PIP5K1A Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
Storage Buffer: PBS supplemented with 0.02% sodium azide, 50% glycerol, pH 7.3. Store at -20°C. Avoid repeated freeze-thaw cycles.
Lead Time
Product shipment typically occurs within 1-3 business days of order receipt. Delivery times may vary depending on shipping method and destination. Please contact your local distributor for precise delivery estimates.
Synonyms
68 kDa type I phosphatidylinositol-4-phosphate 5-kinase alpha antibody; Phosphatidylinositol 4 phosphate 5 kinase type I alpha antibody; Phosphatidylinositol-4-phosphate 5-kinase type I alpha antibody; Phosphatidylinositol-4-phosphate 5-kinase type-1 alpha antibody; PI4P5K Ia antibody; PI51A_HUMAN antibody; PIP5K A antibody; PIP5K1-alpha antibody; Pip5k1a antibody; PIP5KIalpha antibody; PtdIns(4)P-5-kinase 1 alpha antibody
Target Names
Uniprot No.

Target Background

Function

PIP5K1A (Phosphatidylinositol-4-phosphate 5-kinase type I alpha) catalyzes the phosphorylation of phosphatidylinositol 4-phosphate (PI4P) to produce phosphatidylinositol 4,5-bisphosphate (PIP2). PIP2 is a crucial lipid second messenger involved in numerous cellular processes, including signal transduction, vesicular trafficking, actin cytoskeleton dynamics, cell adhesion, and cell motility. It can function directly as a second messenger or serve as a precursor for other second messengers such as inositol 1,4,5-trisphosphate (IP3), diacylglycerol (DAG), and phosphatidylinositol-3,4,5-trisphosphate (PIP3). PIP5K1A-mediated phosphorylation of PI4P is the primary pathway for PIP2 biosynthesis. This enzyme can also utilize phosphatidylinositol (PtdIns) as a substrate *in vitro*. In conjunction with PIP5K1C, PIP5K1A is essential for phagocytosis, with each enzyme regulating distinct aspects of actin remodeling during sequential steps. Specifically, PIP5K1A promotes particle ingestion by activating the WASP (Wiskott-Aldrich syndrome protein) GTPase-binding protein, thereby triggering Arp2/3-dependent actin polymerization at the nascent phagocytic cup. Furthermore, PIP5K1A, along with PIP5K1B, is required for the G-protein coupled receptor-stimulated synthesis of IP3, which subsequently facilitates stable platelet adhesion. PIP5K1A is recruited to the plasma membrane by the E-cadherin/β-catenin complex, where it provides the PIP2 substrate for the generation of PtdIns(3,4,5)P3, IP3, and DAG, ultimately mobilizing intracellular calcium and driving keratinocyte differentiation. In adipocytes, it positively regulates insulin-induced translocation of GLUT4 (SLC2A4) to the cell membrane. PIP5K1A, in collaboration with PIP5K1C, plays a role during embryogenesis. Independent of its catalytic activity, PIP5K1A is crucial for membrane ruffling formation, actin organization, and focal adhesion formation during directed cell migration by controlling integrin-induced translocation of the small GTPase RAC1 to the plasma membrane. Additionally, PIP5K1A functions within the nucleus, where it activates TUT1 (TUTase 1) adenylyltransferase activity in nuclear speckles, thus regulating the polyadenylation of specific mRNAs.

Gene References Into Functions

Selected Research Highlights:

  • Mediates oncogenic KRAS signaling and proliferation. (PMID: 30194290)
  • Inhibition of IQGAP1 interaction with PIPKIα or PI(3)K diminishes PtdIns(3,4,5)P3 generation, signaling, and cancer cell survival. (PMID: 27870828)
  • PP1 (Protein Phosphatase 1) activation of PIP5K1A is required for PLC-γ1 activation and keratinocyte differentiation. (PMID: 27340655)
  • Modified by polySUMO-2 specifically during apoptosis. (PMID: 23994136)
  • Regulated by CD28 in human primary CD4+ T lymphocytes. (PMID: 23589613)
  • Essential for cell migration through spatial regulation of Rac1 activity. (PMID: 20660631)
  • Required for invadopodia formation in breast cancer cells. (PMID: 20426790)
  • Membrane ruffling requires coordination with Rac signaling. (PMID: 12682053)
  • Influences cell migration through regulation of PI(4,5)P2 synthesis via direct activation of PIPKIα. (PMID: 15870270)
  • Downregulated during apoptosis. (PMID: 16979564)
  • Responsible for PIP2 synthesis in natural killer cells. (PMID: 18073347)
  • Regulates receptor internalization through β-arrestin-mediated localization. (PMID: 18534983)
  • Crucial for HIV-1 entry and early infection steps in lymphocytes. (PMID: 18981107)
  • Recruited to the E-cadherin-catenin complex by extracellular calcium. (PMID: 19158393)
  • Involved in FcγRIIA-mediated signaling platform formation. (PMID: 19331818)
Database Links

HGNC: 8994

OMIM: 603275

KEGG: hsa:8394

STRING: 9606.ENSP00000357883

UniGene: Hs.655131

Subcellular Location
Cell membrane. Cytoplasm. Nucleus. Nucleus speckle. Cell projection, ruffle. Cell projection, lamellipodium.
Tissue Specificity
Highly expressed in heart, placenta, skeletal muscle, kidney and pancreas. Detected at lower levels in brain, lung and liver.

Q&A

What is PIP5K1A and why is it important in cellular research?

PIP5K1A (also known as 68 kDa type I phosphatidylinositol 4-phosphate 5-kinase alpha) is an enzyme that catalyzes the phosphorylation of phosphatidylinositol 4-phosphate (PtdIns(4)P/PI4P) to form phosphatidylinositol 4,5-bisphosphate (PtdIns(4,5)P2/PIP2). This 62.6 kilodalton protein plays a central role in multiple cellular processes including signal transduction, vesicle trafficking, actin cytoskeleton dynamics, cell adhesion, and cell motility . PIP5K1A-mediated phosphorylation represents the predominant pathway for PtdIns(4,5)P2 synthesis, which can directly act as a second messenger or serve as a precursor for generating other important signaling molecules such as inositol 1,4,5-trisphosphate (IP3), diacylglycerol (DAG), or phosphatidylinositol-3,4,5-trisphosphate (PtdIns(3,4,5)P3/PIP3) . Its involvement in multiple signaling cascades makes PIP5K1A antibodies invaluable tools for studying these fundamental cellular processes and their dysregulation in disease states.

What applications are PIP5K1A antibodies most commonly used for?

PIP5K1A antibodies are predominantly employed in several key laboratory techniques. Western Blotting (WB) serves as the primary application for detecting and quantifying PIP5K1A protein expression in cell or tissue lysates . Immunohistochemistry (IHC) allows researchers to visualize PIP5K1A distribution in tissue sections, providing spatial information about expression patterns . Immunocytochemistry and Immunofluorescence (ICC/IF) techniques enable subcellular localization studies to determine where PIP5K1A functions within cells . Additionally, co-immunoprecipitation (Co-IP) assays have been successfully employed to study PIP5K1A's interactions with binding partners, particularly in examining its specific interaction with KRAS . Each application requires careful optimization of antibody concentration, incubation conditions, and detection methods to ensure specific and sensitive detection of PIP5K1A.

How do I select an appropriate PIP5K1A antibody for my experiment?

Selection of an appropriate PIP5K1A antibody requires consideration of several key factors. First, define your experimental application (WB, IHC, ICC/IF) as different antibodies may perform optimally in specific applications . Consider target species reactivity, ensuring the antibody recognizes PIP5K1A in your experimental model (human, mouse, rat, etc.) . Evaluate antibody format (polyclonal vs. monoclonal) based on experimental needs - polyclonals often provide higher sensitivity by recognizing multiple epitopes, while monoclonals offer greater specificity and consistency . Review the immunogen information to determine which region of PIP5K1A the antibody targets (N-terminal, C-terminal, or internal domains), which may affect detection of specific isoforms or splice variants . Finally, assess validation data provided by manufacturers, including Western blot images, IHC staining patterns, and positive/negative controls to ensure the antibody performs as expected in your planned application .

