-10 μg/mL with 3-hour incubation at room temperature has been shown to provide optimal results . Counterstaining with DAPI helps to visualize the nuclear localization component.

How can I distinguish between DHX16 isoforms using available antibodies?

DHX16 has multiple isoforms due to alternative splicing, which presents challenges for isoform-specific detection. To distinguish between DHX16 isoforms:

  • Epitope mapping: Check the immunogen sequence of the antibody to determine which isoforms it recognizes. Antibodies raised against C-terminal epitopes (e.g., aa 950 to C-terminus) will detect specific isoforms .

  • Western blot analysis:

    • Use high-resolution SDS-PAGE (6-8% gels) for optimal separation

    • Compare band patterns with predicted molecular weights of known isoforms

    • Include isoform-specific positive controls if available

  • Immunoprecipitation followed by mass spectrometry:

    • Immunoprecipitate using the DHX16 antibody

    • Analyze precipitated proteins by mass spectrometry

    • Identify peptides unique to specific isoforms

  • Validation with genetic tools:

    • Use isoform-specific siRNAs to knock down individual isoforms

    • Verify antibody specificity by observing selective band reduction

For conclusive isoform identification, researchers may need to use a combination of these approaches, particularly when studying tissues or cell types where multiple isoforms are co-expressed.

What strategies can overcome cross-reactivity issues when studying DHX16 in complex samples?

Cross-reactivity with other DEAH-box proteins can complicate DHX16 detection in complex samples. Several strategies can minimize this issue:

  • Pre-adsorption of antibody:

    • Incubate antibody with recombinant proteins from the same family (e.g., DHX9, DHX15)

    • Remove bound antibodies using protein A/G beads

    • Use the pre-adsorbed antibody for your experiment

  • Competitive peptide blocking:

    • Pre-incubate the antibody with excess blocking peptide corresponding to the immunogen

    • Compare staining patterns with and without peptide blocking

    • Specific signals should be eliminated by peptide blocking

  • Genetic validation approaches:

    • Use CRISPR/Cas9 to knockout DHX16 in your experimental system

    • Compare antibody reactivity in wild-type versus knockout samples

    • True DHX16 signals should be absent in knockout samples

  • Sequential immunoprecipitation:

    • Perform an initial IP with antibodies against potential cross-reactive proteins

    • Use the depleted lysate for DHX16 immunoprecipitation

    • This approach helps remove proteins that might cross-react

These strategies are particularly important when working with tissues or cell types that express multiple DEAH-box family members with high sequence similarity.

How does antibody affinity for DHX16 change under different post-translational modification states?

DHX16 undergoes several post-translational modifications (PTMs) that can affect antibody recognition, including phosphorylation, ubiquitination, and SUMOylation. Understanding these effects requires:

  • Phosphorylation effects:

    • DHX16 phosphorylation status changes during the cell cycle and splicing reactions

    • Antibodies raised against regions containing phosphorylation sites may show reduced binding when the protein is phosphorylated

    • To detect total DHX16 regardless of phosphorylation status, select antibodies targeting regions without known phosphorylation sites

  • Ubiquitination considerations:

    • The interaction between DHX16 and unanchored 'Lys-48'-polyubiquitin chains is part of its function in antiviral response

    • This modification can mask epitopes or alter protein conformation

    • For studying ubiquitinated DHX16, use denaturing conditions prior to immunoprecipitation

  • Experimental approaches to address PTM interference:

    • Treat samples with phosphatases or deubiquitinating enzymes before antibody application

    • Compare antibody binding under native versus denaturing conditions

    • Use a panel of antibodies targeting different epitopes to ensure detection regardless of modification state

  • Validation through mass spectrometry:

    • Perform IP-MS analysis to identify PTMs present on the detected protein

    • Correlate antibody binding efficiency with specific modification patterns

This understanding is crucial when studying DHX16 in different cellular contexts, particularly during viral infection when its modification state may change significantly.

