The DDX27 antibody is a polyclonal or monoclonal immunoglobulin designed to specifically target the DEAD-box helicase 27 (DDX27) protein. DDX27, a member of the DEAD-box family of RNA helicases, plays critical roles in RNA processing, ribosome assembly, and cancer progression. Its overexpression has been implicated in colorectal, breast, and gastric cancers, making it a focal point for oncological research . The antibody is primarily used in western blot (WB), immunofluorescence (IF), and immunohistochemistry (IHC) to detect and quantify DDX27 protein expression in cell lysates, tissues, or cultured cells .
Molecular Weight: DDX27 has a calculated molecular weight of 90 kDa, with a conserved DEAD-box motif (Asp-Glu-Ala-Asp) essential for RNA helicase activity .
Subcellular Localization: Primarily localized to the nucleus, where it regulates ribosomal RNA (rRNA) processing and interacts with nucleophosmin (NPM1) to modulate NF-κB signaling .
Helicase Activity: Facilitates RNA secondary structure remodeling, critical for ribosome assembly and transcriptional regulation .
Role in Tumorigenesis: DDX27 amplification and overexpression correlate with poor survival in CRC patients. The antibody has been used to confirm its oncogenic role in promoting proliferation, metastasis, and NF-κB pathway activation .
Experimental Validation: Western blot and IHC studies using the antibody demonstrated DDX27’s nuclear localization in CRC cells (e.g., HCT116, SW480) and its association with stemness markers (e.g., CD44, CD133) .
Stem Cell-Like Properties: Overexpression of DDX27 enhances breast cancer stem cell (BCSC) characteristics, including OCT4 and SOX2 expression. The antibody detected elevated DDX27 levels in mammospheres derived from MCF-7 and T47D cells .
Prognostic Marker: DDX27 overexpression predicts poor prognosis in GC patients. Antibody-based assays confirmed its role in colony formation and drug resistance .
| Antibody Vendor | Type | Applications | Reactivity | Dilution |
|---|---|---|---|---|
| Proteintech | Rabbit Polyclonal | WB, IF/ICC, ELISA | Human, Mouse, Rat | 1:500–1:2000 (WB/IF) |
| Abcam | Mouse Monoclonal | WB, IF/ICC | Human, Mouse | 1:50–1:200 (WB/IF) |
Proteintech’s antibody detected nuclear DDX27 in HeLa, NIH/3T3, and mouse liver tissues .
Abcam’s antibody showed specificity in HeLa and NIH/3T3 lysates via western blot, with observed bands at ~90 kDa .
Biomarker Potential: Elevated DDX27 expression correlates with aggressive tumor phenotypes across cancers, suggesting its utility as a diagnostic or prognostic marker .
Therapeutic Target: Inhibition of DDX27 via RNA interference (RNAi) or small molecules has shown promise in preclinical models, reducing tumor growth and enhancing chemosensitivity .
DDX27 (DEAD-box helicase 27) belongs to the DEAD-box RNA helicases family, characterized by conserved D-E-A-D (Asp-Glu-Ala-Asp) sequences. This ATP-dependent helicase plays critical roles in:
Ribosome biogenesis, specifically regulating 47S ribosomal RNA formation
Association with the PeBow-complex in ribosome synthesis pathways
RNA transportation and degradation processes
Influencing ribosome RNA maturation during skeletal muscle myogenesis
Various cellular metabolic processes including glucose and lipid metabolism
Beyond these normal functions, DDX27 has been implicated in tumorigenesis and cancer development, particularly in breast, hepatocellular, and gastrointestinal cancers .
Based on validated protocols, DDX27 antibodies are primarily utilized in:
Western Blot (WB): Successfully employed for detection in multiple sample types including:
Cell lines: RT-4, U-251 MG
Biological fluids: Human plasma
Tissues: Liver, tonsil
Immunohistochemistry (IHC): Particularly effective in formalin/PFA-fixed paraffin-embedded sections showing:
Strong nucleolar and cytoplasmic positivity in Purkinje cells of human cerebellum (at 1:10-1:20 dilution)
Primarily nuclear localization in breast cancer tissues
These applications enable researchers to assess DDX27 expression levels, compare expression between normal and pathological samples, analyze subcellular localization, and evaluate correlations with other biological markers .
