Phospho-DDX5 (Y593) Antibody

Shipped with Ice Packs
In Stock

Description

2.1. Detection of DDX5 Phosphorylation

The antibody is pivotal in studying DDX5’s phosphorylation-dependent roles in cancer. For example:

  • Tongue Cancer: Phosphorylation at Tyr593/Tyr595 correlates with tumor progression by inhibiting TRAIL-induced apoptosis and enhancing proliferation .

  • Chronic Myeloid Leukemia (CML): FL118, a DDX5-targeting compound, suppresses DDX5 expression and induces apoptosis in BCR-ABL inhibitor-resistant cells .

2.2. Immunohistochemistry and Immunofluorescence

  • Tumor Microenvironment Analysis: High DDX5 expression in tongue cancer tissues correlates with increased CD8+ T cell infiltration and reduced immune exhaustion .

  • Subcellular Localization: Validates nuclear localization of phosphorylated DDX5 in breast carcinoma tissues .

2.3. Western Blot and ELISA

  • Specificity: ELISA assays confirm no cross-reactivity with non-phosphorylated DDX5 .

  • Sensitivity: Detects DDX5 phosphorylation in lysates from MCF-7, A375, and K562 cells .

3.1. DDX5’s Dual Role in Cancer

  • Oncogenic Function: Promotes cancer cell proliferation and drug resistance via transcriptional regulation of oncogenes (e.g., c-Myc) .

  • Tumor-Suppressive Activity: In tongue cancer, high DDX5 expression enhances immune infiltration and cytotoxic T cell activity .

3.2. Therapeutic Targeting

FL118, a camptothecin derivative, downregulates DDX5 expression and inhibits Topoisomerase I, showing potent efficacy against CML cells resistant to BCR-ABL inhibitors .

Product Specs

Buffer
The antibody is provided as a liquid solution in phosphate-buffered saline (PBS) containing 50% glycerol, 0.5% bovine serum albumin (BSA), and 0.02% sodium azide.
Form
Liquid
Lead Time
Typically, we can ship products within 1-3 business days after receiving your order. Delivery times may vary depending on the shipping method or location. Please consult your local distributors for specific delivery time estimates.
Synonyms
ATP dependent RNA helicase DDX5 antibody; DDX 5 antibody; Ddx5 antibody; DDX5_HUMAN antibody; DEAD (Asp Glu Ala Asp) box helicase 5 antibody; DEAD (Asp Glu Ala Asp) box polypeptide 5 antibody; DEAD box 5 antibody; DEAD box protein 5 antibody; DEAD/H (Asp Glu Ala Asp/His) box polypeptide 5 (RNA helicase; 68kD) antibody; G17P1 antibody; HELR antibody; HLR1 antibody; HUMP68 antibody; P68 antibody; p68 RNA helicase antibody; Probable ATP dependent RNA helicase DDX5 antibody; Probable ATP-dependent RNA helicase DDX5 antibody; RNA helicase p68 antibody
Target Names
Uniprot No.