What are the technical considerations for using DHX16 antibodies in chromatin immunoprecipitation (ChIP) experiments?

While DHX16 is primarily known for its role in RNA processing rather than direct DNA binding, researchers investigating its potential chromatin associations should consider:

  • Crosslinking optimization:

    • For protein-RNA-DNA complexes, dual crosslinking may be required

    • Use 1% formaldehyde for 10 minutes followed by a glycine quench

    • Consider additional crosslinkers like DSG for protein-protein interactions

  • Sonication parameters:

    • Optimize sonication conditions to generate chromatin fragments of 200-500 bp

    • Excessive sonication may disrupt DHX16-containing complexes

    • Insufficient fragmentation may lead to high background

  • Antibody selection criteria:

    • Choose antibodies validated for immunoprecipitation applications

    • Confirm epitope accessibility in crosslinked chromatin

    • Use antibodies targeting different epitopes to validate findings

  • Controls and validation:

    • Include IgG controls matched to the host species of the DHX16 antibody

    • Perform ChIP-qPCR at known negative regions

    • Consider ChIP after DHX16 knockdown as a specificity control

    • Validate findings with multiple antibodies if possible

  • Data interpretation:

    • DHX16 enrichment may represent indirect association through protein-protein interactions

    • RNA-dependent interactions may be distinguished by including RNase treatment controls

    • Consider performing RNA immunoprecipitation (RIP) in parallel for comparison

These technical considerations are essential for generating reliable ChIP data when investigating the potential chromatin-associated functions of DHX16.

What are common reasons for false positive or false negative results when using DHX16 antibodies?

Several factors can contribute to misleading results when working with DHX16 antibodies:

Causes of false positives:

  • Cross-reactivity with related DEAH-box helicases due to sequence homology

  • Non-specific binding to denatured proteins in fixed samples

  • Excessive antibody concentration leading to background signal

  • Insufficient blocking or inadequate washing steps

  • Secondary antibody cross-reactivity

Causes of false negatives:

  • Epitope masking due to protein-protein interactions or post-translational modifications

  • Inadequate antigen retrieval for fixed samples

  • Protein degradation during sample preparation

  • Insufficient antibody concentration or incubation time

  • Using antibodies raised against human DHX16 for detection in other species with poor conservation at the epitope region

Verification strategies:

  • Include positive and negative control samples in each experiment

  • Validate results with at least two antibodies targeting different epitopes

  • Confirm specificity through genetic approaches (siRNA, CRISPR knockout)

  • Perform peptide competition assays to verify binding specificity

  • Include appropriate technical controls for each application

By systematically addressing these potential issues, researchers can significantly improve the reliability of their DHX16 antibody-based experiments.

How can I optimize fixation conditions for DHX16 immunostaining in different tissue types?

Fixation conditions significantly impact DHX16 antibody staining due to its nuclear and cytoplasmic localization. Optimization strategies include:

For formalin-fixed paraffin-embedded (FFPE) tissues:

  • Fixation duration:

    • Standard 10% neutral buffered formalin for 24-48 hours

    • Avoid prolonged fixation which can mask epitopes

    • For delicate tissues, reduce fixation time to 12-24 hours

  • Antigen retrieval methods comparison:

    • Heat-induced epitope retrieval (HIER): Test both citrate buffer (pH 6.0) and EDTA buffer (pH 9.0)

    • Enzymatic retrieval: Test proteinase K digestion (1-5 μg/mL for 10-20 minutes)

    • For DHX16, HIER with EDTA buffer (pH 9.0) often yields superior results

  • Tissue-specific considerations:

    • Brain tissue: Extend antigen retrieval time to 30 minutes

    • Muscle tissue: Add a permeabilization step with 0.2% Triton X-100

    • Lung tissue: Reduce background by including 0.3% hydrogen peroxide treatment

For frozen tissues and cells:

