The DDX21 antibody is a specialized immunoglobulin designed to detect the DEAD-box helicase 21 (DDX21) protein, a critical RNA helicase involved in ribosomal RNA biogenesis, RNA editing, and transcription regulation . This antibody is widely utilized in molecular biology research to study DDX21’s role in cellular processes, cancer progression, and immune regulation. Its specificity allows for precise visualization and quantification of DDX21 in tissues and cells via techniques such as Western blotting (WB), immunohistochemistry (IHC), and immunoprecipitation (IP).
Western Blotting (WB): Detects DDX21 protein expression levels in lysates, with reported molecular weights ranging from 75–100 kDa, depending on post-translational modifications .
Immunohistochemistry (IHC): Identifies DDX21 localization in tissue sections, revealing nuclear or nucleolar patterns in cancer samples .
Immunoprecipitation (IP): Isolates DDX21 for interaction studies, such as its binding to c-Jun or RIG-I .
Flow Cytometry (FC): Measures DDX21 expression in intracellular compartments of immune cells .
Cancer Biology: DDX21 antibodies have shown elevated expression in breast, colorectal, and gastric cancers, correlating with tumor progression and prognosis .
Immune Regulation: Studies using DDX21 antibodies revealed its role in modulating type I interferon (IFN-I) signaling, with knockout models demonstrating enhanced antiviral responses .
Viral Interactions: DDX21 antibodies identified its antagonistic effects on foot-and-mouth disease virus (FMDV) IRES-dependent translation .
Breast Cancer: High DDX21 expression promotes cell proliferation and tumor growth by regulating AP-1 transcriptional activity and rRNA processing .
Colorectal Cancer: DDX21 interacts with WDR5 to enhance CDK1 expression, driving cell cycle progression .
Gastric Cancer: Overexpression of DDX21 correlates with poor prognosis and increased Cyclin D1 levels .
Innate Immunity: DDX21 suppresses IFN-β production by competing with RIG-I for dsRNA binding, maintaining immune homeostasis .
Viral Pathogenesis: DDX21 restricts FMDV replication by inhibiting IRES-mediated translation .
Targeting DDX21 may offer new strategies for cancer treatment and antiviral therapies. For instance, its downregulation in colorectal cancer cells halts cell cycle progression , while its inhibition enhances antiviral responses .
DDX21, also known as DEAD (Asp-Glu-Ala-Asp) box polypeptide 21, is a 783 amino acid protein that functions as an RNA helicase with essential roles in RNA metabolism. Its primary functions include:
Unwinding double-stranded RNA and folding single-stranded RNA into secondary structures
Facilitating ribosome and spliceosome assembly
Regulating both RNA Polymerase I and II transcription
Processing ribosomal RNA (rRNA)
Contributing to innate immune responses against viral infections
Promoting cell cycle progression
DDX21 is primarily localized in the nucleus, specifically in the nucleolus, where it participates in the multi-protein B-WICH complex. Its activity can be influenced by various post-translational modifications that affect its helicase activity and protein interactions. In certain cellular contexts, it can undergo translocation from the nucleolus to other cellular compartments in response to specific stimuli .
DDX21 is involved in regulating multiple cellular processes critical for normal cell function:
DDX21 has been identified as playing crucial roles in cancer progression, particularly in colorectal, breast, and pancreatic cancers. Recent research indicates that DDX21 interacts with WDR5 to promote colorectal cancer cell proliferation by activating CDK1 gene expression and influencing histone modifications (H3K4me3) on the CDK1 promoter . This finding provides new insights into potential therapeutic targets for cancer treatment.
DDX21 expression varies between normal and disease states, particularly in cancer and viral infections:
In cancer:
During viral infections:
HCMV infection prevents the normal repression of DDX21 at both protein and mRNA levels
Virus infection can induce DDX21 translocation from the nucleus to the cytoplasm
Caspase-dependent cleavage of DDX21 occurs after virus infection, especially with RNA viruses
Research shows that DDX21 undergoes caspase-dependent cleavage at D126 during viral infections, which promotes its translocation from the nucleus to the cytoplasm. This cleaved form of DDX21 negatively regulates interferon beta (IFN-β) signaling by suppressing the formation of the DDX1-DDX21-DHX36 complex, thus modulating the host immune response .
