KEGG: ddi:DDB_G0286627
STRING: 44689.DDB0229378
For optimal preservation of enzymatic activity, recombinant DDB_G0286627 should be stored at -20°C or -80°C for extended storage periods in appropriate buffer conditions . The protein is typically supplied in a Tris-based buffer with 50% glycerol or similar formulations optimized for stability . For working solutions, aliquots should be maintained at 4°C for no more than one week to preserve activity .
When handling the protein:
Briefly centrifuge vials before opening to bring contents to the bottom
Reconstitute lyophilized protein in deionized sterile water to a concentration of 0.1-1.0 mg/mL
Add glycerol to a final concentration of 5-50% for long-term storage
Avoid repeated freeze-thaw cycles as this can significantly diminish protein activity and stability
When designing experiments to study the kinase activity of DDB_G0286627, implement the following controls:
Negative controls: Include reaction mixtures without the kinase or without substrate to account for background phosphorylation and non-specific signals.
Positive controls: Incorporate well-characterized kinases with known activity profiles (e.g., PKA or PKC) to validate assay conditions.
Inhibitor controls: Use broad-spectrum serine/threonine kinase inhibitors (e.g., staurosporine) and compare with specific inhibitors to examine selectivity profiles.
Heat-inactivated enzyme control: Include a sample of heat-denatured DDB_G0286627 to confirm that observed phosphorylation is enzymatically driven.
Substrate specificity controls: Test multiple substrates with varying recognition motifs to establish specificity profiles.
These controls help distinguish between experimental artifacts and genuine biological activities, particularly important when working with "probable" kinases whose activity profiles may not be fully characterized .
Identifying physiological substrates of DDB_G0286627 requires a multi-faceted experimental approach combining in vitro and in vivo techniques. The following methodological workflow is recommended:
In vitro kinase assays with candidate substrates:
Begin with known substrates of related kinases
Perform radiometric assays using [γ-32P]ATP or non-radiometric assays using phospho-specific antibodies
Verify phosphorylation sites by mass spectrometry
Phosphoproteomic approaches:
Compare phosphoproteomic profiles between wild-type and DDB_G0286627-knockout Dictyostelium cells
Use SILAC (Stable Isotope Labeling with Amino acids in Cell culture) for quantitative comparison
Analyze samples under different developmental or stress conditions to capture context-dependent phosphorylation events
Genetic interaction studies:
Generate DDB_G0286627-deficient Dictyostelium strains
Perform phenotypic analyses under various conditions
Conduct synthetic genetic array analysis to identify functional relationships
Protein-protein interaction studies:
Use co-immunoprecipitation followed by mass spectrometry
Conduct yeast two-hybrid screening or proximity-dependent biotin identification (BioID)
Validate interactions with bimolecular fluorescence complementation (BiFC)
This comprehensive approach allows for the systematic identification of physiological substrates while minimizing false positives through multiple layers of validation .
To systematically investigate DDB_G0286627 function during Dictyostelium development, implement the following experimental design:
Temporal expression profiling:
Analyze DDB_G0286627 expression throughout developmental stages using RT-qPCR and Western blotting
Create reporter constructs (e.g., GFP fusion) to visualize expression patterns in vivo
Compare expression profiles under different environmental conditions
Spatial localization studies:
Generate fluorescently tagged DDB_G0286627 constructs
Perform time-lapse confocal microscopy throughout development
Co-localize with known developmental markers
Loss-of-function and gain-of-function studies:
Create knockout strains using CRISPR-Cas9
Develop conditional expression systems for temporal control
Generate phospho-mimetic and phospho-null mutants of key residues
Compare developmental phenotypes under various conditions (starvation, osmotic stress, etc.)
Substrate identification during development:
Perform stage-specific phosphoproteomic analyses
Compare phosphorylation profiles between wild-type and mutant strains
Validate key substrates through in vitro kinase assays
Integration with signaling networks:
Map interactions with known developmental signaling pathways
Perform epistasis experiments with genes in related pathways
Use computational modeling to predict system-level effects
This comprehensive experimental design allows for rigorous assessment of DDB_G0286627 function throughout the complex developmental process of Dictyostelium discoideum .
