Phospho-KDR (Tyr1175) refers to the vascular endothelial growth factor receptor 2 (VEGFR2/KDR/Flk-1) specifically phosphorylated at tyrosine residue 1175. This phosphorylation site represents one of the major VEGF-dependent autophosphorylation sites on KDR and plays a crucial role in endothelial cell signaling . The significance of this specific phosphorylation lies in its function as a molecular switch that activates multiple downstream signaling pathways critical for angiogenesis, vascular permeability, and endothelial cell proliferation . Mutations at this site have profound effects on vascular development, making phospho-KDR (Tyr1175) antibodies valuable tools for studying VEGF-mediated signaling in both normal physiology and pathological conditions.
KDR/VEGFR2 contains multiple tyrosine phosphorylation sites, with the most prominently studied being Tyr951, Tyr1054, Tyr1059, Tyr1175, and Tyr1214. Among these, Tyr1175 is distinguished by its specific role as the primary binding site for phospholipase C-gamma (PLC-γ), which activates the PKC-MAP kinase pathway crucial for endothelial cell proliferation . While Tyr951 and Tyr996 are located in the receptor's kinase insert domain and Tyr1054/1059 in the catalytic domain , Tyr1175 phosphorylation uniquely triggers both PLC-γ-dependent and PKA-dependent signaling pathways .
Research has demonstrated that mutation of Tyr1175 to phenylalanine (Y1175F) specifically abolishes PLC-γ activation and significantly reduces MAP kinase phosphorylation and DNA synthesis in response to VEGF-A, whereas mutations at other sites produce distinct phenotypes . This makes Tyr1175 phosphorylation particularly critical for vascular development compared to other phosphorylation sites.
Phosphorylated KDR at Tyr1175 is predominantly found in:
Notably, research has shown that human umbilical vein endothelial cells (HUVECs) are frequently used as a positive control for phospho-KDR (Tyr1175) detection . In tumors, phosphorylated KDR can be localized not only at the cell membrane but also within the nucleus of neoplastic cells, particularly after combined hypoxic and VEGF stimulation .
The expression pattern varies significantly based on:
Activation state (VEGF stimulation)
Oxygen conditions (hypoxia increases expression)
Cellular context (normal vs. neoplastic)
Subcellular localization (membrane, cytoplasm, nucleus)
Multiple detection methodologies are suitable for studying Phospho-KDR (Tyr1175), each with specific advantages:
The HTRF-based assay offers unique advantages for quantitative assessment, as it uses two labeled antibodies: one specific for the phosphorylated motif on Tyr1175 and another that recognizes KDR independently of its phosphorylation state. This creates a FRET signal directly proportional to the concentration of phosphorylated protein in the sample .
Designing robust experiments to study KDR Tyr1175 phosphorylation requires careful consideration of multiple factors:
Mutation Studies:
Create Y1175F KDR mutants (tyrosine to phenylalanine substitution) to abolish phosphorylation at this specific site
Express mutated constructs in relevant cell lines (e.g., 293 cells or endothelial cells) using adenoviral vectors or transfection
Compare biological responses (proliferation, migration, tube formation) between wild-type and mutant KDR-expressing cells
Pharmacological Approaches:
Use specific inhibitors of KDR kinase activity (e.g., SU1498) with appropriate controls
Include proteasome/lysosome inhibitors (e.g., leupeptin, Bafilomycin A1) when necessary to prevent protein degradation
Consider time-course experiments to capture both rapid (minutes) and delayed (hours) phosphorylation events
Stimulation Protocols:
Controls to Include:
Well-designed experiments should incorporate multiple readouts to assess both the phosphorylation status and its functional consequences on downstream signaling pathways and cellular behaviors.
Successful Western blot detection of Phospho-KDR (Tyr1175) requires attention to several critical parameters:
Sample Preparation:
Gel Electrophoresis:
Transfer and Blocking:
Extended transfer times or specialized transfer methods may be needed for the high molecular weight KDR
BSA-based blocking solutions (rather than milk) are recommended to prevent interference with phospho-epitopes
Antibody Selection and Dilution:
Use phospho-specific antibodies validated for Western blot applications
Include total KDR antibody on parallel blots or after stripping
Optimize antibody dilutions and incubation conditions empirically
Detection and Analysis:
Following these parameters will help ensure reliable and reproducible detection of Phospho-KDR (Tyr1175) in Western blot applications.