What controls should I include when using PIP5K1A antibodies?

Proper controls are essential for validating PIP5K1A antibody results. Include a positive control sample known to express PIP5K1A (specific cell lines or tissues with documented expression) . For negative controls, use samples where PIP5K1A expression is absent or samples from PIP5K1A knockout models if available. Loading controls (housekeeping proteins like GAPDH, β-actin, or tubulin) are crucial for Western blotting to normalize protein loading and enable accurate quantification . Include isotype controls (non-specific antibodies of the same isotype) for immunostaining applications to distinguish specific binding from background signal . Peptide competition assays, where the antibody is pre-incubated with the immunizing peptide before application, can confirm binding specificity . For advanced validation, consider using siRNA/shRNA knockdown or CRISPR-Cas9 knockout samples to demonstrate antibody specificity for PIP5K1A .

How can I optimize PIP5K1A antibody performance in proximity-dependent labeling experiments?

Proximity-dependent labeling techniques, such as BirA-based proximity labeling, have proven valuable for studying PIP5K1A interactions, particularly with KRAS . For optimal performance, begin by validating your BirA-PIP5K1A fusion construct to ensure it retains both biotinylation activity and the native functions of PIP5K1A . When designing your construct, carefully consider the fusion orientation (N- or C-terminal tagging) as this can impact PIP5K1A's enzymatic activity or protein interactions. Optimize biotin concentration and labeling time through titration experiments to achieve sufficient labeling while minimizing background. Always include appropriate controls, including BirA-only expressing cells and catalytically inactive BirA mutants to distinguish true interactors from nonspecific labeling . For analysis, employ stringent washing conditions and consider using quantitative proteomics with stable isotope labeling to identify enriched proteins with statistical confidence. The proximity labeling approach offers distinct advantages over traditional co-immunoprecipitation by capturing weak or transient interactions in living cells, as demonstrated in studies identifying PIP5K1A as a KRAS-specific interactor .

What are the methodological considerations for using PIP5K1A antibodies in tumor tissue microarrays?

Using PIP5K1A antibodies in tumor tissue microarrays (TMAs) requires careful methodological planning. Begin with antigen retrieval optimization, testing multiple methods (heat-induced epitope retrieval with citrate or EDTA buffers at varying pH) to determine optimal conditions for PIP5K1A detection . Antibody titration is essential - test multiple concentrations to identify the optimal dilution that maximizes specific signal while minimizing background. Include positive control tissues known to express PIP5K1A and negative controls where primary antibody is omitted . For quantitative analysis, establish clear scoring criteria (intensity, percentage of positive cells, subcellular localization) and ensure multiple independent observers score the samples to reduce bias. Consider multiplexed immunostaining to simultaneously visualize PIP5K1A with other markers (such as KRAS or phospho-ERK) to assess pathway activation . Validate TMA findings with orthogonal methods such as Western blotting or qRT-PCR from matched samples when possible. Given PIP5K1A's emerging role in KRAS-driven cancers, correlating its expression patterns with clinical data may provide valuable prognostic insights for patients with KRAS mutations .

How can I detect specific PIP5K1A-protein interactions using antibody-based approaches?

Detecting PIP5K1A-protein interactions requires specialized antibody-based approaches. Co-immunoprecipitation (Co-IP) serves as the gold standard, where PIP5K1A antibodies immobilized on beads can capture PIP5K1A along with its binding partners for subsequent identification . For optimal results, use mild lysis buffers that preserve protein-protein interactions and optimize antibody concentration and incubation conditions. Proximity ligation assay (PLA) offers an alternative approach, using two primary antibodies (one against PIP5K1A, another against the suspected interaction partner) followed by species-specific secondary antibodies linked to DNA oligonucleotides that, when in close proximity, generate fluorescent signals visualizable by microscopy . For studying KRAS-PIP5K1A interactions specifically, reciprocal Co-IP experiments have confirmed this association, with PIP5K1A preferentially co-immunoprecipitating with KRAS G12V compared to NRAS G12V or HRAS G12V . Bimolecular fluorescence complementation (BiFC) and Förster resonance energy transfer (FRET) provide alternative approaches for visualizing PIP5K1A interactions in living cells, offering spatial and temporal resolution that traditional Co-IP cannot achieve.