When faced with contradictory data regarding DDB_G0286627 substrate specificity, employ the following methodological framework to resolve discrepancies:
Systematic evaluation of experimental conditions:
Compare buffer compositions, pH values, and ion concentrations across studies
Assess the influence of different tags (His, GST, etc.) on enzyme activity
Evaluate the impact of protein purity and storage conditions on specificity profiles
Test activity across temperature ranges and incubation times
Comprehensive substrate profiling:
Utilize peptide arrays containing hundreds of potential substrates
Perform parallel reactions with varying ATP concentrations
Compare results from multiple detection methods (radiometric, antibody-based, mass spectrometry)
Develop a consensus sequence motif and validate with mutational analyses
Structural biology approaches:
Obtain crystal structures of DDB_G0286627 alone and in complex with substrates
Perform molecular dynamics simulations to understand substrate binding dynamics
Use hydrogen-deuterium exchange mass spectrometry to identify conformational changes upon substrate binding
Biological context considerations:
Compare in vitro results with in vivo phosphorylation data
Assess the influence of scaffolding proteins and regulatory subunits
Evaluate the impact of post-translational modifications on DDB_G0286627 itself
Meta-analysis and statistical approaches:
Perform quantitative comparisons across studies using standardized metrics
Employ Bayesian statistical methods to integrate contradictory data
Develop predictive models that account for contextual variables
By systematically addressing these aspects, researchers can develop a more nuanced understanding of the apparent contradictions and establish a unified model of DDB_G0286627 substrate specificity .
For robust and reproducible in vitro kinase assays with recombinant DDB_G0286627, the following optimized protocol is recommended:
Buffer composition:
50 mM HEPES or Tris-HCl (pH 7.5)
10 mM MgCl₂ or MnCl₂ (test both separately)
1 mM DTT or 2 mM β-mercaptoethanol
0.1 mg/mL BSA (to prevent non-specific binding)
0.01% Triton X-100 or NP-40 (to prevent aggregation)
Reaction components:
50-200 ng purified recombinant DDB_G0286627
1-5 μg substrate protein or 50-100 μM substrate peptide
100 μM ATP (including trace [γ-³²P]ATP for radiometric detection)
ATP regeneration system (optional): 10 mM creatine phosphate, 1 U/mL creatine kinase
Reaction conditions:
Temperature: 25°C (room temperature) or 30°C
Time: Establish a time course (5, 15, 30, 60 minutes)
Volume: 25-50 μL reaction volume
Termination: Add 5x SDS sample buffer or 100 mM EDTA
Detection methods:
Radiometric: SDS-PAGE followed by autoradiography or phosphorimaging
Non-radiometric: Western blotting with phospho-specific antibodies
Luminescent: ADP-Glo or similar kits that measure ATP consumption
ELISA-based methods for high-throughput applications
Data analysis:
Determine initial velocity conditions (linear phase of reaction)
Calculate kinetic parameters (Km, Vmax) through Michaelis-Menten analysis
Compare relative activities across multiple substrates
Optimize these conditions for your specific experimental questions by performing preliminary experiments that systematically vary key parameters .
To comprehensively analyze DDB_G0286627's role in cell signaling pathways, employ this multi-layered experimental approach:
Pathway perturbation analysis:
Expose cells to pathway-specific stimuli (e.g., cAMP, folate, starvation)
Monitor DDB_G0286627 activation (by phosphorylation state or activity assays)
Compare signaling dynamics in wild-type vs. DDB_G0286627-deficient cells
Use specific pathway inhibitors to determine epistatic relationships
Signaling network mapping:
Perform phosphoproteomic analysis before and after DDB_G0286627 activation
Conduct reverse-phase protein array analysis of key signaling nodes
Use CRISPR-Cas9 screening to identify genetic interactions within pathways
Apply network analysis algorithms to construct signaling diagrams
Live-cell signaling dynamics:
Develop FRET-based biosensors for DDB_G0286627 activity
Perform simultaneous multi-parameter imaging with orthogonal sensors
Analyze single-cell heterogeneity in signaling responses
Track temporal signaling dynamics through development or stress response
Computational modeling:
Develop ordinary differential equation models of pathways involving DDB_G0286627
Parameterize models using experimental data
Perform sensitivity analysis to identify key control points
Use models to predict system-level responses to perturbations
Integration with -omics data:
Correlate signaling activities with transcriptomic changes
Analyze metabolomic consequences of pathway alterations
Examine phenotypic outcomes at cellular and organismal levels
This systematic approach enables researchers to place DDB_G0286627 within the context of broader signaling networks and understand its regulatory impact on downstream cellular processes .