  • Fixative selection:

    • 4% paraformaldehyde (10-15 minutes at room temperature)

    • Methanol fixation (-20°C for 10 minutes) may better preserve certain DHX16 epitopes

    • Combine with 0.1-0.3% Triton X-100 for optimal nuclear staining

  • Post-fixation treatments:

    • Add a mild permeabilization step (0.1% Triton X-100 for 5-10 minutes)

    • Block with 5-10% normal serum from the same species as the secondary antibody

  • Validation approach:

    • Test multiple fixation methods in parallel on the same tissue type

    • Compare staining intensity, specificity, and background

    • Document optimal conditions for each tissue type in your laboratory protocols

These optimization strategies should be systematically tested and documented to establish reliable protocols for DHX16 detection across different experimental systems.

What are the recommended approaches for multiplexing DHX16 antibodies with other markers?

Multiplexed detection allows visualization of DHX16 in relation to other cellular components. Consider these strategies:

  • Antibody compatibility assessment:

    • Select primary antibodies from different host species (e.g., rabbit anti-DHX16 with mouse anti-splicing factor)

    • For antibodies from the same species, use sequential immunostaining with HRP inactivation between rounds

    • Test for potential cross-reactivity between secondary antibodies

  • Fluorophore selection for immunofluorescence:

    • Choose fluorophores with minimal spectral overlap (e.g., Alexa Fluor 488, 555, and 647)

    • Include single-color controls to assess bleed-through

    • Consider brightness differences when selecting fluorophore combinations

  • Optimized protocols for co-detection with organelle markers:

    • For nuclear co-localization: Use rabbit anti-DHX16 with mouse anti-SC35 (splicing speckle marker)

    • For cytoplasmic RNA granules: Combine DHX16 staining with anti-DCP1 (P-body marker)

    • For viral infection studies: Co-stain for DHX16 and viral proteins

  • Sequential multiplex immunohistochemistry:

    • Apply tyramide signal amplification (TSA) for sequential detection

    • Between rounds, completely strip or inactivate previous antibodies

    • Document antibody order effects on staining intensity

  • Validation of multiplex results:

    • Compare multiplex staining patterns with single antibody controls

    • Confirm expected co-localization patterns based on known biology

    • Include appropriate negative controls for each marker

When optimizing multiplex protocols, start with established combinations and systematically introduce new antibodies while validating each step.

How can DHX16 antibodies be utilized to study its role in antiviral immune responses?

Recent research has revealed DHX16's role as a pattern recognition receptor in antiviral immunity . To investigate this function:

  • Infection model systems:

    • Study DHX16 localization before and after viral infection

    • Track temporal changes in DHX16 expression and localization during infection

    • Compare responses across different viral challenges (RNA vs. DNA viruses)

  • Protein-protein interaction analysis:

    • Use DHX16 antibodies for co-immunoprecipitation to identify interaction partners

    • Compare interaction networks in uninfected versus infected cells

    • Validate key interactions through reciprocal co-IP and proximity ligation assays

  • Methodological approach for studying TRIM6-DHX16 interactions:

    • Immunoprecipitate DHX16 and blot for ubiquitin to detect unanchored K48-linked polyubiquitin chains

    • Perform IP under native conditions to preserve these interactions

    • Include RNase treatment controls to determine RNA-dependence of interactions

    • Compare wild-type cells to TRIM6 knockdown cells

  • DHX16 translocation during viral infection:

    • Track DHX16 redistribution using immunofluorescence at multiple time points post-infection

    • Correlate localization changes with activation of downstream antiviral signaling pathways

    • Quantify nuclear-cytoplasmic distribution changes using high-content imaging

These approaches can help elucidate how DHX16 contributes to innate immune sensing and response to viral pathogens, particularly through its interaction with RIGI and TRIM6-mediated signaling pathways .

What considerations should be made when using DHX16 antibodies in patient-derived samples for disease research?