Phospho-KDR (Tyr1175) serves as a critical hub for multiple signaling pathways in endothelial cells:
PLC-γ Pathway:
PKA-dependent Signaling:
MAPK Cascade:
Calcium Signaling:
The integration of these signaling cascades downstream of Phospho-KDR (Tyr1175) orchestrates complex cellular responses including proliferation, migration, survival, and specialized endothelial functions like vWF release. Disruption of this signaling node through Y1175F mutation has profound effects on endothelial cell biology and vascular development.
Research has revealed a complex relationship between KDR nuclear localization and Tyr1175 phosphorylation:
Nuclear Translocation Phenomenon:
Phosphorylated KDR can be located in the nucleus of both normal and neoplastic cells
This nuclear localization was initially observed in animal cell lines and subsequently confirmed in human cells
The translocation appears to be regulated by both VEGF stimulation and environmental factors like hypoxia
Differential Response to Stimuli:
In human neoplastic cell lines, hypoxic stimulation greatly increases nuclear localization of total KDR but has less effect on the phosphorylated form
Combined hypoxia and VEGF stimulation leads to comparable increases in both phosphorylated and total KDR in the nucleus
This suggests differential regulation of KDR phosphorylation and localization
Functional Significance:
Nuclear KDR may have distinct functions from membrane-bound receptor
Although the precise functional meaning of nuclear localization remains to be fully established, it may represent a direct communication mechanism between activated receptors and nuclear processes
This pathway might bypass traditional cytoplasmic signaling cascades
Detection Methods:
The discovery of nuclear-localized phosphorylated KDR adds complexity to our understanding of VEGF signaling and suggests additional mechanisms by which KDR activation may influence cellular function beyond traditional membrane-initiated signaling cascades.
Mutations affecting the Tyr1175 region of KDR have significant implications in pathological conditions:
Embryonic Development:
Cancer Biology:
In neoplastic cells, alterations in KDR phosphorylation at Tyr1175 may contribute to dysregulated angiogenesis
Tumors show variable expression patterns of phosphorylated KDR in the nucleus, suggesting heterogeneous signaling mechanisms
These variations might influence tumor vascularization and response to anti-angiogenic therapies
Therapeutic Implications:
The Tyr1175 region represents a potential therapeutic target for anti-angiogenic strategies
Targeted disruption of this specific phosphorylation site could provide more selective inhibition compared to general KDR kinase inhibitors
Understanding the precise signaling downstream of Tyr1175 may help identify patients more likely to respond to anti-VEGF therapies
Research into Novel Mutations:
The PhosphoAtlas database has identified mutations that recurrently perturb specific phospho-hubs in the human phospho-reactome
In silico mapping of tumor variants has revealed how tumors systematically hijack particular sub-networks
These approaches may identify novel mutations affecting the Tyr1175 region and its signaling in various cancer types
The specificity of antibodies recognizing Phospho-KDR (Tyr1175) makes them valuable tools for studying these pathological conditions and evaluating the efficacy of targeted therapies.
When investigating cross-talk between KDR phosphorylation and other signaling pathways, researchers should consider:
Temporal Dynamics:
VEGF binding to cells expressing native KDR leads to rapid increase in receptor and PLC-γ phosphorylation, followed by slower increases in p125FAK and paxillin phosphorylation
Design time course experiments (minutes to hours) to capture both immediate and delayed signaling events
Consider both acute and chronic VEGF stimulation protocols
Comprehensive Phosphorylation Analysis:
Mutational Approaches:
Inhibitor Studies:
Employ pathway-specific inhibitors to dissect signaling cross-talk
Use both broad and specific kinase inhibitors with appropriate controls
Include inhibitors of phosphatases to understand dynamic regulation
Cellular Context:
Primary endothelial cells may respond differently than immortalized cell lines
Consider tissue-specific endothelial cells for more physiologically relevant insights
Include both normal and pathological (e.g., tumor-derived) endothelial cells
Environmental Factors:
By systematically addressing these factors, researchers can design experiments that effectively uncover the complex cross-talk between KDR phosphorylation and other signaling networks.