Different DDX21 antibodies are optimized for specific experimental applications:
When selecting a DDX21 antibody, researchers should consider:
The specific cellular compartment being investigated (nucleolar vs. nucleoplasmic vs. cytoplasmic)
The experimental technique and required sensitivity
The species of the experimental model
Whether post-translational modifications or specific DDX21 isoforms are being studied
For example, when studying DDX21 translocation during viral infection, immunofluorescence with antibodies validated for subcellular localization would be most appropriate. For examining protein-protein interactions, antibodies optimized for co-immunoprecipitation would be preferred .
For optimal DDX21 immunohistochemical (IHC) staining in tissue samples:
Protocol optimization:
Tissue preparation: Deparaffinize in xylene and rehydrate in graded alcohols
Antigen retrieval: Use 0.01 M sodium citrate buffer (pH 6.0) or TE buffer (pH 9.0) in a steamer
Endogenous peroxidase blocking: Treat with 1% hydrogen peroxide in methanol for 30 minutes
Background blocking: Pre-incubate in 10% normal FBS for 1 hour
Primary antibody: Incubate with DDX21 antibody at appropriate dilution (typically 1:50-1:500) at room temperature for 2 hours
Detection: Follow standard protocol based on DAKO Envision kit using polymer to amplify signals
Critical considerations:
DDX21 is predominantly nucleolar/nuclear in normal cells but may show altered localization in cancer or infected cells
For dual staining, select compatible secondary antibodies to avoid cross-reactivity
Include positive controls (colon cancer tissue, breast cancer tissue) and negative controls (skeletal muscle tissue)
Confirm antibody specificity through western blot validation before IHC studies
Tissue microarray analysis has shown that DDX21 expression is elevated in breast cancer compared to adjacent normal tissues, with potential prognostic implications . Similar patterns have been observed in colorectal cancer, suggesting broader relevance across multiple cancer types.
DDX21 undergoes dynamic subcellular relocalization during viral infection, making it an important aspect of host-pathogen interaction studies:
Methodological approach:
Immunofluorescence analysis:
Fix cells with 4% paraformaldehyde (20 min) and permeabilize with 0.2% Triton X-100 (20 min)
Block with 2% BSA (1 hour) followed by overnight antibody incubation at 4°C
Use specific cellular markers (nucleolar, nuclear, cytoplasmic) for co-localization studies
Analyze using confocal microscopy for precise subcellular localization
Nucleocytoplasmic separation assay:
Time-course experiments:
Monitor DDX21 localization at different time points post-infection
Correlate with viral replication stages and immune response activation
Research has demonstrated that during HCMV infection, DDX21 translocates from the nucleolus to the nucleoplasm . In contrast, PRRSV infection promotes DDX21 translocation from the nucleus to the cytoplasm . For PRRSV, this translocation is independent of DDX21's ATPase, RNA helicase, and foldase activity, highlighting virus-specific mechanisms of DDX21 regulation.
DDX21 plays multiple roles in cancer progression through several mechanisms:
Mechanisms of DDX21-mediated cancer progression:
Cell cycle regulation:
Transcriptional regulation:
RNA metabolism alterations:
Epigenetic modulation:
Pan-cancer analysis has revealed that DDX21 is overexpressed in multiple cancer types and correlates with poor prognosis. For example, DDX21 knockdown significantly inhibits colorectal cancer cell proliferation and blocks cell cycle at G2/M phase, suggesting its potential as a therapeutic target .
In pancreatic cancer, DDX21 is upregulated regardless of molecular subtype (classical or basal-like), indicating its fundamental role in PDAC pathogenesis . This consistent overexpression makes DDX21 a promising biomarker for pancreatic cancer detection and prognosis.
DDX21 has complex, sometimes opposing roles in viral infection and innate immunity:
Pro-viral functions:
Positively regulates replication of certain viruses (e.g., PRRSV, HCMV)
Cleaved DDX21 suppresses IFN-β signaling pathway
Required for proper viral late gene transcription in HCMV infection
Anti-viral functions:
Forms part of DDX1-DDX21-DHX36 complex that senses viral dsRNA
Binds adaptor protein TRIF to initiate immune signaling
Inhibits influenza A virus replication (though counteracted by viral NS1 protein)
Regulatory mechanisms:
Cleavage-dependent regulation:
R-loop regulation:
Experimental evidence shows that DDX21 knockdown significantly reduces vesicular stomatitis virus (VSV) replication but also inhibits interferon production and interferon-stimulated gene expression. This dual effect suggests that DDX21 helps maintain a balance in innate immune responses .