To effectively identify and validate DDB_G0286627 phosphorylation targets in cellular contexts, implement this methodological workflow:
Global phosphoproteomic screening:
Compare phosphoproteomes of wild-type vs. DDB_G0286627-knockout cells
Use stable isotope labeling (SILAC or TMT) for quantitative comparison
Employ TiO₂ or IMAC enrichment for phosphopeptide isolation
Analyze samples by high-resolution LC-MS/MS
Apply stringent statistical criteria for identifying significantly altered phosphosites
Candidate validation strategies:
Generate phospho-specific antibodies against key targets
Develop phospho-null (S/T→A) and phospho-mimetic (S/T→D/E) mutants
Perform rescue experiments in knockout backgrounds
Assess functional consequences of phosphorylation through phenotypic assays
Proximity-based labeling:
Create BioID or TurboID fusions with DDB_G0286627
Identify proximal proteins by streptavidin pulldown and mass spectrometry
Cross-reference with phosphoproteomic data to identify high-confidence targets
Validate physical interactions through co-immunoprecipitation
Substrate specificity profiling:
Use oriented peptide library screening to define consensus motifs
Apply motif information to predict additional substrates in silico
Validate predictions through targeted phosphosite analysis
Compare motif preferences across developmental stages
Spatiotemporal analysis of phosphorylation events:
Employ phospho-specific antibodies for immunofluorescence microscopy
Use FRET-based biosensors to monitor phosphorylation dynamics in real-time
Conduct subcellular fractionation followed by phospho-specific Western blotting
Analyze the timing of phosphorylation events relative to cellular processes
This comprehensive approach enables researchers to build a high-confidence map of DDB_G0286627 phosphorylation targets and understand their functional significance in various cellular contexts .
To comprehensively characterize DDB_G0286627 function in Dictyostelium, implement these phenotypic assays across multiple cellular processes:
Growth and developmental phenotypes:
Measure growth rates in axenic medium and on bacterial lawns
Assess developmental timing through time-lapse imaging
Quantify fruiting body morphology, spore production, and germination efficiency
Evaluate streaming behavior during aggregation phases
Motility and chemotaxis assays:
Perform under-agarose chemotaxis assays toward cAMP and folate
Conduct Dunn chamber or micropipette assays for directional sensing
Track single-cell migration using computer-assisted video microscopy
Analyze parameters such as speed, persistence, and directional accuracy
Stress response characterization:
Test survival under osmotic, oxidative, and mechanical stresses
Assess recovery kinetics after stress exposure
Measure stress-induced gene expression changes
Compare stress granule formation and autophagy induction
Cytoskeletal dynamics:
Visualize F-actin distribution using Lifeact-GFP or phalloidin staining
Monitor focal adhesion dynamics with appropriate markers
Quantify pseudopod formation and retraction cycles
Analyze cytokinesis efficiency and morphology
Quantitative developmental metrics:
Measure cAMP production and pulsing during aggregation
Assess cell-type differentiation ratios (prespore vs. prestalk)
Quantify cell-cell adhesion strength
Evaluate intercellular communication through reporter assays
For all assays, compare DDB_G0286627-knockout, knockdown, or overexpression strains with wild-type controls under identical conditions, using multiple independent clones to ensure reproducibility .
