PIP4K2C (Phosphatidylinositol-5-Phosphate 4-Kinase Type-2 Gamma) antibodies are laboratory tools designed to detect and study the PIP4K2C protein, a lipid kinase involved in phosphoinositide metabolism and immune regulation. PIP4K2C converts phosphatidylinositol-5-phosphate (PI5P) to phosphatidylinositol-4,5-bisphosphate (PI4,5P2), a critical lipid signaling molecule that modulates cellular processes like vesicular trafficking, autophagy, and mTORC1 signaling . These antibodies are essential for elucidating PIP4K2C’s role in autoimmune diseases, cancer, and viral infections .
PIP4K2C antibodies are validated for diverse experimental applications:
Immunohistochemistry (IHC): Visualizes protein localization in tissues like kidney, brain, and immune organs .
Immunofluorescence (IF): Maps subcellular distribution in fixed cells .
Flow cytometry: Analyzes PIP4K2C expression in immune cell populations .
Immunoprecipitation (IP): Isolates PIP4K2C complexes for interaction studies .
Recent studies using PIP4K2C antibodies revealed its role in:
Autoimmunity: Germline Pip4k2c knockout mice exhibit hyperactivated mTORC1 signaling, T-cell dysregulation, and tissue inflammation .
Cancer: PIP4K2C loss in melanoma and colorectal cancer cells promotes liver metastasis via insulin/PI3K/AKT pathway hyperactivation .
Viral infections: PIP4K2C binds SARS-CoV-2 NSP6, impairing autophagic flux; its inhibition reduces viral replication .
Mechanism: PIP4K2C suppresses mTORC1 signaling, which governs T-cell differentiation. Loss of PIP4K2C increases Th1/Th17 cells and reduces regulatory T-cells (Tregs), driving autoimmunity .
Therapeutic relevance: Rapamycin (mTORC1 inhibitor) reverses inflammatory phenotypes in Pip4k2c−/− mice .
Organotropism: Pip4k2c-deficient cancer cells exploit insulin-rich liver microenvironments for metastasis via PI3K/AKT hyperactivation .
Clinical correlation: Human liver metastases show reduced PIP4K2C expression compared to primary tumors .
SARS-CoV-2: PIP4K2C inhibitors (e.g., RMC-113) restore autophagic flux disrupted by viral NSP6, reducing replication .
PIP4K2C is emerging as a druggable target for:
Cancer immunotherapy: Degraders like LRK-A enhance tumor antigen presentation and reduce colorectal cancer growth in preclinical models .
Antiviral therapy: Dual PIP4K2C/PIKfyve inhibitors (e.g., RMC-113) show broad-spectrum antiviral activity .
Autoimmune diseases: Modulating PIP4K2C-mTORC1 axis could restore immune homeostasis .
PIP4K2C is a lipid kinase that belongs to the PI-5-phosphate 4-kinase family, which converts PI5P to PI45P2. Beyond this catalytic function, PIP4K2C plays critical roles in membrane trafficking, vesicular dynamics, autophagy regulation, and immune system modulation . Unlike its family members PIP4K2A and PIP4K2B, PIP4K2C has unique functions in regulating immune responses, particularly in cancer contexts . PIP4K2C also governs multiple aspects of membrane trafficking through its ability to regulate PI45P2 membrane localization and clustering, activities that appear to be independent of its catalytic function .
At the molecular level, PIP4K2C's calculated molecular weight is 47 kDa (421 amino acids) . Recent studies have revealed its crucial involvement in viral infections, including SARS-CoV-2, where it participates in viral entry, RNA replication, and assembly/egress processes .
Selection should be based on:
Target epitope specificity: Consider whether you need antibodies targeting specific domains (e.g., C-terminal region, amino acids 310-381 or 348-364)
Host species compatibility: Ensure the antibody won't cross-react with other proteins in your experimental system. Available options include mouse monoclonal and rabbit polyclonal antibodies
Application requirements: Different antibodies are validated for specific applications:
Reactivity spectrum: Confirm the antibody reacts with your species of interest (human, mouse, rat)
For multiplexed assays requiring paired antibodies, consider validated pairs like those offered for cytometric bead arrays (MP50878-2 or MP50878-3) .