When applying DHX16 antibodies to clinical specimens, researchers should address several important considerations:

  • Preanalytical variables:

    • Fixation time: Clinical samples often have variable fixation durations

    • Tissue processing: Standardize antigen retrieval protocols for FFPE samples

    • Storage effects: Consider the impact of long-term storage on epitope preservation

    • Sample age: Validate antibody performance on archived versus fresh samples

  • Disease-specific optimization:

    • Cancer tissues: May require additional blocking steps to reduce background

    • Inflammatory conditions: Consider the impact of tissue inflammation on antibody specificity

    • Neurodegenerative diseases: May require specialized fixation for optimal detection

  • Quantitative analysis approaches:

    • Develop scoring systems based on staining intensity and distribution

    • Use digital pathology tools for unbiased quantification

    • Include control tissues on the same slide for normalization

  • Validation in disease contexts:

    • Compare results from multiple antibodies targeting different DHX16 epitopes

    • Correlate protein detection with mRNA expression data

    • Validate findings across multiple patient cohorts

  • Ethical and consent considerations:

    • Ensure appropriate ethical approvals for antibody-based studies

    • Consider limitations in consent for archived specimens

    • Document sample handling in accordance with regulatory requirements

These considerations are essential when translating DHX16 research from cell lines and animal models to patient-derived specimens, particularly when investigating its potential role in cancer, inflammatory diseases, or viral infections.

How can we design custom DHX16 antibodies with enhanced specificity for particular experimental applications?

Designing antibodies with improved specificity for DHX16 requires careful consideration of protein structure, homology, and application requirements:

  • Epitope selection strategies:

    • Target unique regions with low homology to other DEAH-box proteins

    • Focus on accessible regions based on protein structure prediction

    • Consider regions that maintain native conformation in your application

    • Avoid regions subject to variable post-translational modifications

  • Computational design approaches:

    • Use bioinformatic tools to identify DHX16-specific regions

    • Compare sequences across species for conserved epitopes (for cross-reactivity)

    • Model epitope accessibility in the folded protein

    • Predict potential cross-reactive epitopes in related proteins

  • Validation methodologies:

    • Test against recombinant DHX16 and related family members

    • Validate in cells with CRISPR-mediated DHX16 knockout

    • Perform epitope mapping to confirm binding to the intended region

    • Cross-validate across multiple applications (WB, IP, IF)

  • Application-specific optimization:

    • For ChIP applications: Target epitopes away from DNA/RNA binding domains

    • For live-cell imaging: Select epitopes accessible in native conditions

    • For detecting specific isoforms: Target unique exon junctions

As demonstrated in antibody engineering research, this approach of combining biophysics-informed modeling with extensive validation can generate antibodies with customized specificity profiles, enabling precise detection of DHX16 even in complex biological samples .

What are the emerging techniques for studying DHX16 protein dynamics using antibody-based approaches?

Novel antibody applications are expanding our ability to study DHX16 dynamics in real-time:

  • Live-cell antibody-based imaging techniques:

    • Nanobody approaches: Using smaller antibody fragments for improved intracellular delivery

    • SNAP/CLIP-tag fusion proteins combined with antibody detection

    • Single-chain variable fragments (scFvs) expressed intracellularly

  • Super-resolution microscopy applications:

    • STORM/PALM imaging of DHX16 within nuclear splicing bodies

    • Expansion microscopy to visualize DHX16 interactions with splicing machinery

    • Correlative light and electron microscopy (CLEM) for ultrastructural localization

  • Proximity-dependent labeling approaches:

    • BioID or TurboID fusions to DHX16 to identify proximal proteins

    • APEX2-mediated proximity labeling followed by antibody detection

    • Integration with quantitative proteomics for temporal interaction mapping

  • Single-molecule tracking methodologies:

    • Quantum dot-conjugated antibodies for long-term tracking

    • Optimal labeling strategies: Use Fab fragments for reduced impact on protein function

    • Analysis approaches: Mean square displacement analysis for diffusion characteristics

  • Förster resonance energy transfer (FRET) applications:

    • Antibody-based FRET sensors for DHX16 conformational changes

    • Detecting DHX16-substrate interactions through FRET pairs

    • Time-resolved FRET for studying transient interactions

These emerging techniques extend beyond traditional static imaging to provide insights into the dynamic behavior of DHX16 during splicing reactions and viral sensing, offering unprecedented resolution of its functional mechanisms in living cells.