What methods can be used to assess PIP5K1A enzymatic activity in conjunction with antibody detection?

Assessing PIP5K1A enzymatic activity in conjunction with antibody detection requires specialized approaches. In vitro kinase assays using immunoprecipitated PIP5K1A can measure the conversion of PI4P to PI(4,5)P2 through radiolabeled ATP incorporation or detection of the phosphorylated product using specific antibodies against PI(4,5)P2 . For cell-based assays, combine PIP5K1A immunodetection with PI(4,5)P2 staining using specific PI(4,5)P2 antibodies or fluorescent protein-tagged lipid-binding domains (such as PH domains) to correlate PIP5K1A localization with its lipid products . Lipid extraction followed by thin-layer chromatography or mass spectrometry enables quantitative analysis of phosphoinositide levels in cells with altered PIP5K1A expression. Following genetic manipulation of PIP5K1A (overexpression, knockdown, or mutation), researchers should monitor downstream signaling events by immunoblotting for phosphorylated effector proteins . When designing these experiments, include appropriate controls such as catalytically inactive PIP5K1A mutants and specific inhibitors of phosphoinositide metabolism to confirm that observed effects are specifically attributable to PIP5K1A activity.

How does PIP5K1A contribute to KRAS-driven oncogenesis and what antibody-based methods best detect this interaction?

PIP5K1A has been identified as a KRAS-specific interactor that contributes significantly to KRAS-driven oncogenesis through several mechanisms . PIP5K1A specifically binds to a unique region in KRAS (but not NRAS or HRAS), suggesting isoform-specific functions in RAS signaling . This interaction promotes KRAS-mediated signaling, as PIP5K1A depletion specifically reduces oncogenic KRAS signaling and proliferation in cancer cell lines . To detect this interaction, BirA proximity-dependent biotin identification has proven particularly effective, capturing weak or transient interactions in living cells that might be missed by traditional co-immunoprecipitation methods . Validation of this interaction requires complementary approaches: co-immunoprecipitation experiments in 293T cells expressing tagged versions of both proteins have confirmed that PIP5K1A preferentially binds to KRAS G12V compared to other RAS isoforms . Functional validation using CRISPR-Cas9 genetic screens demonstrated that sgRNAs targeting PIP5K1A were the most negatively enriched in KRAS G12V-transformed cells, indicating that PIP5K1A is essential for KRAS-driven proliferation . These findings suggest PIP5K1A could be a potential therapeutic target specifically for KRAS-mutant cancers, which have historically been difficult to treat.

What is the significance of PIP5K1A in phosphoinositide signaling, and how can antibodies help map these pathways?

PIP5K1A plays a central role in phosphoinositide signaling by catalyzing the production of PtdIns(4,5)P2, a critical lipid second messenger and precursor for other signaling molecules . This enzyme represents the predominant pathway for PtdIns(4,5)P2 synthesis, which regulates numerous cellular processes including signal transduction, vesicle trafficking, actin cytoskeleton dynamics, cell adhesion, and motility . PIP5K1A-generated PtdIns(4,5)P2 serves as a substrate for phospholipase C to produce inositol 1,4,5-trisphosphate (IP3) and diacylglycerol (DAG), or for PI3K to generate phosphatidylinositol-3,4,5-trisphosphate (PIP3) . Antibody-based approaches are invaluable for mapping these signaling networks through multiplexed immunofluorescence to simultaneously visualize PIP5K1A with its upstream regulators or downstream effectors . Phospho-specific antibodies against signaling molecules activated downstream of PIP5K1A enable tracking of pathway activation in response to stimuli or perturbations . Proximity ligation assays can detect PIP5K1A's interactions with other signaling components in situ with subcellular resolution. Combined with lipid detection methods, these antibody-based approaches provide a comprehensive view of how PIP5K1A coordinates phosphoinositide signaling in normal physiology and disease states .