PIP4K2C antibodies can be employed for:
Protein detection and quantification:
Localization studies:
Protein interaction studies:
Multiplex analysis:
Validation of knockdown efficiency:
Methodological approach for optimization:
Initial concentration titration:
For Western blotting: Begin with 1:500-1:2000 dilution ranges
For ELISA: Start with 1-5 μg/mL and adjust based on signal-to-background ratio
For immunohistochemistry: Test 1:100-1:500 dilutions
Blocking optimization:
Test different blocking agents (BSA, milk, commercial blockers)
Optimize blocking time (1-3 hours)
Consider using specialized blockers for phosphoprotein detection
Sample preparation considerations:
For membrane proteins: Use appropriate lysis buffers that maintain membrane protein integrity
Include phosphatase inhibitors when studying phosphorylation states
Consider using intact cells for surface epitopes and permeabilized cells for intracellular domains
Validation controls:
Storage and handling:
PIP4K2 family members (PIP4K2A, PIP4K2B, and PIP4K2C) share over 60% similarity but exhibit differences in enzymatic activity levels and subcellular distribution . To distinguish them:
Antibody selection strategy:
Functional discrimination:
Expression pattern analysis:
Compare tissue-specific expression profiles
Examine subcellular localization differences using fluorescently-tagged constructs
Analyze relative expression levels across different cell types
Knockout/knockdown validation:
Recent research has uncovered PIP4K2C's involvement in SARS-CoV-2 infection . Key methodological approaches include:
Target engagement and inhibition studies:
Functional assessment of viral life cycle stages:
Mechanistic investigations:
Autophagic flux measurements:
LC3-I to LC3-II conversion assessment by Western blotting
Autophagy flux reporter systems (GFP-LC3-RFP)
Transmission electron microscopy for autophagosome visualization
Degradation of selective autophagy substrates
Model systems:
PIP4K2C has been implicated in autophagy regulation, with its inhibition or knockdown affecting autophagic flux . Methodological applications include:
Monitoring autophagy marker changes:
Co-localization studies:
Double immunofluorescence for PIP4K2C and autophagosome markers
Live-cell imaging of fluorescently-tagged PIP4K2C with autophagy proteins
Super-resolution microscopy to visualize membrane interactions
Functional autophagy assays:
Long-lived protein degradation assays following PIP4K2C knockdown
Mitophagy-specific measurements using MitoTracker-based approaches
Selective substrate degradation monitoring
Signaling pathway analysis:
Genetic rescue experiments:
Complementation with wild-type vs. catalytically inactive PIP4K2C
Domain-specific mutants to identify regions critical for autophagy regulation
Chimeric constructs with other PIP4K2 family members
Given PIP4K2C's role in immune regulation and the link between PIP4K2C SNP (rs1678542) and autoimmunity , several methodological approaches are valuable:
Genetic association studies:
Genotyping for rs1678542 in patients with autoimmune conditions
Correlation analysis between genotype and PIP4K2C expression levels
eQTL (expression quantitative trait loci) analysis
Immune cell phenotyping:
Signaling pathway investigation:
In vivo models:
Therapeutic potential assessment:
Methodological approaches for antibody validation:
Knockout/knockdown controls:
Peptide competition assays:
Pre-incubate antibody with excess immunizing peptide
Parallel processing of competed and non-competed samples
Expect signal elimination in competed samples for specific antibodies
Multiple antibody approach:
Recombinant protein testing:
Mass spectrometry validation:
Immunoprecipitate with the antibody and perform mass spectrometry analysis
Confirm target identification and assess potential cross-reactivities
Common challenges and their solutions:
Cross-reactivity with related kinases:
Use antibodies targeting unique regions not conserved across family members
Include family member knockouts as controls
Perform careful titration to minimize non-specific binding
Detecting low expression levels:
Implement signal amplification strategies (HRP-conjugated secondary antibodies)
Consider immunoprecipitation before Western blotting
Use high-sensitivity detection systems (chemiluminescent substrates)
Phosphorylation-dependent epitope masking:
Test antibody performance with and without phosphatase treatment
Use multiple antibodies targeting different regions
Consider phospho-specific antibodies if phosphorylation status is relevant
Post-translational modification interference:
Optimize sample preparation to preserve or remove modifications as needed
Use denaturation conditions that expose relevant epitopes
Consider modification-specific antibodies for comprehensive analysis
Technical issues in application-specific contexts:
For immunohistochemistry: Optimize antigen retrieval methods
For flow cytometry: Careful fixation and permeabilization protocol selection
For ELISA: Thorough blocking and washing optimization
Methodological considerations for high-throughput applications:
Assay miniaturization and automation:
Adapt protocols to 384- or 1536-well formats
Optimize reagent concentrations for minimal volumes
Implement automated liquid handling systems
Readout optimization:
Select high signal-to-background ratio detection methods
Consider homogeneous (no-wash) assay formats
Implement multiplex readouts to increase information content
ELISA-based screening approaches:
Cell-based assay considerations:
Select appropriate cell models (endogenous vs. overexpression)
Develop stable reporter cell lines
Optimize fixation and antibody incubation times
Target engagement confirmation:
Antibody applications in therapeutic development:
Target validation studies:
Inhibitor screening and characterization:
Degrader development support:
Therapeutic response biomarkers:
Developing assays to monitor PIP4K2C levels in clinical samples
Correlation of target engagement with efficacy measures
Patient stratification based on expression levels
Combination therapy strategies:
Recent studies suggest PIP4K2C inhibition could enhance cancer immunotherapy . Key methodological approaches include:
Immune cell functional assays:
T cell activation studies following PIP4K2C inhibition
Dendritic cell antigen processing and presentation assessment
NK cell cytotoxicity measurements with and without PIP4K2C modulation
Tumor microenvironment analysis:
Multiplex immunohistochemistry to assess immune infiltration
Single-cell RNA sequencing of tumor and immune compartments
Spatial transcriptomics to map PIP4K2C expression patterns
In vivo model systems:
Mechanism delineation approaches:
Translational considerations:
Studies have linked PIP4K2C to mTORC1 signaling regulation . Methodological approaches include:
Biochemical pathway analysis:
Phosphorylation status of mTORC1 substrates (S6K1, 4E-BP1) following PIP4K2C modulation
Co-immunoprecipitation studies to identify physical interactions
In vitro kinase assays to assess direct effects
Genetic epistasis experiments:
Combined knockdown of PIP4K2C and mTOR pathway components
Rescue experiments with constitutively active pathway members
CRISPR screens to identify synthetic interactions
Cellular assays:
Amino acid sensing studies, as mTORC1 is nutrient-responsive
Lysosomal positioning and recruitment of mTORC1 components
Autophagy induction measurements under various nutrient conditions
Systems biology approaches:
Phosphoproteomic analysis following PIP4K2C inhibition
Network modeling of signaling pathway interactions
Integration with transcriptomic and metabolomic data
Model organism studies:
Several antibody pairs have been validated for multiplex applications . Key methodological approaches:
Antibody pair selection:
Conjugation strategies:
Assay development considerations:
Optimize antibody concentrations individually before multiplexing
Validate specificity in the multiplex context with appropriate controls
Develop calibration curves for each target in the multiplex format
Technical platforms:
Data analysis approaches:
Implement compensation algorithms for spectral overlap
Develop appropriate normalization strategies
Apply machine learning for pattern recognition in complex datasets
Methodological considerations for imaging applications:
Super-resolution microscopy:
Select bright, photostable fluorophores for techniques like STORM or PALM
Consider direct conjugation to minimize distance between fluorophore and target
Optimize sample preparation to reduce background autofluorescence
Live-cell imaging approaches:
Develop membrane-permeable nanobody derivatives
Consider genetically encoded tags with complementary fluorophores
Optimize imaging conditions to minimize phototoxicity
Multiplex imaging strategies:
Sequential immunofluorescence with antibody stripping
Spectral unmixing for simultaneous detection of multiple targets
Mass cytometry imaging for highly multiplexed analyses
Correlation with functional readouts:
Combine with activity sensors for phosphoinositides
Integrate with autophagic flux reporters
Correlate with calcium or pH indicators for functional contexts
Tissue and 3D model applications:
Optimize clearing methods for thick specimens
Adapt antibody penetration protocols for organoids
Implement light-sheet microscopy for rapid 3D imaging
This comprehensive FAQ collection provides researchers with both fundamental and advanced methodological guidance for working with PIP4K2C antibodies across various research contexts. The increasing recognition of PIP4K2C's roles in viral infection, cancer immunotherapy, and autoimmunity makes these applications particularly relevant to current research priorities.