What is the optimal protocol for immunoprecipitation of DHX16-associated RNA-protein complexes?

For effective isolation of DHX16 ribonucleoprotein complexes:

Detailed IP-RIP (Immunoprecipitation-RNA Immunoprecipitation) Protocol:

  • Cell preparation:

    • Harvest 10-20 million cells at 80% confluency

    • Wash twice with ice-cold PBS

    • Resuspend in 1 mL of polysome lysis buffer (100 mM KCl, 5 mM MgCl₂, 10 mM HEPES pH 7.0, 0.5% NP-40, 1 mM DTT)

    • Add RNase inhibitors (40 U/μL) and protease inhibitors

  • Antibody coupling:

    • Pre-couple 5 μg of DHX16 antibody to 50 μL of Protein A/G magnetic beads

    • Rotate for 2 hours at 4°C in NT2 buffer (50 mM Tris-HCl pH 7.4, 150 mM NaCl, 1 mM MgCl₂, 0.05% NP-40)

  • Immunoprecipitation:

    • Add cleared cell lysate to antibody-coupled beads

    • Incubate overnight at 4°C with gentle rotation

    • Include IgG control IP in parallel

  • Washing conditions:

    • Wash 5 times with NT2 buffer containing 300 mM NaCl

    • Perform one final wash with NT2 buffer (150 mM NaCl)

  • RNA isolation:

    • Split the beads for protein and RNA analysis

    • For RNA: Add TRIzol directly to beads, isolate RNA per manufacturer's protocol

    • For protein: Elute with SDS sample buffer for Western blot verification

  • RNA analysis options:

    • RT-qPCR for known RNA targets

    • RNA-seq for unbiased profiling

    • Include input RNA control for normalization

This protocol has been optimized to maintain RNA integrity while ensuring specific immunoprecipitation of DHX16-associated complexes. The high-salt washing steps are critical for reducing non-specific RNA binding while preserving authentic interactions.

What are the considerations for using DHX16 antibodies in in situ proximity ligation assays (PLA)?

In situ PLA enables visualization of protein-protein interactions at the single-molecule level. For DHX16 PLA:

  • Antibody selection criteria:

    • Primary antibodies must be from different species (e.g., rabbit anti-DHX16 with mouse anti-interaction partner)

    • Validate individual antibodies by immunofluorescence before PLA

    • Confirm epitope accessibility in fixed samples

    • Consider using monoclonal antibodies for improved specificity

  • Fixation and permeabilization optimization:

    • Test multiple fixation methods (4% PFA, methanol, or combination)

    • For nuclear proteins like DHX16, use 0.5% Triton X-100 for 10 minutes to ensure nuclear permeabilization

    • Include antigen retrieval step for formalin-fixed samples

  • Critical controls:

    • Negative controls: Omit one primary antibody

    • Biological negative control: Use cells with knockdown of one interaction partner

    • Positive control: Known interacting proteins expressed in your cell system

    • Technical validation: Single antibody controls to assess non-specific oligonucleotide binding

  • Interaction-specific considerations:

    • For RNA-dependent interactions: Include RNase treatment controls

    • For transient interactions: Consider mild crosslinking (0.5-1% formaldehyde)

    • For splicing complex interactions: Compare results under active transcription versus transcriptional inhibition

  • Quantification approaches:

    • Count PLA signals per nucleus/cell

    • Analyze colocalization with nuclear compartment markers

    • Compare signal intensity and distribution across experimental conditions

This approach is particularly valuable for studying DHX16 interactions with components of the splicing machinery or viral RNA sensing complexes, as it provides spatial information about where these interactions occur within the cell.