Distinguishing direct from indirect effects of DDB_G0286627 in signaling cascades requires a multi-faceted approach combining temporal, chemical, and genetic strategies:
Temporal resolution strategies:
Implement rapid induction or inhibition systems (e.g., rapamycin-inducible dimerization)
Perform high-resolution time-course experiments with dense sampling
Use computational methods to infer causal relationships from temporal data
Compare response kinetics between direct and downstream effects
Substrate engineering approaches:
Develop analog-sensitive DDB_G0286627 mutants that use bulky ATP analogs
Create substrate mutations at putative phosphorylation sites
Implement "bump-and-hole" strategies for specific inhibition
Use caged kinase or substrate variants for precise temporal control
In vitro validation:
Reconstitute minimal signaling modules with purified components
Perform sequential kinase reactions with intermediate purification steps
Use kinase-dead mutants as negative controls
Implement quantitative biochemical assays for direct phosphorylation
Proximity-based methods:
Use FRET biosensors to detect direct interactions in real-time
Implement split-luciferase complementation assays
Apply crosslinking strategies followed by mass spectrometry
Utilize nanobody-based sensors for activation state detection
Genetic epistasis experiments:
Create combinatorial knockouts of DDB_G0286627 with upstream or downstream factors
Perform rescue experiments with targeted pathway components
Implement orthogonal control systems from different species
Use inducible expression systems to establish dependency relationships
By integrating multiple lines of evidence from these approaches, researchers can build strong cases for direct versus indirect effects of DDB_G0286627 in complex signaling networks .
Advanced computational approaches can significantly enhance the analysis and interpretation of DDB_G0286627 functional data:
Sequence-based predictions and evolutionary analysis:
Perform multiple sequence alignments with characterized kinases
Identify conserved functional domains and regulatory motifs
Construct phylogenetic trees to infer evolutionary relationships
Use conservation patterns to predict functionally important residues
Structural bioinformatics:
Generate homology models based on related kinase structures
Perform molecular dynamics simulations to study conformational flexibility
Use docking studies to predict substrate binding modes
Apply machine learning approaches to identify substrate preferences
Network analysis and pathway reconstruction:
Integrate proteomic, genetic, and functional data into interaction networks
Apply graph theory algorithms to identify signaling modules
Use Bayesian networks to infer causal relationships
Perform enrichment analyses to identify affected biological processes
Dynamic modeling of signaling pathways:
Develop ordinary differential equation models of pathways involving DDB_G0286627
Use parameter estimation techniques to fit models to experimental data
Perform sensitivity analyses to identify critical control points
Simulate the effects of perturbations for experimental design
Image analysis and quantification:
Implement machine learning for automated phenotype classification
Apply computer vision algorithms for tracking cells in time-lapse experiments
Develop custom workflows for quantifying complex morphological features
Use spatial statistics to analyze pattern formation during development
Multi-omics data integration:
Correlate phosphoproteomic data with transcriptomic and metabolomic changes
Apply dimension reduction techniques to visualize complex datasets
Use clustering approaches to identify co-regulated genes and proteins
Develop predictive models of cellular responses based on integrated data
These computational approaches transform raw experimental data into mechanistic insights about DDB_G0286627 function within the broader cellular context .
Research on DDB_G0286627 provides valuable opportunities to elucidate fundamental principles of cellular signaling through several research angles:
Evolutionary conservation of kinase signaling networks:
Compare DDB_G0286627 with homologous kinases across species
Identify conserved substrate recognition patterns
Trace the evolution of regulatory mechanisms
Determine how signaling modules are repurposed during evolution
Spatiotemporal regulation of kinase activity:
Investigate subcellular localization dynamics during signaling events
Examine how scaffolding proteins and microdomains affect signaling specificity
Study temporal patterns of activation and deactivation
Analyze the coordination of multiple signaling pathways during complex cellular processes
Systems-level properties of kinase networks:
Explore functional redundancy and compensation mechanisms
Study signal amplification and attenuation dynamics
Investigate signal integration from multiple inputs
Analyze feedback and feedforward loops in pathway regulation
Quantitative aspects of phosphorylation signaling:
Determine how phosphorylation stoichiometry affects downstream responses
Investigate ultrasensitivity and threshold effects in signaling cascades
Study how multisite phosphorylation creates complex response patterns
Analyze the kinetics of signal propagation through pathways
Developmental context of signaling pathways:
Examine how signaling networks are rewired during development
Study the coordination of individual cell signaling with multicellular patterns
Investigate how environmental cues are translated into developmental decisions
Analyze the integration of multiple signaling pathways during morphogenesis
By addressing these fundamental questions through research on DDB_G0286627, investigators can contribute broadly to our understanding of cellular signaling principles that apply across diverse biological systems .