For HCMV, DDX21 mRNA and protein levels are maintained during infection (while they decline in uninfected cells), and its translocation from nucleolus to nucleoplasm is essential for proper viral replication, particularly late gene expression .
Knockdown and overexpression models provide complementary approaches to understand DDX21 function:
Knockdown methodologies:
siRNA approach:
shRNA approach:
CRISPR/Cas9 gene editing:
Overexpression systems:
Transient transfection:
Lentiviral overexpression:
Experimental readouts:
Cell proliferation (CCK8 assay, colony formation)
Cell cycle analysis (flow cytometry)
Viral replication (viral titers, viral protein expression)
Immune signaling (IFN-β luciferase reporter, ISG expression)
Gene expression (qRT-PCR, RNA-seq)
Protein-protein interactions (co-IP, proximity ligation)
Research has shown that DDX21 knockdown in lung adenocarcinoma cells significantly inhibits cell proliferation and migration, while overexpression enhances these processes . In viral studies, knockdown and overexpression models have revealed context-dependent roles for DDX21, with discrepancies between these models highlighting the importance of post-translational modifications in regulating DDX21 function .
ChIP (Chromatin Immunoprecipitation) and DRIP (DNA-RNA Immunoprecipitation) assays are powerful techniques to investigate DDX21's chromatin functions:
ChIP assay applications:
Promoter binding analysis:
Histone modification correlation:
Complex formation analysis:
Use sequential ChIP (Re-ChIP) to identify co-binding partners (e.g., WDR5)
Examine recruitment of RNA polymerases and transcription factors
Map DDX21-containing complexes genome-wide
DRIP assay applications:
R-loop detection:
R-loop resolution mechanisms:
RNase H treatment controls to confirm specificity
Rescue experiments using RNase H overexpression
Time-course analyses to track dynamic changes in R-loop formation
Recent research has demonstrated that DDX21 knockdown leads to increased R-loop accumulation on viral late genes during HCMV infection, impairing their transcription. This defect can be rescued by RNase H overexpression, confirming the causal relationship between R-loops and transcriptional impairment . In cancer research, ChIP assays have revealed that DDX21 binds to the CDK1 promoter in colorectal cancer cells and influences the deposition of H3K4me3 marks, providing mechanistic insight into how DDX21 promotes cancer cell proliferation .
DDX21 forms context-specific protein complexes that mediate its diverse cellular functions:
Major DDX21 protein complexes:
Methods to study DDX21 protein interactions:
Co-immunoprecipitation (Co-IP):
Proximity ligation assays:
Detect protein-protein interactions in situ
Visualize DDX21 interactions within specific subcellular compartments
Quantify interaction frequencies under different conditions
Domain mapping experiments:
Research shows that in colorectal cancer, DDX21 directly interacts with WDR5, a core component of histone methyltransferase complexes. This interaction influences the deposition of H3K4me3 marks on the CDK1 promoter, thereby promoting cell cycle progression . In antiviral immunity, DDX21 forms part of the DDX1-DDX21-DHX36 complex that senses viral dsRNA, but caspase-dependent cleavage of DDX21 during infection disrupts this complex and modulates the immune response .
Post-translational modifications (PTMs) of DDX21 critically regulate its activity, localization, and protein interactions:
Key DDX21 modifications:
Caspase-dependent cleavage:
Deacetylation by SIRT7:
Phosphorylation:
Affects DDX21 helicase activity and interactions
Regulates nucleolar/nucleoplasmic distribution
Responsive to cellular stresses and metabolic changes
SUMOylation:
Modifies DDX21 localization and function
Influences interactions with specific protein partners
Potentially regulated during cell cycle progression
Experimental approaches to study PTMs:
Site-directed mutagenesis:
Mass spectrometry:
Identify specific modification sites
Quantify modification changes under different conditions
Map modification patterns across the protein
PTM-specific antibodies:
Detect modified forms of DDX21
Monitor dynamic changes in modification status
Determine subcellular localization of modified DDX21
Recent research has demonstrated that caspase-dependent cleavage of DDX21 at D126 occurs following virus infection and treatment with RNA/DNA ligands, especially for RNA viruses. This cleavage event promotes the translocation of DDX21 from the nucleus to the cytoplasm, where the cleaved form negatively regulates the IFN-β signaling pathway by suppressing the formation of the DDX1-DDX21-DHX36 complex. This mechanism helps maintain immune balance during viral infection .