Target Background

Function
DDX5, also known as p68, is a DEAD-box RNA helicase involved in various cellular processes, including pre-mRNA splicing regulation, transcriptional regulation, and viral infection. DDX5 plays a critical role in regulating alternative splicing, particularly by increasing the inclusion of exon 10 in the tau pre-mRNA in a RBM4-dependent manner. This activity is essential for the proper function of the tau protein. DDX5 binds to the tau pre-mRNA in the stem-loop region downstream of exon 10. Its RNA helicase activity is highly stimulated by single-stranded RNA, which contributes to its role in RNA metabolism. Beyond its role in splicing, DDX5 is also implicated in transcriptional regulation. Notably, DDX5 acts as a transcriptional coactivator for various transcription factors, including androgen receptor (AR), estrogen receptor 1 (ESR1), MYOD1, p53/TP53, RUNX2, and others. DDX5 interacts with these transcription factors and enhances their transcriptional activity. In some cases, DDX5 can act as a transcriptional repressor, likely by associating with histone deacetylases, such as HDAC1. As a component of a large PER complex, DDX5 plays a role in inhibiting 3' transcriptional termination of circadian target genes such as PER1 and NR1D1, contributing to the regulation of circadian rhythms.
Gene References Into Functions
  1. RIP-seq analysis in HEK293T cells has identified a comprehensive repertoire of DDX5/p68 interacting transcripts, including the LOC284454 lncRNA. PMID: 29227193
  2. Research has indicated that DDX5 is significantly upregulated in gastric cancer tissues and plays a novel role in promoting gastric cancer cell proliferation through the mTOR pathway. PMID: 28216662
  3. DDX5 is known to be involved in RNA metabolism and viral infection. Notably, DDX5 appears to be hijacked by RNA viruses to facilitate their replication. However, DDX5 also exhibits antiviral activity during HBV and MYXV infection. This contrasting behavior between DNA and RNA viruses likely reflects the different modes of RNA and DNA virus biosynthesis. [review] PMID: 29642538
  4. A significant overlap has been observed between hnRNPA1 and DDX5 splicing targets, with these factors often sharing closely linked binding sites. PMID: 30042133
  5. DDX5 has been shown to play a role in regulating esophageal cancer cell proliferation and tumorigenesis. It is highly expressed in esophageal cancer. PMID: 28244855
  6. Downregulation of p68 RNA Helicase (DDX5) has been found to activate a survival pathway involving mTOR and MDM2 signaling. PMID: 28557706
  7. Studies have provided refined biochemical and biological comparisons between the yeast Dbp2 and human DDX5 enzymes. Human DDX5 exhibits a 10-fold higher unwinding activity than Dbp2, partially attributed to the presence of a mammalian/avian specific C-terminal extension. Moreover, ectopic expression of DDX5 rescues the cold sensitivity, cryptic initiation defects, and impaired glucose import in dbp2Delta cells, suggesting functional conservation between these two proteins. PMID: 28411202
  8. p53 gain-of-function mutations have been shown to accelerate endometrial carcinoma progression and metastasis by interfering with Drosha and p68 binding and pri-miR-26a-1 processing, resulting in reduced miR-26a expression and EZH2 overexpression. PMID: 26587974
  9. Cervical cancer cell DDX5 gene is transcriptionally upregulated by calcitriol through a VDRE located in its proximal promoter. PMID: 26314252
  10. Systematic analysis of the human Cyclin Dependent Kinase (CDK)-9 interactome has identified novel functions in RNA splicing mediated by the DEAD Box DDX5 and DDX17 RNA Helicases. PMID: 26209609
  11. LMTK3, a protein involved in tumor suppression, escapes the regulation of tumor suppressor miRNAs through sequestration of DDX5. PMID: 26739063
  12. Research has provided a model demonstrating the interplay between p68 and p53 in regulating PLK1 expression, offering insights into the behavior of these molecules and the outcome of their interaction in human breast cancer. PMID: 24626184
  13. Studies have revealed a novel mechanism of oncogenesis attributed to p68, involving the upregulation of AKT, leading to nuclear exclusion and degradation of the tumor suppressor FOXO3a. PMID: 25745998
  14. DDX5 has been found to play a crucial role in the proliferation and tumorigenesis of non-small-cell lung cancer cells by activating the beta-catenin signaling pathway. PMID: 26212035
  15. Research suggests that correcting p68 expression may reduce the toxicity of mutant RNAs in myotonic dystrophy types 1 and 2 (DM1 and DM2). PMID: 26080402
  16. Data indicate that cyclooxygenase 2 (COX-2) is inversely correlated with microRNA 183 (miR-183) and directly correlated with DEAD-box helicase p68 (DDX5). PMID: 25963660
  17. DDX5 protein is essential for normal cell proliferation, and its concentration increases during the transition from the G1 to S/G2 phase of the cell cycle. PMID: 26035968
  18. Studies have shown that acute myeloid leukemia (AML) is dependent on DDX5, and inhibiting DDX5 expression slows down AML cell proliferation. PMID: 24910429
  19. Downregulation of DDX5 and DDX17 protein expression during myogenesis and epithelial-to-mesenchymal transdifferentiation contributes to the switching of splicing programs during these processes. PMID: 24910439
  20. Data indicate that the armadillo repeat protein ARVCF interacts with the splicing factors SRSF1 (SF2/ASF), the RNA helicase p68 (DDX5), and the heterogeneous nuclear ribonucleoprotein hnRNP H2. PMID: 24644279
  21. Researchers have identified DDX5 as a positive regulator for Japanese encephalitis virus replication through its binding to the viral 3' UTR. PMID: 24035833
  22. DDX5 facilitates HIV-1 replication as a cellular co-factor of Rev. PMID: 23741449
  23. DDX5 could be critical for NOTCH1-mediated T-ALL pathogenesis, suggesting it as a potential new target for modulating Notch signaling in leukemia. PMID: 23108395
  24. p68, in the presence of Ca-calmodulin, can function as a microtubule motor. PMID: 23322042
  25. Studies have shown a novel role for DDX5 in cancer cell proliferation, suggesting DDX5 as a potential therapeutic target in breast cancer treatment. PMID: 22750847
  26. High p68 RNA helicase expression has been associated with glioma. PMID: 22810421
  27. Data indicate that transcriptional coregulator ddx5/ddx17 RNA helicases can simultaneously regulate the transcriptional activity and alternative splicing of NFAT5 transcription factor. PMID: 22266867
  28. RNA helicases Ddx17 and Ddx5 contribute to tumor-cell invasiveness by regulating alternative splicing of several DNA- and chromatin-binding factors, including the macroH2A1 histone. PMID: 23022728
  29. The DEAD box RNA helicase p68, also referred to as DDX5, directly interacts with the vitamin D receptor (VDR). PMID: 22476084
  30. There is a direct interaction between DDX5 and the hepatitis C virus (HCV) NS5B polymerase, and DDX5 has two independent NS5B-binding sites. PMID: 22640416
  31. High DDX5 expression is associated with basal breast cancer cells. PMID: 22086602
  32. Using an RNA affinity pulldown-coupled mass spectrometry approach, DDX5/RNA helicase p68 has been identified as an activator of TAU exon 10 splicing. PMID: 21343338
  33. A striking inverse association between p68 and delta133p53 expression has been observed in primary breast cancers. PMID: 20818423
  34. DEAD-box RNA helicase p68 (DDX5) and its associated noncoding RNA, steroid receptor RNA activator (SRA), form a complex with CTCF that is essential for insulator function. PMID: 20966046
  35. p300-mediated acetylation has pleiotropic effects on p68 and p72 RNA helicase. PMID: 20663877
  36. Crystallization and preliminary diffraction analysis of the N-terminal region of DDX5 have been reported. X-ray diffraction data were processed to a resolution of 2.7 A. PMID: 20124720
  37. DDX5 acts as a repressor of fibrogenic genes in hepatic stellate cells (HSCs) through interaction with transcriptional complexes. PMID: 20022962
  38. DDX5 is essential for pre-mRNA splicing in vitro and may function in destabilizing the U1-5'ss interaction. Depletion of p68 RNA helicase has been found to arrest spliceosome assembly at the prespliceosome stage. PMID: 12101238
  39. DDX5 exhibits synergy with transcriptional coactivators CBP and p300. PMID: 12527917
  40. DDX5 plays a role in c-H-ras alternative splicing regulation. PMID: 12665590
  41. p68 is an important transcriptional regulator, functioning as a co-activator and/or co-repressor depending on the context of the promoter and the transcriptional complex. The amino acids 1-478 of p68 can repress transcription, as can the full-length protein. PMID: 15298701
  42. There is a tightly controlled expression and nucleolar localization of p68 in keratinocytes in vitro and during skin repair in vivo, which contributes to keratinocyte proliferation and gene expression. PMID: 15304501
  43. Research has investigated how p68 RNA helicase may act as a tumor cosuppressor in governing p53 transcriptional activity. PMID: 15660129
  44. Data suggest that the functions of p68 RNA helicase may be subject to regulation by multiple cell signaling pathways. PMID: 15927448
  45. Increasing Tlk1 activity in HT1080 cells through forced Tlk1 overexpression leads to increased phosphorylation of endogenous p68. PMID: 15950181
  46. Patients with chronic hepatitis C carrying DDX5 haplotypes are at an increased risk of developing advanced liver fibrosis. PMID: 16697732
  47. SUMO modification of the DEAD box protein p68 modulates its transcriptional activity and promotes its interaction with HDAC1. PMID: 17369852
  48. A mutant carrying mutations at the phosphorylation sites (Y593/595F) dramatically sensitizes TRAIL-resistant cells to TRAIL-induced apoptosis, suggesting a potential therapeutic strategy to overcome TRAIL resistance. PMID: 17384675
  49. The percentage correlation between Q-RT-PCR and microarray data were 70% and 48%, respectively, using DDX5 and GAPDH as internal controls. PMID: 17540040
  50. p68/p72 may contribute to colon cancer formation by directly upregulating proto-oncogenes and indirectly by downregulating the growth suppressor p21(WAF1/CIP1). PMID: 17699760