Investigating DDB_G0286627's role in developmental signaling presents several methodological challenges that require specialized approaches:
Temporal complexity challenges:
Challenge: Capturing rapid signaling events during key developmental transitions
Solution: Implement high-temporal resolution approaches such as optogenetic control systems, synchronized development protocols, and automated time-lapse imaging with computational analysis
Spatial heterogeneity issues:
Challenge: Distinguishing cell-type specific signaling patterns in multicellular structures
Solution: Apply single-cell phosphoproteomics, cell-type specific reporters, spatial transcriptomics, and advanced imaging techniques like light-sheet microscopy with computational image analysis
Multifactorial signaling integration:
Challenge: Determining how DDB_G0286627 integrates with other signaling pathways
Solution: Perform combinatorial perturbations, develop multi-parameter biosensors, and apply systems biology modeling approaches to capture pathway crosstalk
Technical limitations in manipulation:
Challenge: Achieving precise temporal control of DDB_G0286627 activity during development
Solution: Develop stage-specific inducible systems, apply chemical genetics approaches with analog-sensitive kinase mutants, and implement CRISPR-based methods for rapid genetic manipulation
Distinguishing primary and secondary effects:
Challenge: Separating direct consequences of DDB_G0286627 activity from downstream events
Solution: Perform acute inhibition studies, use phosphoproteomics with high temporal resolution, and implement mathematical modeling to infer causal relationships
Quantitative assessment of developmental phenotypes:
Challenge: Objectively measuring complex developmental outcomes
Solution: Develop computational image analysis pipelines, implement machine learning for phenotype classification, and establish standardized quantitative metrics for developmental progression
Addressing these methodological challenges requires interdisciplinary approaches combining advanced genetic tools, sophisticated imaging techniques, biochemical assays, and computational methods to fully elucidate DDB_G0286627's role in developmental signaling .
To comprehensively identify and validate substrates and interacting partners of DDB_G0286627, implement this systematic experimental workflow:
Initial discovery phase:
Affinity purification-mass spectrometry (AP-MS):
Express tagged DDB_G0286627 in Dictyostelium cells
Perform immunoprecipitation under various conditions (e.g., different developmental stages)
Identify co-purified proteins by mass spectrometry
Use appropriate controls (tag-only, kinase-dead mutants)
Proximity labeling approaches:
Generate BioID or TurboID fusions with DDB_G0286627
Induce biotinylation in living cells during relevant biological processes
Purify biotinylated proteins and identify by mass spectrometry
Compare spatial interactomes across cellular compartments
Phosphoproteomic screening:
Compare phosphoproteomes between wild-type and DDB_G0286627-deficient cells
Analyze phosphorylation changes upon acute activation/inhibition
Enrich for phosphopeptides using TiO₂ or IMAC techniques
Apply quantitative proteomics (SILAC, TMT) for accurate comparisons
Candidate validation phase:
In vitro kinase assays:
Express and purify candidate substrates
Perform kinase reactions with recombinant DDB_G0286627
Map phosphorylation sites by mass spectrometry
Generate phospho-specific antibodies for key sites
Cellular validation:
Create phospho-null and phospho-mimetic mutants of candidate substrates
Assess functional consequences through phenotypic assays
Perform co-localization studies during relevant processes
Use FRET-based approaches to detect direct interactions in living cells
Functional characterization phase:
Genetic interaction studies:
Create double knockouts of DDB_G0286627 and interacting partners
Perform phenotypic analyses under various conditions
Implement genetic rescue experiments with modified variants
Apply synthetic genetic array approaches for systematic analysis
Pathway mapping:
Position interactions within known signaling cascades
Determine epistatic relationships through sequential perturbations
Assess effects on downstream cellular processes
Develop computational models incorporating new interactions
Advanced structural and dynamic analyses:
Structural biology approaches:
Obtain structures of DDB_G0286627 in complex with interacting partners
Use hydrogen-deuterium exchange mass spectrometry to map interaction surfaces
Perform molecular dynamics simulations to understand binding dynamics
Create domain deletion constructs to map minimal interaction regions
Live-cell dynamics:
Track spatiotemporal dynamics of interactions during relevant processes
Implement optogenetic approaches to manipulate interactions with temporal precision
Correlate interaction dynamics with cellular behaviors
Analyze interaction stoichiometry using fluorescence fluctuation spectroscopy
This comprehensive approach enables researchers to build a validated network of DDB_G0286627 substrates and interacting partners with functional significance .