The Phospho-DOK1 (Y398) Antibody is a rabbit polyclonal antibody that specifically recognizes DOK1 phosphorylated at tyrosine residue 398 (Y398). This phosphorylation event occurs in response to growth factor signaling, such as platelet-derived growth factor (PDGF)-BB stimulation, and is critical for DOK1's role as a scaffold protein in signal transduction complexes .
Immunogen: Derived from a synthetic peptide corresponding to the Y398 phosphorylation site in human DOK1 .
Phosphorylation of DOK1 at Y398 (and Y362) is essential for its role in PDGF-BB-mediated signaling:
Mechanistic Role:
PDGF-BB activates SRC family kinases (SFKs), which phosphorylate DOK1 at Y362 and Y398 .
Phosphorylated DOK1 facilitates p130Cas tyrosine phosphorylation and subsequent activation of Rap1 GTPase, driving glioma cell migration and invasion .
Inhibition of DOK1 phosphorylation (e.g., via Y398F mutation) disrupts p130Cas–Rap1 signaling and reduces 3D tumor spheroid invasion .
This antibody is widely used to study DOK1 phosphorylation dynamics in cancer and signaling pathways:
Glioma Cell Invasion:
Kinase Dependencies:
Clinical Relevance:
DOK1 (Docking protein 1, also known as p62dok or pp62) is an enzymatically inert adaptor or scaffolding protein that provides a docking platform for the assembly of multimolecular signaling complexes . DOK1 is an abundant Ras-GTPase-activating protein-associated tyrosine kinase substrate that plays several important regulatory roles in cellular signaling :
It negatively regulates cell growth and proliferation
It promotes cell migration and motility
It appears to function as a negative regulator of the insulin signaling pathway
It modulates integrin activation by competing with talin for the same binding site on ITGB3
It regulates platelet-derived growth factor (PDGF)-BB-stimulated glioma cell motility
DOK1's activity is primarily regulated through phosphorylation at multiple tyrosine and serine residues, with Y398 being one of the critical regulatory sites.
Y398 is a key tyrosine phosphorylation site in DOK1 that becomes phosphorylated in response to various stimuli. Research indicates that phosphorylation at this site is crucial for DOK1's downstream signaling functions. Specifically:
PDGF-BB stimulates DOK1 phosphorylation on both Tyr362 and Tyr398
This phosphorylation is critical for PDGF-BB-stimulated tyrosine phosphorylation of p130Cas, a key component in cell migration signaling pathways
Phosphorylation at Y398 plays an essential role in DOK1's ability to regulate cell motility and invasion
It may influence DOK1's interaction with other signaling molecules in various cellular contexts
This site-specific phosphorylation represents a critical regulatory mechanism that determines DOK1's functional activity in various signaling pathways.
For optimal antibody performance and longevity, follow these research-validated storage and handling recommendations:
When working with this antibody, it's advisable to make small aliquots upon first thaw to minimize freeze-thaw cycles, which can degrade antibody performance over time.
The specificity of Phospho-DOK1 (Y398) antibody has been validated through multiple complementary approaches:
Western blot analysis: The antibody detects a specific band at approximately 62 kDa corresponding to phosphorylated DOK1 in extracts from K562 cells
Immunohistochemistry: Specific staining has been demonstrated in paraffin-embedded human breast carcinoma tissues
Peptide competition assays: Similar to validation approaches used for other phospho-specific DOK1 antibodies, specificity can be confirmed by demonstrating loss of signal when the antibody is pre-incubated with the phosphorylated immunizing peptide
Phosphatase treatment controls: As demonstrated with other phospho-DOK1 antibodies, lambda phosphatase treatment eliminates the specific band, confirming phospho-specificity
These validation approaches collectively establish that the antibody specifically recognizes the phosphorylated Y398 epitope of DOK1 without significant cross-reactivity to unphosphorylated DOK1 or other phosphorylated proteins.