Show More

Hide All

Database Links

HGNC: 2746

OMIM: 180630

KEGG: hsa:1655

STRING: 9606.ENSP00000225792

UniGene: Hs.279806

Protein Families
DEAD box helicase family, DDX5/DBP2 subfamily
Subcellular Location
Nucleus. Nucleus, nucleolus. Cytoplasm.

Q&A

What is the biological significance of DDX5 phosphorylation at Y593?

Phosphorylation of DDX5 at the Y593 residue is a critical post-translational modification that significantly alters its functionality. When phosphorylated at Y593, DDX5 exhibits enhanced coactivation of androgen receptor transcription . More significantly, this phosphorylation promotes the dissociation of histone deacetylase 1 (HDAC1) from the Snail1 promoter, thereby activating Snail1 transcription, inhibiting E-cadherin expression, and ultimately promoting epithelial-mesenchymal transition (EMT) .

Additionally, Y593-phosphorylated DDX5 facilitates the nuclear translocation of β-catenin by blocking GSK-3β-induced β-catenin phosphorylation and replacing Axin in the β-catenin complex . This leads to activation of β-catenin target genes including cyclin D1 and c-Myc, stimulating cell proliferation . In glioblastoma, dual phosphorylation at Y593/Y595 inhibits cell apoptosis by suppressing XAF1 expression .

How does phosphorylated DDX5 (Y593) differ functionally from unphosphorylated DDX5?

Unphosphorylated DDX5 functions primarily as an RNA helicase involved in RNA metabolism, including pre-mRNA splicing, where its RNA helicase activity increases tau exon 10 inclusion . It also serves as a transcriptional regulator independent of its RNA helicase activity .

In contrast, Y593-phosphorylated DDX5 acquires additional oncogenic functions that significantly alter cellular processes:

  • Enhanced transcriptional coactivation: Phospho-DDX5 (Y593) exhibits increased coactivation of androgen receptor transcription

  • Altered protein interactions: Forms complexes with β-catenin, blocking its degradation and enabling nuclear translocation

  • Epigenetic regulation: Promotes dissociation of HDAC1 from target promoters, altering transcriptional landscapes

  • Cell survival promotion: Inhibits apoptotic pathways, particularly when dual-phosphorylated at Y593/Y595

  • Metastatic potential: Activates EMT-related genes through Snail1 upregulation

These phosphorylation-dependent functions make phospho-DDX5 (Y593) a critical player in cancer progression and potential therapeutic target .

What are the optimal storage conditions for phospho-DDX5 (Y593) antibodies?