How can mass spectrometry be combined with DHX16 antibody-based enrichment for comprehensive protein characterization?

Integrating immunoprecipitation with mass spectrometry offers powerful insights into DHX16 complexes and modifications:

  • Sample preparation for IP-MS:

    • Scale up: Use 10-fold more cells than standard IP

    • Minimize keratin contamination: Work in a clean environment

    • Crosslinking options: Consider reversible crosslinkers like DSP (dithiobis[succinimidyl propionate])

    • Elution methods: Use on-bead digestion or gentle elution with glycine (pH 2.5)

  • DHX16-specific protocol adjustments:

    • Include RNase treatment controls to distinguish direct versus RNA-mediated interactions

    • Compare native versus denaturing conditions to assess complex integrity

    • Consider size exclusion chromatography pre-IP to isolate specific complexes

  • Post-translational modification analysis:

    • Enrichment strategies for phosphopeptides: TiO₂ or IMAC

    • Ubiquitination analysis: Include deubiquitinase inhibitors during lysis

    • SUMOylation detection: Use SUMO-specific enrichment before or after IP

  • Data analysis approaches:

    • Compare DHX16 interactome across different cellular conditions

    • Identify condition-specific interactions (e.g., during viral infection)

    • Quantify changes in PTM status under different stimuli

    • Network analysis to place DHX16 in functional protein communities

  • Validation of MS findings:

    • Confirm key interactions by reciprocal IP

    • Validate PTM sites with phospho-specific antibodies (if available)

    • Perform functional studies on identified interaction partners

This integrated approach provides a comprehensive view of DHX16's dynamic protein interactions and modification states across different cellular conditions, offering insights beyond what can be achieved with antibody-based detection alone.

How do we interpret contradictory results between different DHX16 antibodies in the same experimental system?

When faced with discrepancies between different antibodies targeting DHX16:

  • Systematic evaluation approach:

    • Compare epitopes: Map each antibody's target region on DHX16

    • Review validation data: Assess evidence for specificity of each antibody

    • Check for isoform specificity: Determine if antibodies recognize different isoforms

    • Consider PTM interference: Evaluate if modifications might block epitope access

  • Technical confirmation strategies:

    • Perform genetic validation: siRNA or CRISPR knockout to confirm specificity

    • Use peptide competition: Block with immunizing peptide to verify signal specificity

    • Compare multiple detection methods: IF versus WB versus IP results

    • Check subcellular fractionation: Compare nuclear versus cytoplasmic signals

  • Reconciliation framework:

    ScenarioLikely ExplanationResolution Approach
    Different band patterns in WBIsoform specificity or PTM sensitivityUse RNA-seq to confirm isoform expression; perform phosphatase treatment
    Different subcellular localizationEpitope masking in specific compartmentsUse multiple antibodies; validate with tagged DHX16
    Discrepant interaction partnersEpitope interference with specific complexesUse alternative techniques (BioID, crosslinking)
    Varying expression levelsAntibody affinity differencesCalibrate with recombinant standards
  • Publication recommendations:

    • Report all antibodies used with catalog numbers

    • Document discrepancies transparently

    • Include all relevant controls for each antibody

    • Consider orthogonal validation approaches

When interpreting contradictory results, consider that each antibody may be revealing different aspects of DHX16 biology rather than one being simply "right" or "wrong."

How does DHX16 expression and localization vary across different tissue types and disease states?