Commercial DDX27 antibodies (such as PAB24527) typically have the following specifications:
| Parameter | Specification |
|---|---|
| Host | Rabbit |
| Type | Polyclonal |
| Immunogen | Recombinant protein corresponding to amino acids of human DDX27 |
| Reactivity | Human |
| Form | Liquid |
| Purification | Antigen affinity purification |
| Isotype | IgG |
| Storage Buffer | PBS, pH 7.2 (40% glycerol, 0.02% sodium azide) |
| Recommended Dilution (IHC) | 1:10-1:20 |
| Storage Conditions | 4°C short-term; -20°C long-term |
| Target Sequence | EDKEAKSGKLEKEKEAKEGSEPKEQEDLQENDEEGSEDEASETDYSSADENILTKADTLKVKDRKKKKKKGQEAGVFFEDASQYDENLSFQ |
Note that optimal working dilutions should be determined by the end user for specific applications and experimental conditions .
Validating antibody specificity is crucial for ensuring reliable experimental results. For DDX27 antibodies, consider these validation approaches:
Positive and negative control samples:
Positive controls: Use breast cancer tissues or cell lines with known high DDX27 expression
Negative controls: Include normal breast tissue (lower expression) or DDX27-knockout cells
Critical validation experiments:
Western blot analysis confirming a single band at the expected molecular weight
Pre-absorption with immunizing peptide to demonstrate binding specificity
DDX27 knockdown/knockout experiments comparing staining patterns with wild-type cells
Correlation of protein detection with mRNA expression levels from RT-qPCR or RNA-seq data
Multi-antibody verification:
Compare staining patterns using antibodies targeting different DDX27 epitopes
Verify consistent subcellular localization patterns (nucleolar/nuclear)
Staining pattern assessment:
DDX27 expression shows significant correlations with cancer progression parameters:
Expression profile:
Significantly higher expression in breast cancer compared to normal breast tissue
58.8% of breast cancer samples (97/165) show high DDX27 expression versus only 27.5% (11/40) in normal breast tissue
Clinical correlation analysis:
| Clinical Parameter | Correlation with High DDX27 Expression | Statistical Significance |
|---|---|---|
| Tumor size | Larger | p = 0.0005 |
| Lymph node status | Positive nodes | p = 0.0008 |
| Histological grade | Higher grade | p = 0.0040 |
| Ki-67 expression | Higher expression | p = 0.0063 |
| TNM stage | Later stage | p < 0.0001 |
These findings suggest DDX27 may serve as a potential prognostic biomarker and therapeutic target in breast cancer research .
Successful DDX27 immunohistochemistry requires careful protocol optimization:
Tissue fixation and processing:
Recommended: Formalin/PFA fixation with standard paraffin embedding
Section thickness: 4 μm sections for optimal staining
Antigen retrieval protocol:
Method: Citrate buffer with high-pressure heat-induced epitope retrieval
Critical step: Complete cooling to room temperature before antibody application
Blocking and antibody dilution:
Blocking: 3% hydrogen peroxide to inhibit endogenous peroxidase activity
Primary antibody: 1:10-1:20 dilution range for PAB24527 antibody
Incubation: 4°C overnight for primary antibody; room temperature for 1 hour for secondary
Detection system:
Recommended: Diaminobenzidine (DAB) with hematoxylin counterstaining
Expected patterns: Nuclear localization in breast cancer; nucleolar and cytoplasmic positivity in Purkinje cells
Standardized evaluation system:
Intensity scoring: Deep (3), medium (2), light (1), negative (0)
Percentage scoring: 0 (0-5%), 1 (6-25%), 2 (26-50%), 3 (51-75%), 4 (76-100%)
Score calculation: Intensity × percentage
Classification: High expression (≥4); low expression (≤3)
For tissues with high background or weak signal, further optimization of antibody concentration, incubation time, and antigen retrieval conditions may be necessary .