How can PIP5K1A antibodies be used to investigate its role in cell migration and invasion?

PIP5K1A antibodies offer multiple methodological approaches for investigating its role in cell migration and invasion. Immunofluorescence staining can reveal PIP5K1A's subcellular localization during migration, particularly its enrichment at leading edges where actin remodeling and membrane protrusion occur . Time-course experiments examining PIP5K1A expression and localization during epithelial-to-mesenchymal transition can elucidate its role in this cancer-relevant process. For functional studies, combine PIP5K1A knockdown or overexpression with migration assays (wound healing, Boyden chamber, or live-cell tracking), followed by immunoblotting to confirm manipulation of PIP5K1A levels . Co-immunostaining of PIP5K1A with focal adhesion markers, actin cytoskeleton components, or phosphoinositide-binding proteins can reveal mechanistic connections between PIP5K1A activity and migratory machinery . In 3D invasion models, confocal microscopy with PIP5K1A antibodies can visualize its distribution in invadopodia or other invasive structures. Phospho-specific antibodies against proteins regulated by PIP5K1A-generated PIP2 (such as components of the WASP-Arp2/3 pathway) can connect PIP5K1A activity to specific cytoskeletal remodeling events that drive migration and invasion .

What is known about PIP5K1A expression in different cancer types and how can immunohistochemistry quantify this?

Research using PIP5K1A antibodies for immunohistochemistry has revealed variable expression patterns across cancer types, though comprehensive data remains limited. PIP5K1A expression appears particularly relevant in KRAS-mutant cancers, including pancreatic, colorectal, and lung adenocarcinomas, where it may contribute to oncogenic signaling . To quantify PIP5K1A expression in tumor samples, implement a systematic immunohistochemistry scoring approach. First, optimize antigen retrieval and antibody dilution using positive control tissues (such as normal tissues known to express PIP5K1A) . Develop a scoring system that accounts for both staining intensity (0=negative, 1=weak, 2=moderate, 3=strong) and percentage of positive cells to calculate an H-score (0-300) or quickscore . For more objective quantification, employ digital pathology with image analysis software to measure staining intensity and distribution . Validate scoring consistency with multiple independent observers and correlate findings with other markers in the same pathway (such as KRAS mutation status or phospho-ERK levels) . In advanced studies, multiplex immunohistochemistry can simultaneously visualize PIP5K1A with other biomarkers to better characterize tumor heterogeneity and signaling pathway activation .

How should experiments be designed to compare different PIP5K1A antibodies for specific applications?

Designing rigorous comparison experiments for PIP5K1A antibodies requires systematic methodology. Begin by selecting antibodies targeting different epitopes of PIP5K1A (N-terminal, C-terminal, internal domains) from multiple manufacturers . Prepare a panel of validation samples including positive controls (cell lines or tissues with known PIP5K1A expression), negative controls (samples with low or no expression), and when possible, PIP5K1A knockout or knockdown samples . For Western blotting comparisons, run identical amounts of the same lysates in parallel, process membranes simultaneously with different antibodies at their respective optimal dilutions, and evaluate based on band specificity, signal-to-noise ratio, and detection of expected molecular weight (approximately 62.6 kDa) . For immunostaining applications, process identical tissue or cell sections in parallel with each antibody, maintaining consistent protocols except for antibody-specific optimization steps . Document all comparison data in a standardized table format recording sensitivity (minimum detectable amount), specificity (absence of non-specific bands/staining), reproducibility across replicates, and performance across applications. This systematic comparison approach ensures selection of the most appropriate antibody for each specific experimental context and application.

What strategies can address cross-reactivity and non-specific binding when using PIP5K1A antibodies?