Rigorous validation of Phospho-KDR (Tyr1175) antibodies is essential for experimental reliability. Researchers should implement the following validation strategies:
Positive and Negative Controls:
Mutant Expression:
Peptide Competition:
Perform peptide competition assays using the phosphorylated peptide epitope
Include non-phosphorylated peptide controls to confirm phospho-specificity
Titrate peptide concentrations to demonstrate dose-dependent inhibition
Multiple Detection Methods:
Validate antibody specificity across multiple applications (Western blot, immunofluorescence, ELISA)
Compare results between different detection methods
Confirm subcellular localization patterns are consistent with published literature
Cross-Reactivity Testing:
Test for cross-reactivity with other phosphorylated tyrosine sites on KDR (e.g., pY951, pY1214)
Assess potential cross-reactivity with related receptor tyrosine kinases
Confirm species specificity when working with non-human models
Recombinant Protein Controls:
Use in vitro kinase assays with recombinant KDR to generate defined phosphorylation states
Compare antibody reactivity with phosphorylated versus non-phosphorylated recombinant proteins
A comprehensive validation approach combining these strategies will ensure reliable and specific detection of Phospho-KDR (Tyr1175) in experimental settings.
When designing phospho-KDR assays for drug screening applications, several key considerations should be addressed:
Assay Format Selection:
HTRF-based assays offer significant advantages for high-throughput screening due to their homogeneous (no-wash) format, quantitative readout, and compatibility with 384-well plates
Consider the trade-offs between throughput, sensitivity, and information content when selecting an assay format
Automation compatibility should be evaluated for large-scale screening campaigns
Cell Model Optimization:
Select appropriate cell models (e.g., HUVECs, engineered cell lines)
Optimize cell density and culture conditions for consistent KDR expression
Consider using stable cell lines expressing KDR to reduce variability
For the 2-plate protocol, ensure consistent cell transfer between culture and detection plates
Stimulation Parameters:
Assay Controls and Quality Metrics:
Include positive controls (VEGF stimulation), negative controls (unstimulated), and technical controls
Establish acceptance criteria for Z' factor, signal-to-background ratio, and coefficient of variation
Incorporate control lysates with known phospho-KDR (Tyr1175) levels
Include KDR kinase inhibitors (e.g., SU1498) as reference compounds
Counter-Screening Strategy:
Design parallel assays to identify false positives (e.g., compounds affecting assay readout rather than KDR phosphorylation)
Consider counter-screens for cytotoxicity and general kinase inhibition
Include orthogonal assays (e.g., Western blot, cell-based functional assays) for hit validation
Data Analysis Approach:
Develop standardized data normalization methods
Establish dose-response parameters for compound characterization
Consider multiplexed readouts to simultaneously assess total KDR and phospho-KDR
Implement robust statistical methods appropriate for high-throughput data
By carefully addressing these considerations, researchers can develop phospho-KDR (Tyr1175) assays suitable for reliable drug screening applications in angiogenesis, cancer, and vascular disease research.
Researchers frequently encounter several challenges when detecting Phospho-KDR (Tyr1175). Here are common issues and their solutions:
Low Signal Intensity:
High Background:
Cause: Non-specific antibody binding, inadequate blocking, or contaminating phosphoproteins
Solution: Use BSA instead of milk for blocking, optimize antibody dilutions, include additional washing steps, and consider using more selective antibodies
Inconsistent Results:
Detection of Multiple Bands:
Cause: Protein degradation, alternative splice variants, or cross-reactivity
Solution: Add protease inhibitors to all buffers, confirm band identity with total KDR antibody, and validate with additional phospho-specific antibodies
Poor Resolution of Phosphorylated Species:
Nuclear Localization Challenges:
Sample Degradation:
Cause: Rapid dephosphorylation during sample processing
Solution: Process samples rapidly at cold temperatures, add phosphatase inhibitors immediately, and consider direct lysis in SDS sample buffer for Western blot applications
By anticipating and addressing these challenges, researchers can significantly improve the reliability and sensitivity of Phospho-KDR (Tyr1175) detection in various experimental settings.