To maintain optimal antibody activity, phospho-DDX5 (Y593) antibodies should be stored according to the following evidence-based recommendations:

  • Short-term storage (up to 2 weeks): Maintain at 4°C in refrigerated conditions

  • Long-term storage: Store at -20°C or -80°C

  • Aliquoting: Divide into small aliquots to avoid repeated freeze-thaw cycles, which can degrade antibody quality

  • Buffer composition: Most commercial antibodies come in optimized buffers that typically include:

    • 30-50% glycerol (to prevent freezing damage)

    • Phosphate buffered saline (PBS) or Tris-buffered saline (TBS)

    • 0.01-0.02% sodium azide (as preservative)

    • Sometimes includes 0.5% BSA (as stabilizer)

  • Temperature fluctuations: Avoid repeated freeze-thaw cycles which demonstrably reduce antibody efficacy

Testing has shown that antibodies stored under these conditions maintain their specificity and reactivity for the duration of their expected shelf life (typically 12 months from receipt) .

What are the recommended protocols for detecting phospho-DDX5 (Y593) in different experimental systems?

Based on validated research protocols, the following are recommended approaches for detecting phospho-DDX5 (Y593) in various experimental systems:

Western Blot Analysis:

  • Dilution range: 1:500-1:2000

  • Sample preparation: Total cell lysates (40μg protein typically sufficient)

  • Recommended positive controls: MCF-7, A375, U-87MG, CT-26, C6 cell lines

  • Molecular weight: Expected band at approximately 70 kDa

  • Blocking: 5% BSA in TBST recommended over milk (phospho-epitopes can be masked by milk proteins)

Immunohistochemistry:

  • Dilution range: 1:100-1:200

  • Sample preparation: Formalin-fixed paraffin-embedded tissue sections

  • Antigen retrieval: Heat-induced epitope retrieval with citrate buffer (pH 6.0)

  • Detection: ABC method or polymer-based detection systems

  • Validated tissues: Human prostate cancer has shown clear phospho-DDX5 (Y593) staining

Immunofluorescence:

  • Dilution range: 1:100-1:500

  • Fixation: 4% paraformaldehyde for 15 minutes at room temperature

  • Permeabilization: 0.1% Triton X-100 for 5 minutes

  • Expected subcellular localization: Predominantly nuclear staining

ELISA:

  • Validated for detecting phospho-DDX5 (Y593)

  • Capture antibody concentration: 1-2 μg/ml

  • Detection antibody dilution: 1:1000-1:5000

How can I reliably quantify changes in DDX5 phosphorylation status following experimental treatments?

For reliable quantification of DDX5 phosphorylation at Y593 following experimental treatments, researchers should implement multiple complementary approaches:

Western Blot Quantification:

  • Always run parallel blots with both phospho-specific and total DDX5 antibodies

  • Calculate the phospho-DDX5/total DDX5 ratio to normalize for variations in total protein expression

  • Include appropriate loading controls (β-actin, GAPDH)

  • Use digital image analysis software (ImageJ, Li-Cor, etc.) for densitometric analysis

  • Perform at least three independent experiments for statistical validity

Phosphorylation-Specific Flow Cytometry:

  • Fix cells with 4% paraformaldehyde

  • Permeabilize with cold methanol

  • Stain with fluorophore-conjugated phospho-DDX5 (Y593) antibodies (like Cy5.5 conjugated antibodies)

  • This allows quantification at single-cell resolution and detection of heterogeneous responses

Mass Spectrometry-Based Approaches:

  • Immunoprecipitate DDX5 using total DDX5 antibodies

  • Perform tryptic digestion

  • Use targeted mass spectrometry to quantify the absolute stoichiometry of Y593 phosphorylation

  • This provides the most accurate measurement of phosphorylation levels

Proximity Ligation Assay (PLA):

  • Use antibodies against total DDX5 and phosphotyrosine

  • PLA signals will only appear when DDX5 is phosphorylated

  • This approach allows in situ visualization of phosphorylation events in intact cells

Research has demonstrated that integrating multiple quantification methods provides the most reliable assessment of phosphorylation changes, particularly when studying signaling dynamics or drug responses .

What are the most effective strategies for validating phospho-DDX5 (Y593) antibody specificity?

Rigorous validation of phospho-DDX5 (Y593) antibody specificity is critical for reliable research outcomes. Based on established research practices, implementation of the following comprehensive strategy is recommended:

1. Peptide Competition Assays:

  • Pre-incubate antibody with phosphorylated and non-phosphorylated Y593 peptides

  • Specific binding should be blocked only by the phospho-peptide and not by non-phosphorylated peptide

  • This confirms phospho-specificity of the antibody

2. Phosphatase Treatment Controls:

  • Treat one sample set with lambda phosphatase before immunoblotting

  • Signal should be eliminated in phosphatase-treated samples while total DDX5 remains detectable

  • This verifies that the antibody specifically recognizes the phosphorylated form

3. Genetic Validation Approaches:

  • Use CRISPR/Cas9 to generate Y593F mutant cell lines (phenylalanine cannot be phosphorylated)

  • Compare antibody reactivity between wild-type and Y593F mutant samples

  • Absence of signal in Y593F mutants confirms specificity

4. Kinase Modulation:

  • Treat cells with kinase inhibitors known to affect DDX5 phosphorylation

  • Alternatively, overexpress kinases that target DDX5

  • Observe corresponding changes in phospho-Y593 signal

5. Cross-Reactivity Assessment:

  • Test antibody against recombinant phosphorylated and non-phosphorylated DDX5

  • Include the highly homologous DDX17 to ensure no cross-reactivity

  • Western blot analysis should show a single band at the expected molecular weight (~70 kDa)

6. Application-Specific Validation:

  • Validate separately for each application (WB, IHC, IF, etc.)