Addressing cross-reactivity and non-specific binding with PIP5K1A antibodies requires multiple strategic approaches. Begin with antibody selection, favoring those validated against PIP5K1A knockout or knockdown samples and those that specifically recognize the intended species orthologue . Implement stringent blocking procedures using species-appropriate blocking agents (5% BSA or 5% non-fat dry milk) in TBST or PBST, with extended blocking times (1-2 hours at room temperature or overnight at 4°C) to minimize non-specific binding . For Western blotting, optimize antibody dilution through titration experiments and extend washing steps (at least 3-4 times for 10 minutes each) with agitation to remove weakly bound antibodies . When non-specific bands persist, employ peptide competition assays where the antibody is pre-incubated with excess immunizing peptide to confirm specific binding . For immunostaining, include isotype controls at matching concentrations to distinguish specific from non-specific binding . Consider pre-adsorption of antibodies with tissue homogenates from species with high homology to remove cross-reactive antibodies. Finally, validate key findings with a second antibody targeting a different epitope of PIP5K1A or with complementary non-antibody approaches to confirm specificity .

How can I optimize PIP5K1A antibody-based detection in tissues with low expression levels?

Detecting low abundance PIP5K1A in tissues requires specialized signal amplification strategies. For immunohistochemistry, implement heat-induced epitope retrieval optimization by testing multiple buffers (citrate pH 6.0, EDTA pH 9.0) at different temperatures and incubation times to maximize antigen accessibility . Extend primary antibody incubation to overnight at 4°C at optimized concentration to increase binding opportunities . Employ signal amplification systems such as tyramide signal amplification (TSA), which can enhance signal 10-100 fold through catalyzed reporter deposition . For fluorescence applications, consider using highly sensitive detection systems like Quantum Dots or Alexa Fluor 647, which offer greater sensitivity and photostability than traditional fluorophores . When working with tissue microarrays, increase section thickness (5-7 μm instead of standard 3-4 μm) to capture more antigen . For Western blotting of low abundance samples, implement protein concentration steps through immunoprecipitation with PIP5K1A antibodies prior to SDS-PAGE . Use high-sensitivity chemiluminescent substrates with extended exposure times or switch to more sensitive detection methods like multiplexed fluorescent Western blotting with near-infrared detection systems .

What are the best approaches for multiplexed detection of PIP5K1A with other signaling proteins?

Multiplexed detection of PIP5K1A with other signaling proteins requires carefully optimized protocols. For immunofluorescence, sequential staining approaches work well when using antibodies from the same species: apply the first primary antibody at lower concentration, detect with a directly labeled secondary antibody, block with excess unconjugated host species IgG, then apply the second primary antibody followed by a distinctly labeled secondary antibody . Alternatively, directly conjugate PIP5K1A antibodies with one fluorophore and other target antibodies with spectrally distinct fluorophores . For chromogenic multiplexing in IHC, sequential chromogenic detection with thorough antibody stripping between rounds or multiplex immunohistochemistry platforms like Opal/Vectra can visualize up to 7 proteins on a single tissue section . In the context of KRAS signaling research, co-detection of PIP5K1A with KRAS, phospho-ERK, and phospho-AKT has proven valuable for understanding pathway activation . For Western blotting, fluorescent multiplexing using secondary antibodies with different spectral properties allows simultaneous detection of multiple proteins, including PIP5K1A and its interacting partners . Mass cytometry (CyTOF) with metal-conjugated antibodies provides another powerful approach for highly multiplexed protein detection without spectral overlap limitations .

How should researchers interpret discrepancies in PIP5K1A detection between different antibodies or techniques?

Interpreting discrepancies in PIP5K1A detection requires systematic troubleshooting and contextual understanding. First, evaluate the epitopes targeted by different antibodies - discrepancies may reflect detection of different isoforms, splice variants, or post-translationally modified forms of PIP5K1A . Consider technique-specific factors: Western blotting detects denatured protein and may miss conformationally-dependent epitopes, while immunostaining preserves native conformation but may have limited epitope accessibility . Differing sensitivity thresholds between techniques may explain why PIP5K1A is detected by one method but not another in samples with low expression . Cross-reactivity with related proteins (such as PIP5K1B or PIP5K1C) may cause false positives in some assays, particularly with antibodies targeting conserved domains . To resolve discrepancies, implement orthogonal validation approaches: if Western blotting and immunostaining yield contradictory results, confirm with mRNA expression analysis, mass spectrometry, or functional assays . When different antibodies produce inconsistent results, prioritize findings from antibodies with the most extensive validation (especially those tested in knockout systems) and confirm key discoveries with genetic approaches such as RNA interference or CRISPR-based gene editing .

What statistical approaches are recommended for quantifying PIP5K1A expression in tissue samples?

Quantifying PIP5K1A expression in tissue samples requires robust statistical approaches. For immunohistochemistry scoring, implement standardized semi-quantitative methods such as H-score (intensity × percentage of positive cells) or Allred score (intensity + proportion) with clearly defined thresholds for categorization (negative, low, moderate, high expression) . Ensure inter-observer reliability by having multiple independent scorers and calculating Cohen's kappa coefficient (>0.75 indicates excellent agreement) . For digital image analysis, employ automated algorithms to measure staining intensity, area, and distribution, reporting quantitative metrics with appropriate statistical parameters (mean, median, interquartile range) . When comparing PIP5K1A expression between groups (e.g., tumor vs. normal, different cancer stages), select appropriate statistical tests based on data distribution: parametric tests (t-test, ANOVA) for normally distributed data or non-parametric alternatives (Mann-Whitney, Kruskal-Wallis) for non-normal distributions . For survival analysis, use Kaplan-Meier curves with log-rank tests to assess prognostic significance of PIP5K1A expression levels . In multivariate analyses, employ Cox proportional hazards models to determine whether PIP5K1A expression independently predicts outcomes when accounting for other clinical variables .

How can researchers distinguish between specific PIP5K1A isoforms and splice variants using antibodies?

Distinguishing between PIP5K1A isoforms and splice variants requires strategic antibody selection and complementary techniques. First, carefully select isoform-specific antibodies targeting unique regions that differ between PIP5K1A variants, reviewing the immunogen sequence information to confirm specificity . Validate these antibodies using overexpression systems with tagged versions of specific PIP5K1A variants to confirm selective detection . When analyzing endogenous expression, combine antibody-based detection with RT-PCR using variant-specific primers to correlate protein and mRNA expression of particular isoforms . For Western blotting applications, use high-resolution gel systems (gradient gels or Phos-tag gels) that can separate closely related isoforms with slight molecular weight differences or different phosphorylation states . Consider two-dimensional gel electrophoresis to separate isoforms based on both molecular weight and isoelectric point before immunoblotting . For immunoprecipitation-mass spectrometry approaches, use antibodies that capture all PIP5K1A forms, then identify specific variants through peptide sequencing, focusing on regions that differ between isoforms . If available, recombinant protein standards representing different PIP5K1A isoforms can serve as positive controls to validate detection specificity and establish migration patterns .

What approaches can resolve contradictory findings regarding PIP5K1A function in different experimental systems?

Resolving contradictory findings about PIP5K1A function requires comprehensive methodological approaches. Begin by thoroughly characterizing the experimental systems, documenting cell type-specific differences in PIP5K1A expression, subcellular localization, and interacting partners using well-validated antibodies . Implement genetic manipulation strategies across multiple systems using both transient (siRNA) and stable (shRNA, CRISPR) approaches to rule out off-target effects or compensation mechanisms that might explain discrepancies . Perform rescue experiments by re-expressing siRNA-resistant PIP5K1A in knockdown cells to confirm phenotype specificity . When contradictory results emerge from different cancer models, consider the genetic background, particularly KRAS mutation status, given PIP5K1A's specific interaction with KRAS . For discrepancies between in vitro and in vivo findings, develop appropriate animal models (conditional knockout, tissue-specific expression) to validate cellular observations in physiological contexts . Employ multiple functional readouts (proliferation, migration, signaling pathway activation) assessed by complementary techniques to build converging evidence . Consider kinetics and dosage effects, as PIP5K1A may have different functions at various expression levels or activation states . Finally, collaborate with other laboratories to independently validate key findings using standardized protocols to distinguish genuine biological differences from technical artifacts .

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.