Phosphoproteomics offers powerful capabilities for comprehensively analyzing KDR signaling networks:
Global Phosphorylation Landscape:
Phosphoproteomics can simultaneously identify hundreds of tyrosine phosphorylated peptides altered in response to KDR modulation
This approach revealed 491 tyrosine phosphorylated peptides significantly affected by KDR inhibition in one study
Enables unbiased discovery of novel KDR-dependent phosphorylation events
Pathway Mapping:
Phosphoproteomic analysis has identified significant alterations in JAK-STAT pathway components (JAK1, JAK2, JAK3, STAT1) following KDR inhibition
MAP Kinases (ERK1, ERK2, JNK1, JNK2, p38) phosphorylation status can be simultaneously monitored
Allows comprehensive pathway mapping beyond predicted signaling nodes
Integration with Computational Tools:
The PhosphoAtlas database has created a comprehensive map of human kinase circuits by curating data from 38 public resources
This resource catalogs 2,617 stringently verified heptameric peptide regions (HPRs) at the catalytic interface of kinases and substrates
Enables in silico mapping of mutations that may perturb phosphorylation networks
Temporal Dynamics:
Time-resolved phosphoproteomics can capture the sequence of phosphorylation events following KDR activation
Reveals primary (direct) versus secondary (downstream) phosphorylation events
Helps establish causality in complex signaling networks
Therapeutic Target Identification:
Methodological Considerations:
By applying phosphoproteomics approaches, researchers can move beyond the study of individual phosphorylation sites to comprehensively understand the complex and interconnected signaling networks downstream of KDR activation.
Several cutting-edge techniques are advancing our ability to monitor KDR phosphorylation dynamics in real-time:
FRET-Based Biosensors:
Genetically encoded FRET biosensors containing KDR phosphorylation motifs can report on phosphorylation events in living cells
These constructs typically contain a phospho-binding domain, a specific KDR substrate sequence, and FRET donor/acceptor pairs
Provides temporal and spatial information on phosphorylation dynamics with subcellular resolution
Can be combined with other fluorescent markers to correlate phosphorylation with cellular processes
Phospho-Specific Nanobodies:
Single-domain antibody fragments (nanobodies) recognizing Phospho-KDR (Tyr1175) can be used for intracellular tracking
When fused to fluorescent proteins, these enable visualization of phosphorylation events in living cells
Smaller size compared to conventional antibodies allows better access to subcellular compartments
Live-Cell Immunofluorescence:
Cell-permeable phospho-specific antibody fragments conjugated to quantum dots or organic dyes
Enables tracking of phosphorylation in living cells without fixation artifacts
Requires careful validation to ensure specificity and minimal perturbation of cellular processes
Time-Resolved HTRF Assays:
Mass Spectrometry with Stable Isotope Labeling:
Pulse-chase experiments with stable isotope labeling (SILAC, TMT) allow tracking of phosphorylation dynamics
Provides quantitative data on phosphorylation site occupancy over time
Can be combined with phosphatase inhibitor washout experiments to determine site-specific dephosphorylation rates
Microfluidic Approaches:
Microfluidic devices coupled with on-chip immunoassays enable real-time monitoring with precise control of stimulation
Allow rapid solution exchange to study acute responses
Can be integrated with imaging systems for simultaneous visualization of cellular responses
Optogenetic Control of KDR Activity:
Light-controllable KDR variants allow precise spatial and temporal activation
When combined with phosphorylation sensors, enables unprecedented control over signaling dynamics
Facilitates dissection of downstream pathway activation thresholds and kinetics
These emerging techniques are transforming our ability to study KDR phosphorylation from static snapshots to dynamic processes, providing new insights into the temporal and spatial regulation of VEGF signaling.
Understanding Phospho-KDR (Tyr1175) signaling offers multiple opportunities for developing more effective anti-angiogenic therapies:
Site-Specific Targeting Approach:
The critical role of Tyr1175 phosphorylation in endothelial cell proliferation makes it an attractive therapeutic target
Compounds specifically disrupting Tyr1175 phosphorylation or its downstream interactions may provide more selective inhibition than general KDR inhibitors
This specificity could potentially reduce off-target effects associated with broader kinase inhibition
Overcoming Resistance Mechanisms:
Research has shown that Y1175 is a molecular switch for multiple signaling pathways
Understanding the complete signaling network downstream of this phosphorylation site may reveal bypass mechanisms leading to therapy resistance
Targeting multiple nodes in the network simultaneously could prevent the development of resistance
Biomarker Development:
Phospho-KDR (Tyr1175) levels could serve as biomarkers for predicting response to anti-angiogenic therapies
The nuclear localization of phosphorylated KDR in neoplastic cells might provide additional prognostic information
Monitoring phosphorylation changes during treatment could help identify early signs of resistance
Combination Therapy Rationale:
Phosphoproteomics studies have revealed that KDR inhibition affects multiple signaling pathways including JAK-STAT and MAPK cascades
This provides a scientific basis for combining KDR inhibitors with other targeted therapies
Such combinations might produce synergistic effects by blocking multiple aspects of tumor angiogenesis
Novel Target Identification:
Precision Medicine Applications:
By focusing on the specific signaling node of Phospho-KDR (Tyr1175) rather than broadly targeting KDR kinase activity, researchers may develop more refined therapeutic approaches with improved efficacy and reduced toxicity.