When encountering signal issues with DDX27 antibodies, consider these methodological solutions:
For weak signals:
Antibody optimization:
Decrease dilution ratio (increase concentration) within recommended ranges
For IHC, start with 1:10 dilution rather than 1:20
Extend primary antibody incubation time (24-48 hours at 4°C)
Antigen retrieval enhancement:
Increase retrieval duration or pressure
Ensure complete cooling before antibody application
Consider alternative buffers if citrate buffer yields insufficient results
Detection system sensitivity:
Use amplification-based detection systems
For Western blot, employ more sensitive chemiluminescent substrates
Increase exposure time while monitoring background levels
For non-specific signals:
Background reduction strategies:
Increase blocking time or concentration
Use more stringent washing protocols (more washes with higher detergent concentration)
Ensure proper quenching of endogenous peroxidase with 3% hydrogen peroxide
Antibody specificity improvement:
Increase antibody dilution to reduce non-specific binding
Pre-absorb antibody with non-specific proteins
Consider using a more specific monoclonal antibody alternative
Sample-specific adjustments:
Standardized quantification approaches ensure reliable DDX27 expression measurements:
Western blot quantification:
Normalize to appropriate loading controls (β-actin, GAPDH)
Use densitometric analysis with consistent exposure settings
Include internal reference samples across all blots for inter-experimental normalization
Perform replicate experiments (minimum n=3) and report means with appropriate statistical measures
IHC quantification:
Implement the standardized scoring system described in section 2.3
Employ multiple independent observers for objective scoring
Consider digital image analysis for more objective quantification
Analyze multiple regions to account for tumor heterogeneity
mRNA expression analysis:
For RT-qPCR: Use validated reference genes with stable expression
For RNA-seq: Implement proper normalization as described in research protocols:
Use edgR package for normalization (as used in TCGA data analysis)
Optimize data from the same patients and exclude formalin-fixed samples
Apply appropriate normalization methods (TPM, FPKM)
Cross-platform normalization:
DDX27's role in cancer stemness can be investigated through these methodological approaches:
Expression correlation analysis:
DDX27 positively correlates with established stemness markers:
OCT4 (p < 0.0001)
SOX2 (p = 0.0032)
IHC co-staining shows positive association between DDX27 and OCT4 expression (p < 0.0001, r = 0.428)
Functional analysis in cancer stem cell models:
DDX27 expression is significantly elevated in mammosphere models:
Higher expression in MCF-7 MS compared to parental MCF-7 cells
Higher expression in T47D MS compared to parental T47D cells
This suggests association with stem cell-like phenotypes
Overexpression studies:
DDX27 overexpression in breast cancer cells results in:
Upregulation of stemness biomarkers SOX2 and OCT4
Enhanced proliferation (measurable by CCK-8 assay)
Increased migration capability (quantifiable by Transwell assay)
Clinical correlation methodologies:
Analyze correlation between DDX27 expression and:
Cancer recurrence rates
Therapy resistance
Metastatic potential
Patient survival metrics (OS, DFS)
These approaches can establish DDX27's mechanistic role in cancer stemness and potential utility as a therapeutic target in cancer treatment strategies .
When investigating DDX27's function in ribosome biogenesis, researchers should implement these experimental design principles:
Subcellular localization analysis:
Perform immunofluorescence co-localization with nucleolar markers
Conduct subcellular fractionation followed by Western blot analysis
Verify nucleolar accumulation pattern in Purkinje cells and other relevant cell types
Ribosome biogenesis assessment:
Analyze pre-rRNA processing through Northern blot or qRT-PCR
Measure 47S rRNA formation rates with and without DDX27 manipulation
Evaluate polysome profiles following DDX27 depletion or overexpression
Interaction studies:
Investigate DDX27's association with the PeBow complex
Perform co-immunoprecipitation to identify interacting partners
Consider proximity labeling approaches (BioID, APEX) to identify the DDX27 interactome
Functional manipulation approaches:
Use RNA interference (siRNA, shRNA) for DDX27 knockdown
Apply CRISPR/Cas9 for DDX27 knockout models
Develop DDX27 overexpression systems with appropriate controls
Assess the effects of DDX27 mutations on ribosome synthesis
Translation efficiency measurement:
Conduct polysome profiling following DDX27 manipulation
Measure global protein synthesis using techniques like puromycin incorporation
Analyze translation of specific mRNAs important in cancer progression
These approaches enable systematic investigation of DDX27's mechanistic role in ribosome biogenesis and its downstream effects on cellular physiology .