Understanding DHX16 tissue distribution provides context for experimental findings:

  • Normal tissue expression patterns:

    • DHX16 is ubiquitously expressed across tissues, with highest levels in metabolically active and proliferating tissues

    • Nuclear localization predominates in most cell types, with concentration in nuclear speckles

    • Cytoplasmic presence increases in certain cell types, particularly immune cells

  • Cell type-specific considerations:

    • Neurons: Predominantly nuclear with enrichment in Cajal bodies

    • Immune cells: Notable cytoplasmic fraction, increasing upon activation

    • Proliferating cells: Higher expression levels compared to quiescent cells

    • Specialized secretory cells: Distinct localization patterns near ER-associated splicing sites

  • Disease-associated changes:

    • Cancer: Often upregulated, with altered nuclear/cytoplasmic distribution

    • Viral infection: Redistribution associated with innate immune function

    • Neurodegenerative disorders: Potential accumulation in pathological inclusions

    • Autoimmune conditions: Can be targeted by autoantibodies in some disorders

  • Developmental dynamics:

    • Expression increases during embryonic development

    • Cell differentiation associated with changing DHX16 levels

    • Tissue-specific isoform expression patterns emerge during development

When designing experiments, these tissue-specific patterns should inform the selection of appropriate positive controls and guide the interpretation of DHX16 antibody staining patterns in different biological contexts.

What emerging technologies will enhance DHX16 antibody applications in single-cell analysis?

Several cutting-edge technologies are poised to revolutionize DHX16 antibody applications at the single-cell level:

  • Single-cell proteomics integration:

    • Mass cytometry (CyTOF) with metal-conjugated DHX16 antibodies

    • Antibody-based single-cell Western blotting

    • Microfluidic antibody capture for single-cell protein quantification

    • Integration with single-cell transcriptomics for multi-omic analysis

  • Spatial transcriptomics applications:

    • In situ sequencing combined with DHX16 antibody detection

    • Spatial mapping of DHX16-associated RNA processing events

    • Correlation of DHX16 localization with local transcriptome profiles

    • Multiplexed imaging with cyclic immunofluorescence for tissue architecture context

  • Microfluidic approaches:

    • Droplet-based single-cell antibody assays

    • Microfluidic trapping devices for dynamic antibody-based measurements

    • Integration with live-cell imaging for temporal analysis

  • Improvements in sensitivity:

    • Signal amplification methods (e.g., tyramide signal amplification, rolling circle amplification)

    • Ultrasensitive detection using quantum dots or photonic crystals

    • Single-molecule pull-down assays with antibody-based capture

These technologies will enable researchers to move beyond population averages to understand cell-to-cell variation in DHX16 expression, localization, interactions, and function, particularly in heterogeneous tissues and during dynamic processes like viral infection or cellular differentiation.

How can machine learning approaches improve the analysis of DHX16 antibody-based imaging data?

Machine learning is transforming the analysis of complex antibody staining patterns:

  • Automated pattern recognition applications:

    • Classification of DHX16 subcellular localization patterns

    • Detection of subtle redistribution following stimuli

    • Quantification of colocalization with other proteins

    • Identification of rare cells with altered DHX16 expression

  • Deep learning for image analysis:

    • Convolutional neural networks for DHX16 signal segmentation

    • Attention-based models for identifying regions of interest

    • Transfer learning to apply trained models across different tissues

    • Generative models to predict DHX16 localization based on other markers

  • Multi-parameter data integration:

    • Correlation of DHX16 staining with multiple cellular markers

    • Clustering of cells based on DHX16 and other protein patterns

    • Trajectory analysis to map DHX16 changes during cellular processes

    • Network analysis of DHX16 interactions in spatial context

  • Practical implementation considerations:

    • Data preprocessing requirements for machine learning applications

    • Training data needs and annotation approaches

    • Validation strategies for AI-based findings

    • Computational requirements and software tools

By leveraging these computational approaches, researchers can extract more information from DHX16 antibody staining than is possible with traditional analysis methods, enabling new insights into its function in complex tissues and heterogeneous cell populations.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.