Researchers may encounter several challenges when working with DDX21 antibodies:
Common challenges and solutions:
Background signal issues:
Cross-reactivity concerns:
Subcellular localization discrepancies:
Detection sensitivity limitations:
Epitope masking due to protein interactions:
Problem: Reduced detection in specific cellular contexts
Solution: Use multiple antibodies targeting different DDX21 domains, try different extraction methods, and consider native vs. denaturing conditions depending on application
When selecting DDX21 antibodies, researchers should consider the specific application, target species, and whether post-translational modifications or specific protein complexes are being studied. Validation using multiple approaches, particularly including knockdown or knockout controls, is essential for ensuring specificity and reproducibility.
Distinguishing between different functional states of DDX21 requires specialized approaches:
Methodological strategies:
Analysis of subcellular localization:
Nucleolar DDX21: Associated with rRNA processing and ribosome biogenesis
Nucleoplasmic DDX21: Involved in RNA polymerase II regulation and 7SK binding
Cytoplasmic DDX21: Often associated with innate immune sensing or cleaved form
Use confocal microscopy with specific subcellular markers for precise localization
Detection of specific protein complexes:
Use co-immunoprecipitation followed by western blotting for known partners
Apply proximity ligation assays to visualize specific interactions in situ
Employ glycerol gradient fractionation to separate distinct DDX21-containing complexes
Identification of post-translationally modified forms:
Assessment of enzymatic activity:
RNA-binding analysis:
Perform RNA immunoprecipitation (RIP) to identify bound RNAs (rRNAs, snoRNAs, 7SK)
Use CLIP-seq to map RNA binding sites genome-wide
Compare RNA binding profiles in different cellular contexts
Research has shown that DDX21 undergoes dramatic relocalization during viral infection, moving from the nucleolus to the nucleoplasm during HCMV infection or from the nucleus to the cytoplasm during PRRSV infection . Additionally, caspase-cleaved DDX21 shows distinct functional properties compared to the full-length protein, particularly in regulating innate immune responses .
Integrating DDX21 research with -omics approaches provides comprehensive insights into its functions:
Multi-omics strategies:
Transcriptomic integration:
Perform RNA-seq after DDX21 knockdown or overexpression
Identify directly and indirectly regulated genes
Conduct pathway enrichment analysis on differentially expressed genes
Example: Gene Ontology analysis of top 100 genes co-expressed with DDX21 reveals enrichment in RNA processing and ribosome biogenesis pathways
Proteomic approaches:
Use IP-MS (immunoprecipitation coupled with mass spectrometry) to identify DDX21 interactome
Compare interactomes across different cell types or conditions
Apply quantitative proteomics to measure proteome-wide changes upon DDX21 manipulation
Research shows DDX21 interactions with factors involved in RNA processing, transcription, and chromatin modification
Chromatin profiling:
RNA-protein interaction mapping:
Apply CLIP-seq or PAR-CLIP to identify DDX21-bound RNAs
Determine RNA binding motifs and structural preferences
Correlate with functional outcomes for bound transcripts
Integrative bioinformatic analysis:
Use tools like GEPIA2 and STRING to analyze co-expression networks
Identify key correlation patterns with genes like RPL5, RPL30, ZC3H18, NCL, FTSJ3, and STAU1
Apply BioGRID data to map protein-protein interaction networks
Pan-cancer analysis through TCGA, GEO, GTEx, CPTAC, and HPA datasets reveals DDX21 as a potential biomarker across multiple cancer types
Recent pan-cancer analysis integrated multiple -omics datasets to identify DDX21 as a potential biomarker for cancer prognosis. This approach combined transcriptomic data from TCGA with protein expression data from the Human Protein Atlas, revealing that DDX21 overexpression correlates with poor survival across multiple cancer types. Co-expression network analysis further identified key DDX21-associated genes involved in RNA processing and cell cycle regulation .