For optimal Western blot results with Phospho-DOK1 (Y398) antibody, the following protocol elements are recommended:
Sample preparation:
Electrophoresis and transfer:
Use 8-10% SDS-PAGE gels to achieve optimal separation around the 62 kDa region
Transfer to PVDF or nitrocellulose membrane using standard protocols
Antibody incubation:
Detection:
Controls:
For immunohistochemical detection of phosphorylated DOK1 (Y398) in tissue samples:
Sample preparation:
Fix tissues in formalin and embed in paraffin
Section tissues at 4-6 μm thickness
Mount on positively charged slides
Deparaffinization and antigen retrieval:
Deparaffinize in xylene and rehydrate through graded alcohols
Perform heat-induced epitope retrieval in citrate buffer (pH 6.0) or EDTA buffer (pH 9.0)
Cool slides to room temperature
Staining procedure:
Block endogenous peroxidase activity with 3% H₂O₂
Block non-specific binding with 5% normal goat serum
Incubate overnight at 4°C or 1-2 hours at room temperature
Detect using an appropriate detection system (e.g., HRP-polymer and DAB)
Counterstain with hematoxylin, dehydrate, and mount
Controls:
DOK1 Y398 phosphorylation plays a crucial role in PDGF-BB signaling cascades, particularly in the context of cell motility and migration:
Activation mechanism:
Downstream effectors:
Functional consequences:
When Y362 and Y398 are mutated to phenylalanine (DOK1FF), PDGF-BB-stimulated p130Cas tyrosine phosphorylation is significantly decreased
DOK1FF expression also results in decreased Rap1 activation in response to PDGF-BB
These molecular changes translate to inhibition of cell motility and invasiveness
This signaling axis represents a critical mechanism by which PDGF-BB promotes cell migration in various cell types, including glioma cells, with potential implications for understanding tumor cell invasion.
DOK1 undergoes complex regulation through both tyrosine and serine phosphorylation, with cross-talk between these modifications:
Tyrosine phosphorylation sites:
Serine phosphorylation sites:
Functional interplay:
Serine phosphorylation is required for DOK1's ability to inhibit PDGF-induced ERK1/2 activation and cell proliferation
DOK1 with mutations in these serine residues (S439A, S443A, S446A, S450A) fails to inhibit cell proliferation or promote cell motility
Phosphorylation of Y450 may mimic the acidic residues that facilitate IKKβ recognition and enhance the association of tyrosine-phosphorylated DOK1 with IKKβ
This dual phosphorylation system represents a sophisticated regulatory mechanism allowing DOK1 to integrate signals from multiple upstream pathways and modulate diverse cellular responses.
Mutation studies have provided critical insights into the functional significance of specific DOK1 phosphorylation sites:
Tyrosine site mutations:
Mutation of Y362 and Y398 to phenylalanine (DOK1FF) prevents PDGF-BB-stimulated phosphorylation
DOK1FF acts in a dominant-negative manner, competing with endogenous DOK1 binding partners to form non-functional complexes
This mutant significantly decreases PDGF-BB-stimulated p130Cas tyrosine phosphorylation and Rap1 activation
Functionally, DOK1FF inhibits spheroid outgrowth, cell motility, and invasion
Serine site mutations:
Phosphomimetic mutations:
These mutation studies collectively demonstrate that both tyrosine and serine phosphorylation events are essential for DOK1's biological functions, particularly in regulating cell proliferation, motility, and migration.
The phospho-DOK1 (Y398) antibody serves as a valuable tool for investigating tumor cell invasion and migration through several experimental approaches:
Correlation with invasive phenotypes:
Pathway analysis in 3D models:
Response to microenvironmental cues:
Western blotting with phospho-DOK1 (Y398) antibody can measure how various extracellular stimuli (growth factors, ECM components, inflammatory mediators) trigger DOK1 activation
Co-immunoprecipitation coupled with phospho-DOK1 detection can identify context-specific protein complexes formed during invasion
Therapeutic intervention assessment:
Monitoring changes in DOK1 Y398 phosphorylation following treatment with kinase inhibitors or other therapeutic agents can provide mechanistic insights into drug effects on cell migration
This approach has been demonstrated in studies using PI3K inhibitors like LY294002, which reduce PDGF-stimulated DOK1 phosphorylation
These applications make phospho-DOK1 (Y398) antibody an important tool for understanding the molecular mechanisms underlying tumor cell invasion and for developing therapeutic strategies targeting metastasis.
Understanding the structural changes induced by Y398 phosphorylation requires sophisticated biophysical and biochemical approaches:
Protein crystallography and structural biology:
While the crystal structure of the DOK1 PTB domain has been determined , similar approaches could be applied to study larger DOK1 fragments containing the Y398 site
Comparative crystallography of phosphorylated versus non-phosphorylated DOK1 could reveal conformational changes induced by Y398 phosphorylation
Hydrogen-deuterium exchange mass spectrometry (HDX-MS):
This technique can identify regions of DOK1 that undergo changes in solvent accessibility upon Y398 phosphorylation
HDX-MS can reveal allosteric effects of phosphorylation on distant protein domains
Nuclear magnetic resonance (NMR) spectroscopy:
NMR can provide dynamic information about conformational changes induced by phosphorylation
Chemical shift perturbation experiments can map the effects of Y398 phosphorylation on the entire protein structure
Molecular dynamics simulations:
Computational approaches can model the structural consequences of Y398 phosphorylation
Simulations can predict changes in protein flexibility, surface properties, and binding interfaces
Protein-protein interaction studies:
Phospho-specific pull-down assays using synthetic peptides containing phosphorylated Y398 can identify phosphorylation-dependent binding partners
Comparison with similar assays using non-phosphorylated peptides can define phosphorylation-specific interactions
These approaches provide complementary information about how Y398 phosphorylation affects DOK1 structure and function, potentially revealing mechanisms by which this modification regulates DOK1's scaffolding activities.
Integrating phospho-DOK1 (Y398) detection with other signaling markers enables comprehensive mapping of interconnected pathways:
Multiplex immunofluorescence or immunohistochemistry:
Co-staining tissues for phospho-DOK1 (Y398) and other phosphorylated signaling proteins (e.g., phospho-p130Cas, phospho-AKT, phospho-ERK) can reveal spatial relationships between different activated pathways
This approach can identify cell populations with coordinated signaling activation within heterogeneous tissues
Phosphoproteomic analysis:
Mass spectrometry-based phosphoproteomics can quantify changes in DOK1 Y398 phosphorylation alongside hundreds of other phosphorylation events
This global approach can identify novel signaling relationships between DOK1 and other pathways
Phosphoproteomic time-course experiments can establish the temporal sequence of phosphorylation events following stimulus
Multi-parameter flow cytometry:
For single-cell analysis, phospho-flow cytometry can simultaneously measure DOK1 Y398 phosphorylation and other phosphoproteins
This technique is particularly valuable for analyzing heterogeneous cell populations like tumor samples or mixed immune cells
Reverse-phase protein arrays (RPPA):
RPPA allows high-throughput, quantitative analysis of many phosphorylated proteins, including phospho-DOK1 (Y398), across large sample sets
This approach is ideal for screening the effects of multiple treatments or genetic perturbations on DOK1 signaling in the context of broader pathway activation
Computational pathway analysis:
Integration of phospho-DOK1 data with other phosphoprotein measurements allows computational reconstruction of signaling networks
Machine learning approaches can identify patterns of coordinated phosphorylation events associated with specific cellular phenotypes
These integrative approaches provide a systems-level understanding of how DOK1 Y398 phosphorylation fits into broader signaling networks regulating cell behavior.