  • Use positive controls with known DDX5 Y593 phosphorylation (e.g., cells treated with growth factors)

  • Include negative controls (untransfected cells, secondary antibody only)

Implementation of these validation strategies ensures that experimental findings truly reflect specific phospho-DDX5 (Y593) detection rather than non-specific or artifactual signals.

How does the phosphorylation status of DDX5 at Y593 impact its role in β-catenin signaling in cancer?

The phosphorylation of DDX5 at Y593 creates a critical molecular switch that fundamentally alters β-catenin signaling with profound implications for cancer progression. Mechanistically, phospho-DDX5 (Y593) promotes β-catenin nuclear translocation through multiple coordinated pathways:

Direct Mechanism of Action:

  • Phospho-DDX5 (Y593) physically blocks GSK-3β-mediated phosphorylation of β-catenin

  • This prevents β-catenin targeting for proteasomal degradation

  • Phospho-DDX5 (Y593) replaces Axin in the β-catenin destruction complex, further stabilizing β-catenin

  • The stabilized β-catenin accumulates and translocates to the nucleus

  • In the nucleus, phospho-DDX5 (Y593) enhances β-catenin/TCF transcriptional activity

Oncogenic Consequences:

Therapeutic Targeting:
Compounds like RX-5902 (a quinoxalinyl-piperazine compound) have been developed to specifically:

  • Attenuate phosphorylation of DDX5 at Y593

  • Lead to cytoplasmic accumulation of β-catenin (preventing nuclear activity)

  • Downregulate c-MYC expression

  • Show promise in AML treatment by targeting this specific mechanism

This phosphorylation-dependent regulation represents a targetable vulnerability in cancers dependent on aberrant β-catenin signaling, including AML, colorectal cancer, and several other malignancies .

What is the relationship between DDX5 Y593 phosphorylation and tumor immune microenvironment modulation?

Recent single-cell and multi-omic analyses have uncovered a previously unrecognized relationship between DDX5 Y593 phosphorylation status and tumor immune microenvironment (TIME) modulation, with potentially significant implications for cancer immunotherapy.

Impact on Immune Cell Infiltration:

  • High DDX5 expression tumors demonstrate:

    • Significantly reduced B cell infiltration

    • Reduced macrophage infiltration, particularly M2-type

    • Increased T cell infiltration

  • DDX5 expression shows a strong negative correlation specifically with M2 macrophage (Macro_spp1) infiltration in the tumor microenvironment, as verified by multi-immunofluorescence staining

Mechanistic Insights:
Analysis of macrophage subtype distribution in relation to DDX5 expression reveals:

  • Elevation of Macro_apoec3a macrophages in DDX5-high tumors

  • Significant reduction in Macro_spp1, Macro_thbs1, and Macro_ccnl1 macrophages in DDX5-high tumors

  • These alterations in macrophage polarization may explain the decreased pro-tumor immune environment in DDX5-high contexts

Clinical Correlations:
Multi-immunofluorescence staining of clinical tongue cancer samples confirmed a negative association between DDX5 expression and M2-type macrophage infiltration in the tumor microenvironment

This evidence suggests that phospho-DDX5 status may serve as a biomarker for immunotherapy responsiveness and raises the possibility that targeting DDX5 phosphorylation could potentially reprogram the tumor immune microenvironment. The specific molecular mechanisms connecting Y593 phosphorylation to these immune modulatory effects requires further investigation, but appears to involve alteration of cancer cell-derived signals that influence immune cell recruitment and polarization .

How do different phosphorylation sites on DDX5 interact with Y593 phosphorylation to regulate its function?

DDX5 function is regulated by a complex interplay of multiple phosphorylation sites that can work synergistically, antagonistically, or independently of Y593 phosphorylation. This combinatorial code creates a sophisticated regulatory network with context-dependent outcomes.

Multiple Phosphorylation Sites and Their Interactions:

Phosphorylation SiteKinase ResponsibleInteraction with Y593Functional Outcome
Y593c-Abl, Src familyPrimary site of interestEnhances AR coactivation, β-catenin stabilization, EMT promotion
Y595c-Abl, Src familySynergisticDual Y593/Y595 phosphorylation inhibits cell apoptosis in glioblastoma by suppressing XAF1
T69PAK5Independent pathwayIncreases miR-10b production by enhancing Drosha/DGCR8 binding, promoting breast cancer progression
S557UnknownIndependent pathwayCreates microtubule motor functionality, promotes cell metastasis
T564/T446p38 MAP kinaseAntagonisticPromotes colon cancer cell apoptosis during chemotherapy, opposing the anti-apoptotic effect of Y593/Y595 phosphorylation

Regulatory Complexity:

  • Phosphorylation at different residues can exert opposite effects even within the same protein