Several promising research directions are emerging for understanding KDR phosphorylation in angiogenesis:
Spatiotemporal Regulation:
Investigating how KDR phosphorylation patterns differ across vascular beds and developmental stages
Understanding how local microenvironmental factors influence site-specific phosphorylation
Mapping phosphorylation dynamics during vessel formation, maturation, and regression
Cross-Talk with Other Receptor Systems:
Exploring how KDR phosphorylation is influenced by other receptor tyrosine kinases (e.g., EGFR, FGFR)
Understanding the integration of multiple growth factor signals at the level of shared downstream pathways
Investigating how Phospho-KDR (Tyr1175) interacts with other signaling hubs in endothelial cells
Nuclear Function of Phosphorylated KDR:
Endothelial Cell Heterogeneity:
Characterizing differences in KDR phosphorylation patterns across diverse endothelial cell populations
Understanding how these differences contribute to organ-specific vascular biology
Exploring how endothelial heterogeneity influences response to anti-angiogenic therapies
Metabolic Regulation of KDR Phosphorylation:
Investigating how cellular metabolism influences KDR phosphorylation and signaling
Understanding the reciprocal relationship between KDR activation and metabolic adaptation
Exploring metabolic vulnerabilities in pathological angiogenesis
Single-Cell Phosphoproteomics:
Developing techniques to analyze KDR phosphorylation at single-cell resolution
Understanding cell-to-cell variability in phosphorylation responses
Identifying rare cell populations with distinct phosphorylation patterns
Computational Modeling:
Creating predictive models of KDR phosphorylation dynamics and downstream signaling
Simulating the effects of therapeutic interventions on the phosphorylation network
Integrating multi-omics data to understand system-level responses to KDR activation
These research directions promise to deepen our understanding of how KDR phosphorylation regulates both normal and pathological angiogenesis, potentially leading to novel therapeutic strategies for various vascular diseases.
Systematic phosphoproteomic mapping offers transformative potential for understanding and improving KDR-targeted therapies:
Network-Level Resistance Mechanisms:
Comprehensive phosphoproteomic analysis before and after KDR inhibition can reveal adaptive reprogramming of kinase cascades
Identification of compensatory phosphorylation events that maintain downstream signaling despite KDR inhibition
Discovery of feedback loop mechanisms that contribute to therapeutic resistance
Patient Stratification Strategies:
Phosphoproteomic profiling may identify distinct patterns of pathway activation in different patient subgroups
These patterns could predict differential responses to KDR-targeted therapies
Enables more precise patient selection for clinical trials and treatment
Rational Combination Therapies:
Mapping reveals how kinase networks are interconnected and identifies critical nodes where multiple pathways converge
This knowledge can guide the development of combination therapies targeting complementary signaling pathways
For example, phosphoproteomics has identified the JAK-STAT pathway as significantly affected by KDR inhibition, suggesting potential synergy between KDR and JAK inhibitors
Biomarker Development:
Phosphoproteomic analysis can identify key phosphorylation events that correlate with response or resistance to therapy
These phosphorylation sites could serve as pharmacodynamic biomarkers to monitor treatment efficacy
May enable early detection of emerging resistance before clinical progression
Understanding On-Target vs. Off-Target Effects:
Comprehensive phosphoproteomic profiling can distinguish direct targets of KDR inhibitors from downstream or off-target effects
This knowledge is crucial for differentiating therapeutic effects from mechanisms contributing to toxicity
Could guide the development of next-generation inhibitors with improved specificity profiles
Tumor-Specific Network Vulnerabilities:
Drug Repurposing Opportunities:
Phosphoproteomic data may reveal unexpected connections between KDR signaling and other pathways
These connections could suggest novel applications for existing drugs that target connected pathways
Accelerates therapeutic development by leveraging compounds with established safety profiles
By implementing systematic phosphoproteomic mapping approaches, researchers can gain unprecedented insights into the complex responses to KDR-targeted therapies, ultimately leading to more effective treatment strategies for angiogenesis-dependent diseases.