Researchers may encounter several challenges when working with phospho-DOK1 (Y398) antibodies:
Low signal intensity:
Cause: Rapid dephosphorylation during sample preparation or low abundance of phosphorylated protein
Solution: Include phosphatase inhibitors (sodium orthovanadate, sodium fluoride, β-glycerophosphate) in lysis buffers; enrich phosphoproteins using immunoprecipitation prior to Western blotting
High background or non-specific bands:
Cause: Insufficient blocking, cross-reactivity with similar phosphoepitopes, or incomplete antibody optimization
Solution: Use alternative blocking agents (5% BSA is preferred over milk for phospho-antibodies); increase washing time and volume; optimize antibody dilution through titration experiments
Inconsistent phosphorylation levels:
Cause: Variation in cell culture conditions or serum starvation protocols affecting basal phosphorylation
Solution: Standardize culture conditions; implement consistent serum starvation periods (typically 16-24 hours) before stimulation
Phospho-epitope masking in IHC applications:
Cause: Formalin fixation can mask phospho-epitopes through protein cross-linking
Solution: Optimize antigen retrieval methods; test both citrate (pH 6.0) and EDTA (pH 9.0) buffers; extend retrieval time if necessary
Variable phosphorylation kinetics:
Cause: Phosphorylation may be transient or have cell type-specific temporal patterns
Solution: Perform detailed time-course experiments to determine optimal time points for phosphorylation detection after stimulation
Implementing these technical solutions can significantly improve the reliability and sensitivity of phospho-DOK1 (Y398) detection in experimental applications.
Rigorous validation of phospho-specificity is essential for reliable interpretation of experimental results:
Peptide competition assays:
Pre-incubate the antibody with the phosphorylated immunizing peptide before application
A specific signal should be abolished or substantially reduced
Pre-incubation with the corresponding non-phosphorylated peptide should not affect signal intensity
Phosphatase treatment controls:
siRNA or CRISPR knockdown validation:
Reduce DOK1 expression using RNA interference or gene editing
A specific signal should decrease proportionally to the reduction in total DOK1
This approach confirms that the detected signal derives from DOK1 rather than cross-reactive proteins
Stimulation experiments:
Mutation studies:
Express wild-type DOK1 or Y398F mutant in cells with endogenous DOK1 knockdown
Only wild-type DOK1 should show reactivity with the phospho-specific antibody
This approach definitively confirms epitope specificity
These validation strategies provide complementary evidence for phospho-specificity and should be selected based on the specific experimental context and available resources.
Studying the interplay between tyrosine and serine phosphorylation of DOK1 requires careful experimental design:
Buffer optimization for comprehensive phosphorylation detection:
Include inhibitors targeting both tyrosine and serine/threonine phosphatases (sodium orthovanadate, sodium fluoride, β-glycerophosphate)
Protect from degradation using protease inhibitors
Maintain sample temperature at 4°C during processing to minimize dephosphorylation
Sequential immunoprecipitation strategies:
Initial immunoprecipitation with phospho-tyrosine antibodies followed by immunoblotting with phospho-serine antibodies (or vice versa)
This approach can reveal populations of DOK1 with both modifications
Alternatively, immunoprecipitate with total DOK1 antibodies and probe with phospho-specific antibodies
Time-course experiments:
Pathway inhibitor studies:
Mass spectrometry-based approaches:
Phosphopeptide mapping can simultaneously identify multiple phosphorylation sites
This approach can determine whether specific tyrosine and serine phosphorylations co-occur on the same DOK1 molecule
Quantitative MS can measure the stoichiometry of different phosphorylation events
These considerations help researchers design experiments that capture the complex interplay between different phosphorylation events on DOK1 and their functional consequences.
Several promising research directions are emerging at the intersection of DOK1 phosphorylation and cancer biology:
DOK1 as a biomarker for therapeutic response:
Investigating whether phospho-DOK1 (Y398) levels predict response to receptor tyrosine kinase inhibitors or other targeted therapies
Determining if DOK1 phosphorylation status correlates with response to immunotherapy or conventional chemotherapy
This could lead to the development of companion diagnostic assays using phospho-DOK1 antibodies
DOK1 in the tumor microenvironment:
Exploring how DOK1 phosphorylation in stromal cells (fibroblasts, immune cells) influences tumor-stroma interactions
Investigating whether cancer cells and stromal cells exhibit different patterns of DOK1 phosphorylation
This could reveal new mechanisms of tumor-stroma communication
DOK1 in therapy resistance mechanisms:
Determining if altered DOK1 phosphorylation contributes to acquired resistance to targeted therapies
Investigating whether combined inhibition of pathways that regulate DOK1 phosphorylation can overcome resistance
This may lead to rational combination therapy approaches
DOK1 in cancer stem cell biology:
Examining whether cancer stem cells exhibit distinct patterns of DOK1 phosphorylation
Investigating if DOK1 signaling contributes to stem cell maintenance or differentiation in tumors
This could identify new therapeutic vulnerabilities in treatment-resistant cancer stem cells
Integration with immune checkpoint regulation:
Exploring potential roles of DOK1 phosphorylation in immune cell function within the tumor microenvironment
Investigating whether DOK1 signaling interacts with immune checkpoint pathways
This may reveal unexpected connections between DOK1 and immunotherapy response
These emerging research areas highlight the potential significance of DOK1 phosphorylation in various aspects of cancer biology and therapeutic response.
Cutting-edge technologies are poised to revolutionize our understanding of DOK1 phosphorylation:
Live-cell phosphorylation sensors:
Genetically encoded FRET-based biosensors can monitor DOK1 phosphorylation in real-time in living cells
This approach would reveal spatiotemporal dynamics of DOK1 activation with subcellular resolution
Could identify previously unknown compartmentalization of DOK1 signaling
Single-cell phosphoproteomics:
Emerging mass spectrometry techniques allow phosphoproteomic analysis at the single-cell level
This could reveal heterogeneity in DOK1 phosphorylation within cell populations
May identify rare cell subpopulations with distinct DOK1 activation patterns
CRISPR-based phosphorylation screens:
CRISPR activation/interference libraries targeting kinases and phosphatases
Readout based on phospho-DOK1 (Y398) levels
This approach could identify novel regulators of DOK1 phosphorylation
Intravital microscopy with phospho-specific probes:
In vivo imaging of DOK1 phosphorylation in tumor models
Could reveal dynamic changes in DOK1 activation during processes like metastatic invasion
May identify spatiotemporal relationships between DOK1 activation and tissue microenvironments
Proximity labeling proteomics:
BioID or APEX2 fused to DOK1 to identify proteins that interact specifically with phosphorylated DOK1
Comparing interactomes of wild-type versus phospho-deficient DOK1 mutants
This approach could map phosphorylation-dependent protein interaction networks
These technological advances will provide unprecedented insights into the dynamics, regulation, and functional consequences of DOK1 phosphorylation in normal and disease states.
Interdisciplinary research strategies hold particular promise for advancing our understanding of DOK1 Y398 phosphorylation:
Systems biology and computational modeling:
Integration of phosphoproteomic data with transcriptomic and metabolomic datasets
Mathematical modeling of DOK1 signaling networks to predict system responses to perturbations
This approach could identify non-intuitive relationships between DOK1 phosphorylation and cellular phenotypes
Patient-derived models and precision medicine:
Analysis of DOK1 phosphorylation in patient-derived xenografts and organoids
Correlation with treatment response and clinical outcomes
Could lead to personalized therapeutic strategies based on DOK1 phosphorylation status
Structural biology and drug discovery:
Structure-based design of compounds that specifically target phosphorylated DOK1 or its interaction interfaces
Development of proteolysis-targeting chimeras (PROTACs) that selectively degrade phosphorylated DOK1
This could yield first-in-class therapeutics targeting DOK1-dependent pathways
Immunology and cancer biology integration:
Investigation of DOK1 phosphorylation in tumor-infiltrating immune cells
Examination of how tumor-derived factors affect DOK1 phosphorylation in immune cells
May reveal immunomodulatory roles of DOK1 signaling in the tumor microenvironment
Developmental biology perspectives:
Comparative analysis of DOK1 phosphorylation in embryonic development and cancer progression
Identification of developmental programs that are aberrantly reactivated in cancer
Could reveal fundamental mechanisms underlying DOK1's role in cell migration and invasion
These interdisciplinary approaches combine diverse methodologies and perspectives to address complex questions about DOK1 phosphorylation that cannot be answered through any single discipline alone.