    • Y593/Y595 phosphorylation promotes cell survival

    • T564/T446 phosphorylation promotes apoptosis during chemotherapy

  • The temporal sequence of phosphorylation events matters:

    • Y593 phosphorylation appears to be a primary event that enables subsequent protein interactions

    • Secondary phosphorylation events may fine-tune these interactions

  • Context-dependent outcomes:

    • In some cancer types, certain phosphorylation combinations predominate

    • The kinase/phosphatase balance in different cellular contexts determines which sites are phosphorylated

This complex phosphorylation interplay highlights the need for comprehensive phosphoproteomic analysis when studying DDX5 function, as focusing solely on Y593 may miss critical regulatory mechanisms. Additionally, therapeutic strategies targeting DDX5 should consider this multisite phosphorylation network to achieve desired specificity .

How does phospho-DDX5 (Y593) contribute differently to solid tumors versus hematological malignancies?

Phospho-DDX5 (Y593) exhibits distinct mechanisms of action and clinical implications in solid tumors compared to hematological malignancies, highlighting the context-dependent nature of its oncogenic functions.

Solid Tumors:

  • Epithelial-Mesenchymal Transition (EMT) Regulation:

    • In solid tumors, phospho-DDX5 (Y593) promotes EMT by activating Snail1 transcription through dissociation of HDAC1 from the Snail1 promoter

    • This leads to repression of E-cadherin and increased invasiveness

    • This mechanism is particularly important in carcinomas where EMT drives metastasis

  • Hormone Receptor Coactivation:

    • Enhances transcriptional coactivation of androgen receptor in prostate cancer

    • Modulates estrogen receptor signaling in breast cancer

    • This hormone-dependent regulation is specific to certain solid tumors

  • Tissue-Specific Effects:

    • Acts as a tumor suppressor in tongue cancer, with high expression associated with better outcomes

    • Functions as an oncogene in glioblastoma through dual Y593/Y595 phosphorylation

    • Required for proliferation in colon cancer cell lines like HCT116

Hematological Malignancies (AML):

These distinctions highlight the importance of cancer-specific approaches when targeting phospho-DDX5 (Y593), as strategies effective in hematological malignancies may not translate directly to solid tumors due to these fundamental differences in molecular mechanism and cellular context .

What are the most effective experimental models for studying phospho-DDX5 (Y593) function in different cancer types?

Selecting appropriate experimental models is crucial for investigating phospho-DDX5 (Y593) functions in cancer research. Based on validated research approaches, the following models offer distinct advantages for different research questions:

Cell Line Models:

Cancer TypeRecommended Cell LinesJustificationApplications
Breast CancerMCF-7High endogenous phospho-DDX5 (Y593) Hormone receptor interactions, β-catenin signaling
GlioblastomaU-87MG, C6Validated phospho-DDX5 (Y593) expression Dual Y593/Y595 phosphorylation studies
Colorectal CancerHCT116DDX5-dependent proliferation demonstrated DNA replication, cell cycle studies
AMLMultiple AML cell linesConstitutive Y593 phosphorylation β-catenin nuclear shuttling, RX-5902 efficacy testing
Tongue CancerVarious SCC linesContext where DDX5 acts as tumor suppressor Immune microenvironment interactions

Genetic Modification Approaches:

  • CRISPR/Cas9 Y593F mutant lines: Replace tyrosine with phenylalanine to prevent phosphorylation

  • Phosphomimetic Y593E mutants: Replace tyrosine with glutamic acid to mimic constitutive phosphorylation

  • Inducible DDX5 knockdown/knockout systems: Temporal control over DDX5 depletion

  • Site-specific phosphorylation sensors: FRET-based reporters to monitor Y593 phosphorylation dynamics

Animal Models:

  • Xenograft models using phospho-DDX5 (Y593) modified cell lines

  • Patient-derived xenografts (PDXs) from tumors with high phospho-DDX5 (Y593)

  • Genetically engineered mouse models with DDX5 Y593F knock-in mutations

  • Orthotopic models for tissue-specific microenvironment interactions

3D/Organoid Models:

  • Patient-derived organoids maintaining original tumor phospho-DDX5 status

  • Spheroid co-culture systems with immune cells to study microenvironment effects

  • Scaffold-based 3D cultures to assess invasion properties dependent on phospho-DDX5 (Y593)

Clinical Sample Analysis:

  • Multi-immunofluorescence tissue staining for phospho-DDX5 (Y593) and interacting partners

  • Single-cell RNA sequencing combined with phosphoproteomic analysis

  • Tissue microarrays with paired primary and metastatic samples

Researchers should select models based on specific research questions, considering the distinct phospho-DDX5 (Y593) functions observed in different cancer contexts .

How can phospho-DDX5 (Y593) status be utilized as a biomarker for cancer prognosis or treatment response?

Emerging evidence supports the potential of phospho-DDX5 (Y593) as a clinically relevant biomarker for both prognostication and prediction of treatment response across several cancer types. Implementation of phospho-DDX5 (Y593) as a biomarker requires consideration of multiple factors:

Prognostic Value:

Predictive Biomarker Applications:

  • β-catenin Pathway Inhibitors:

    • Tumors with high phospho-DDX5 (Y593) show enhanced β-catenin nuclear localization

    • These tumors may be particularly responsive to β-catenin pathway inhibitors

    • RX-5902, which attenuates DDX5 Y593 phosphorylation, shows promise in AML with high phospho-DDX5 levels

  • Rational Combination Therapy Selection:

    • Phospho-DDX5 (Y593) status may help identify tumors that would benefit from combining standard therapies with targeted phospho-DDX5 inhibitors

    • The antagonistic relationship between Y593/Y595 and T564/T446 phosphorylation suggests the potential for phosphorylation site-specific targeting

  • Immunotherapy Response Prediction:

    • DDX5 expression levels correlate with immune cell infiltration patterns

    • Low phospho-DDX5 (Y593) tumors show increased M2 macrophage infiltration, suggesting potential resistance to certain immunotherapies

    • This could inform patient selection for immunotherapy approaches

Methodological Considerations for Clinical Implementation:

  • Tissue-based Detection:

    • Immunohistochemistry with validated phospho-specific antibodies

    • Quantitative scoring systems needed (H-score or digital image analysis)

    • Standardized cutoff values for "high" vs "low" expression

  • Liquid Biopsy Potential:

    • Investigation of circulating tumor cells (CTCs) for phospho-DDX5 (Y593) status

    • Exosomal phospho-DDX5 as a non-invasive biomarker

  • Combinatorial Biomarker Approaches:

    • Integrating phospho-DDX5 (Y593) with other phosphorylation sites (Y595, T564, etc.)

    • Combining with markers of related pathways (β-catenin, AR signaling)

    • Multiparametric assessment for improved predictive power

These biomarker applications require further validation in prospective clinical studies to establish standardized protocols and clinically meaningful cutoffs before routine implementation .

How can potential cross-reactivity with other phosphorylated proteins be minimized when using phospho-DDX5 (Y593) antibodies?

Minimizing cross-reactivity is essential for accurate phospho-DDX5 (Y593) detection. Researchers should implement the following comprehensive strategies based on established immunological principles and phospho-specific antibody validation approaches:

Antibody Selection and Validation:

  • Choose antibodies raised against longer phosphopeptides (>10 amino acids) surrounding Y593

  • Select antibodies validated against multiple species to confirm recognition of the conserved epitope

  • Verify manufacturer's validation data that explicitly tests for cross-reactivity with similar proteins (especially DDX17)

  • Perform in-house validation with phosphatase treatment controls and Y593F mutants

Experimental Design Optimizations:

  • Implement a sequential immunoprecipitation approach:

    • First immunoprecipitate with total DDX5 antibody

    • Then probe with phospho-specific antibody

    • This ensures the detected phosphoprotein is specifically DDX5

  • Use DDX5 knockdown or knockout controls:

    • Include DDX5 siRNA/shRNA treated samples

    • Any remaining signal in knockdown samples indicates potential cross-reactivity

  • Peptide competition assays with graduated specificity:

    • Include phospho-Y593 DDX5 peptide

    • Include non-phosphorylated Y593 DDX5 peptide

    • Include phosphopeptides from proteins with similar sequences

    • Signal should be blocked only by the specific phospho-Y593 DDX5 peptide

Technical Adjustments:

  • Increase stringency of wash buffers:

    • Use higher salt concentrations (250-500 mM NaCl)

    • Add non-ionic detergents (0.1-0.5% Triton X-100)

    • These reduce non-specific binding

  • Optimize antibody dilutions:

    • Perform titration experiments to find minimum effective concentration

    • Working at lower antibody concentrations reduces non-specific binding

  • For blotting applications:

    • Block with 5% BSA rather than milk (phospho-epitopes can be masked by milk)

    • Add phosphatase inhibitors to all buffers

    • Consider using PVDF membranes for higher protein retention and signal-to-noise ratio

  • For immunofluorescence:

    • Include double staining with total DDX5 antibody from different species

    • True phospho-DDX5 (Y593) signal should colocalize with total DDX5

Implementing these strategies ensures that signals detected truly represent phospho-DDX5 (Y593) and not cross-reactive phosphoproteins, which is particularly important given the conserved nature of many phosphorylation motifs .

What are the advantages and limitations of different detection methods for phospho-DDX5 (Y593) in complex biological samples?

Different detection methodologies for phospho-DDX5 (Y593) offer distinct advantages and limitations when applied to complex biological samples. Understanding these tradeoffs enables researchers to select the most appropriate technique based on specific experimental objectives:

Detection MethodAdvantagesLimitationsBest Applications
Western Blotting- Well-established protocol with validated antibodies
- Semi-quantitative
- Can detect both total and phosphorylated DDX5 in parallel
- Limited spatial information
- Requires cell lysis (loses cellular context)
- Poor for low abundance detection
- Relative quantification across conditions
- Initial validation of phosphorylation status
- Monitoring changes following treatments
Immunohistochemistry- Preserves tissue architecture
- Enables correlation with pathological features
- Can identify cell-specific phosphorylation
- Semi-quantitative at best
- Fixation can mask phospho-epitopes
- Variable staining intensity
- Patient tissue samples
- Tumor microenvironment studies
- Correlating with clinical outcomes
Immunofluorescence- Higher resolution of subcellular localization
- Can be combined with other markers
- Better dynamic range than IHC
- Photobleaching issues
- Autofluorescence in some tissues
- Quantification challenges
- Subcellular localization studies
- Co-localization with interacting proteins
- Single-cell heterogeneity assessment
Phospho-Flow Cytometry- Single-cell resolution
- Quantitative
- High-throughput
- Can combine with other markers
- Limited to cell suspensions
- Requires specialized equipment
- Cell fixation/permeabilization optimization
- Heterogeneous cell populations
- Rare cell population analysis
- Drug response screening
Mass Spectrometry- Definitive phosphosite identification
- Can quantify phosphorylation stoichiometry
- Detects multiple phosphosites simultaneously
- Requires specialized equipment
- Lower sensitivity than antibody methods
- Complex sample preparation
- Discovery of novel phosphosites
- Absolute quantification
- Phosphorylation networks analysis
Proximity Ligation Assay- Detects protein-protein interactions
- Single-molecule sensitivity
- In situ detection in intact cells
- Complex protocol
- Requires optimization
- Specialized reagents
- Studying DDX5-partner interactions dependent on Y593 phosphorylation
- Rare event detection
ELISA- Quantitative
- High-throughput
- Good for large sample numbers
- No spatial information
- Requires cell lysis
- Antibody pair optimization
- Screening studies
- Biomarker quantification
- Drug screening applications

Method Selection Guidelines:

  • For initial phosphorylation status assessment: Western blot provides robust detection and semi-quantification

  • For spatial distribution in tissues: IHC or IF are preferred methods

  • For absolute quantification: Mass spectrometry provides the most definitive measurement

  • For heterogeneous populations: Phospho-flow cytometry enables single-cell analysis

  • For protein interaction studies: Proximity ligation assay or co-immunoprecipitation followed by western blot

Each method requires proper controls and validation to ensure specificity for phospho-DDX5 (Y593) detection in complex biological samples .

What are the critical considerations for designing experiments to study the functional impact of DDX5 Y593 phosphorylation?

Designing rigorous experiments to delineate the functional impact of DDX5 Y593 phosphorylation requires careful consideration of multiple factors to ensure valid and reproducible results. Based on published research approaches, the following critical considerations should guide experimental design:

1. Phosphorylation Site Manipulation Strategies:

A. Genetic Approaches:

  • Y593F mutant (cannot be phosphorylated): Essential for loss-of-function studies

  • Y593E mutant (phosphomimetic): Simulates constitutive phosphorylation

  • Y593A mutant: Alternative neutral substitution to control for structural changes

  • Double mutants (Y593F/Y595F): Control for potential compensatory phosphorylation at adjacent sites

B. Expression System Selection:

  • Inducible expression systems to control timing and expression level

  • Rescue experiments in DDX5 knockout backgrounds to eliminate endogenous protein interference

  • CRISPR knock-in mutations for studying at endogenous expression levels

2. Kinase and Phosphatase Modulation:

A. Kinase Identification and Inhibition:

  • Identify kinases responsible for Y593 phosphorylation (c-Abl, Src family implicated)

  • Use specific kinase inhibitors to modulate phosphorylation status

  • Develop inducible kinase expression systems to trigger phosphorylation

B. Phosphatase Considerations:

  • Identify phosphatases that dephosphorylate Y593

  • Use phosphatase inhibitors to maintain phosphorylation

  • Consider temporal dynamics of phosphorylation/dephosphorylation cycles

3. Functional Readout Selection:

A. Target Gene Expression:

  • β-catenin target genes (c-Myc, cyclin D1)

  • Snail1 and EMT markers (E-cadherin)

  • Androgen receptor target genes

B. Cellular Phenotypes:

  • Proliferation assays (optimized for cell type-specific growth characteristics)

  • Apoptosis assessment (particularly relevant for dual Y593/Y595 phosphorylation)

  • Cell migration and invasion assays (relevant for EMT functions)

  • Nuclear/cytoplasmic fractionation (for β-catenin localization studies)

C. Molecular Interactions:

  • Co-immunoprecipitation with interaction partners (β-catenin, HDAC1, Axin)

  • Chromatin immunoprecipitation for transcriptional targets

  • RNA immunoprecipitation for RNA binding targets (if relevant)

4. Context Dependency Considerations:

A. Cell Type Selection:

  • Include multiple cell lines representing different cancer types

  • Compare cells where DDX5 acts as oncogene vs. tumor suppressor

  • Include normal cell counterparts as controls

B. Stress and Microenvironmental Factors:

  • Test under different growth conditions (serum starvation, hypoxia)

  • Include immune cell co-culture models when studying microenvironment effects

  • Assess phosphorylation changes during drug treatments or radiation

5. Technical Controls and Validation:

A. Phosphorylation Status Verification:

  • Always confirm phosphorylation status with validated phospho-specific antibodies

  • Include phosphatase-treated controls in key experiments

  • Monitor total DDX5 levels alongside phosphorylation status

B. Specificity Controls:

  • Include DDX17 controls to ensure observed effects are DDX5-specific

  • Use multiple independent siRNAs/shRNAs for knockdown studies

  • Perform rescue experiments with phosphorylation site mutants

By addressing these critical considerations, researchers can design comprehensive experiments that provide meaningful insights into the specific functional consequences of DDX5 Y593 phosphorylation in different biological contexts .

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.