Given DDX27's established role in cancer progression, potential therapeutic approaches include:
Direct targeting strategies:
Small molecule inhibitors of DDX27's helicase activity
Peptide inhibitors disrupting DDX27 interactions with the PeBow complex
Targeted degradation approaches (PROTACs) specific to DDX27
Therapeutic rationale based on research findings:
DDX27 correlates with poor prognosis (OS: p = 0.0087; DFS: p = 0.0235)
Enhances cancer stem cell properties that contribute to therapy resistance
Associated with aggressive clinicopathological features:
| Clinical Feature | Association | Therapeutic Implication |
|---|---|---|
| Tumor size | Larger tumors with high DDX27 | Potential for tumor reduction |
| Lymph node status | More positive nodes | Could reduce metastatic potential |
| Histological grade | Higher grade | May address aggressive phenotypes |
| TNM stage | Later stage disease | Possible benefit for advanced cancer |
Combinatorial therapy approaches:
Combining DDX27 inhibition with conventional chemotherapy
Targeting DDX27 alongside cancer stemness pathways
Using DDX27 expression as a biomarker for therapy selection
Translational research considerations:
Advanced methodologies for elucidating DDX27's cancer-related mechanisms include:
Single-cell analysis approaches:
Single-cell RNA sequencing to identify DDX27-high subpopulations
Mass cytometry (CyTOF) for simultaneous detection of DDX27 and stemness markers
Spatial transcriptomics to analyze DDX27 expression in the tumor microenvironment
High-throughput functional genomics:
CRISPR-Cas9 screens to identify synthetic lethal interactions with DDX27
RNA-seq following DDX27 manipulation to identify downstream effectors
Ribosome profiling to assess translation impacts of DDX27 activity
Advanced imaging technologies:
Super-resolution microscopy for detailed DDX27 localization
Live-cell imaging with fluorescently tagged DDX27 to track dynamics
FRET-based approaches to study DDX27 protein interactions in real-time
Structural biology approaches:
Cryo-EM studies of DDX27 in ribosome biogenesis complexes
Structure-based drug design for DDX27 inhibitors
Hydrogen-deuterium exchange mass spectrometry to map DDX27 interaction domains
In vivo models:
To differentiate DDX27's cancer-specific functions from other DEAD-box family members:
Comparative expression analysis:
Systematically analyze expression patterns of multiple DEAD-box helicases across cancer types
Identify cancer types where DDX27 shows unique expression patterns
Perform correlation analysis between DDX27 and other family members
Functional redundancy assessment:
Conduct rescue experiments using other DEAD-box helicases after DDX27 depletion
Compare phenotypic effects of knockdown/overexpression of multiple family members
Identify DDX27-specific cellular functions not complemented by other helicases
Domain-specific investigations:
Create chimeric proteins swapping domains between DDX27 and other DEAD-box helicases
Perform mutagenesis of conserved versus unique DDX27 regions
Develop domain-specific antibodies for functional studies
Interactome mapping:
Compare protein interaction networks of DDX27 with other family members
Identify DDX27-specific interaction partners relevant to cancer progression
Analyze unique vs. shared cellular pathways affected by different helicases
Cancer-specific relevance:
The following optimized Western blot protocol is recommended for DDX27 detection:
Sample preparation:
Extract proteins using RIPA buffer supplemented with protease inhibitors
For tissue samples: Homogenize thoroughly in cold buffer
For cell lines: Lyse directly in wells after PBS washing
Quantify protein concentration using BCA or Bradford assay
Prepare samples in Laemmli buffer with reducing agent (50-100 μg total protein)
Gel electrophoresis and transfer:
Separate proteins on 10% SDS-PAGE gel
Transfer to PVDF membrane (0.45 μm) at 100V for 90 minutes in cold transfer buffer
Immunoblotting:
Block membrane with 5% non-fat milk in TBST for 1 hour at room temperature
Incubate with primary DDX27 antibody at 1:1000 dilution in blocking buffer overnight at 4°C
Wash 3× with TBST, 10 minutes each
Incubate with HRP-conjugated secondary antibody (1:5000) for 1 hour at room temperature
Wash 3× with TBST, 10 minutes each
Development and analysis:
Develop using ECL substrate and appropriate imaging system
Expected band size: ~89.8 kDa
Normalize to loading control (β-actin or GAPDH)
Validated sample types:
For rigorous IHC assessment of DDX27 in clinical samples:
Specimen preparation protocol:
Fix tissues in 10% neutral buffered formalin for 24-48 hours
Process and embed in paraffin following standard protocols
Section at 4 μm thickness
Use positive control tissues in each batch (breast cancer with known high DDX27)
Optimized IHC protocol:
Deparaffinize sections in xylene and rehydrate through graded alcohols
Perform antigen retrieval in citrate buffer (pH 6.0) using high-pressure method
Block endogenous peroxidase with 3% hydrogen peroxide for 10 minutes
Apply primary DDX27 antibody at 1:10-1:20 dilution
Incubate overnight at 4°C in humidified chamber
Apply appropriate secondary antibody for 1 hour at room temperature
Develop with DAB and counterstain with hematoxylin
Standardized evaluation method:
Intensity scoring: deep (3), medium (2), light (1), negative (0)
Percentage scoring: 0 (0-5%), 1 (6-25%), 2 (26-50%), 3 (51-75%), 4 (76-100%)
Calculate final score = intensity × percentage
Define high expression: score ≥4; low expression: score ≤3
Quality control measures:
Include technical negative control (primary antibody omitted)
Use multi-tissue controls with known DDX27 expression
Have two independent pathologists score blindly
Document staining pattern (nuclear, nucleolar, cytoplasmic)
Data analysis recommendations: