pho Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship products within 1-3 business days of receiving your order. Delivery time may vary depending on the purchasing method or location. For specific delivery times, please consult your local distributor.
Synonyms
Polycomb protein PHO (Protein pleiohomeotic) (Transcription factor YY1 homolog) pho CG17743
Target Names
pho
Uniprot No.

Target Background

Function
Pleiohomeotic (Pho) is a Polycomb group (PcG) protein that binds to the 5'-CNGCCATNNNNG-3' sequence located in the regulatory regions of numerous genes. PcG proteins function by forming multiprotein complexes, which are crucial for maintaining the transcriptionally repressive state of homeotic genes throughout development. While PcG proteins are not necessary for initiating repression, they are essential for maintaining it during later developmental stages. These proteins likely exert their influence through histone methylation, leading to heritable changes in chromatin expressibility. Pho is believed to target the Esc/E(z) complex to DNA. Although necessary, Pho is not sufficient to recruit a fully functional PcG repressive complex that silences target genes. This suggests that recruitment of the distinct PRC1 complex is also required for subsequent repression. Pho is proposed as a core component of the chromatin remodeling Ino80 complex, which plays a role in transcriptional regulation, DNA replication, and potentially DNA repair.
Gene References Into Functions
  1. Mutations within the protein-protein interaction domain (REPO domain) of Pho disrupt the dynamic interaction with Spt5. Consequently, the transcriptional kinetics of the heat shock response are negatively affected by mutations in the REPO domain of Pho. PMID: 29208012
  2. Spt5 engages in both physical and genetic interactions with the Polycomb Group (PcG) protein Pleiohomeotic (Pho). PMID: 23894613
  3. The TF homolog pair Pho/Yy1 appears to have distinct roles in PcG targeting between Drosophila and mouse. PMID: 20353814
  4. Pleiohomeotic protein is involved in polycomb response element-mediated silencing for transcriptional repression. PMID: 12370294
  5. This protein acts as a PcG protein in vivo. PMID: 12628927
  6. The mammalian transcription factor YY1 can function as a PcG protein in Drosophila, leading to transcriptional repression and correction of phenotypic defects caused by mutations in its homologue, Pleiohomeotic (PHO). PMID: 16052488
  7. Pleiohomeotic (PHO) and a multiprotein Polycomb core complex (PCC) exhibit highly cooperative binding to polycomb response elements. PMID: 16077005
  8. Grainyhead interacts genetically with pleiohomeotic in a transgene-based, pairing-dependent silencing assay. PMID: 16449654
  9. YY1 sequences 201-226 are both necessary and sufficient for Polycomb group transcriptional repression in vivo. PMID: 17158804
  10. These findings suggest that pho plays a role in maintaining the expression of genes involved in the formation of ventral appendages and the proximo-distal axis. PMID: 18495104
  11. Pho is a general component of the maintenance machinery, as the majority of Pc targets are also associated with Pho (Pleiohomeotic) binding. PMID: 18773083
  12. A DNA-binding complex requiring Pho is differentially regulated to facilitate epigenetic transcriptional memory of both the active and repressed states. PMID: 19029043

Show More

Hide All

Database Links

KEGG: dme:Dmel_CG17743

STRING: 7227.FBpp0088268

UniGene: Dm.1406

Subcellular Location
Nucleus.

Q&A

What criteria should be used to select a phospho-specific antibody for experimental use?

When selecting a phospho-specific antibody, researchers should consider several critical factors to ensure reliable experimental outcomes. First, confirm the antibody specifically detects the protein of interest only when phosphorylated at the indicated site, not in its unphosphorylated state . Check that the antibody detects a band at the correct molecular weight for your phosphorylated target protein, keeping in mind that each phosphorylation adds approximately 80Da to the protein's molecular weight .

The validation method used by the manufacturer is also important - look for antibodies validated by comparing phosphorylation levels in treated versus untreated cell lysates and by dephosphorylating western blot membranes . This dual validation approach provides quantitative measurements of the antibody's specificity and performance under different conditions.

For high-throughput screening applications, alkaline phosphatase (AP) treatment can serve as an independent predictive factor to assess phosphorylation antibodies. Studies have demonstrated that antibodies showing a negative log fold change (logFC ≤ -0.792) following AP treatment typically exhibit superior performance, with approximately 85% showing meaningful single bands at expected sizes in subsequent western blot verification .

How can I verify if my phospho-antibody is truly detecting phosphorylated proteins?

Rigorous validation of phospho-antibody specificity requires multiple complementary approaches. The most reliable verification method involves using appropriate controls:

  • Positive controls: Use cell lysates or tissues known to contain the phosphorylated protein of interest, ideally after treatment with specific activators of the relevant signaling pathway .

  • Negative controls: Treat samples with alkaline phosphatase to remove phosphate groups from all proteins. This should significantly reduce or eliminate signal from true phospho-specific antibodies .

  • Peptide competition assays: Compare blocking with phosphorylated versus non-phosphorylated epitope peptides. A true phospho-specific antibody will only be blocked by the phosphorylated peptide .

  • Genetic controls: When possible, use samples from knockout models lacking your protein of interest. Any signal detected in these samples indicates cross-reactivity with other proteins .

  • Phosphorylation site mutants: For advanced validation, use cells expressing mutant proteins where the phosphorylation site has been altered (e.g., serine to alanine mutation) .

Importantly, verification should be performed in the specific experimental context where the antibody will be used, as antibody performance can vary between applications (western blot, immunohistochemistry, etc.) and sample types (cell lines versus tissues) .

What are the best practices for western blot detection of phosphorylation events?

For optimal western blot detection of phosphorylation events, follow these methodological guidelines:

  • Sample preparation: Keep samples on ice and pre-chill all buffers to minimize enzyme activity that could degrade or dephosphorylate proteins. Supplement lysis buffers with both protease and phosphatase inhibitor cocktails .

  • Blocking reagent selection: For phosphorylated protein analysis, start with casein in TBS or BSA rather than milk. Milk contains proteins that can interfere with detection of phosphorylated proteins, potentially increasing background signal .

  • Antibody probing sequence: When planning to strip and re-probe blots, always probe for the phosphoprotein first. The stripping process often leads to loss or degradation of phosphoprotein antigens .

  • Incubation conditions: Due to the typically low abundance of phosphoproteins, incubate with primary anti-phosphoprotein antibodies overnight at 4°C for optimal sensitivity .

  • Loading controls: Use an antibody that detects the total level of your target protein to determine the phosphorylated fraction relative to the total fraction. This allows comparison of different treatments and provides an internal loading control .

  • Multiplexing: Consider fluorescent western blotting with dual detection of phosphorylated and total protein simultaneously, which provides more accurate quantification of phosphorylation status .

  • Sample enrichment: For low-abundance phosphoproteins, consider immunoprecipitation to concentrate the antigen before western blotting to avoid false negative results .

How should I troubleshoot non-specific bands when using phospho-antibodies?

Non-specific bands are a common challenge when working with phospho-antibodies. Systematic troubleshooting should include:

  • Verify with knockout/knockdown samples: Test the antibody in samples where your protein of interest is absent. Any bands appearing in these samples are definitively non-specific .

  • Molecular weight assessment: Carefully evaluate whether detected bands match the expected molecular weight of your phosphorylated protein. Remember that phosphorylation increases molecular weight by approximately 80Da per phosphate group .

  • Analyze protein sequence homology: Cross-reactivity often occurs with proteins containing similar phosphorylation motifs. Search databases for proteins with sequence similarity around the phosphorylation site of interest .

  • Optimize blocking conditions: Test alternative blocking reagents beyond BSA or casein, such as chicken ovalbumin or fish gelatin, which may reduce non-specific binding .

  • Adjust antibody concentration: Titrate the antibody to find the optimal concentration that maximizes specific signal while minimizing background .

  • Perform dephosphorylation controls: Treat duplicate samples with alkaline phosphatase - true phospho-specific signals should disappear or significantly diminish, while non-specific signals will remain unchanged .

Research has shown that many commercially available phospho-antibodies exhibit cross-reactivity with multiple proteins. For example, studies on phospho-S129 alpha-synuclein antibodies demonstrated that they detect non-specific low and high molecular weight bands in alpha-synuclein knockout samples that could easily be mistaken for monomeric or high molecular weight alpha-synuclein species .

How does sample preparation affect phospho-antibody performance across different experimental systems?

Sample preparation significantly impacts phospho-antibody performance, and protocols must be adapted to different experimental systems:

  • Cell line samples: For phosphoprotein preservation in cell lines, rapid lysis in ice-cold buffers containing phosphatase inhibitors is essential. Compared to other sample types, cell lines typically provide the most consistent results with phospho-antibodies due to their homogeneity .

  • Tissue samples: Fresh frozen (FF) versus formalin-fixed paraffin-embedded (FFPE) tissues show variable correlation in phosphoprotein detection. Studies have reported approximately 40% concordance rate for antibodies profiled on FFPE samples compared to matching FF samples . For FFPE samples, efficient protein extraction requires SDS-based denaturation and a heating step for crosslinking reversal .

  • Primary neuron cultures: These require particularly gentle lysis conditions to preserve phosphorylation states, and may benefit from specialized lysis buffers that maintain the native state of complex protein structures .

  • In vivo models: Different phospho-antibodies exhibit differential ability to capture the diversity of protein aggregates in neuronal and in vivo models of pathology. For instance, not all phospho-S129 antibodies capture the biochemical and morphological diversity of alpha-synuclein pathology equally well .

When transitioning between sample types, researchers should not assume antibody performance will remain consistent. For tissue-based profiling, separate antibody validation is recommended as discrepancies may exist between tissues and cell lines due to heterogeneity and preanalytical variables affecting phosphoprotein detection .

How can multiplexing techniques enhance phosphorylation analysis?

Multiplexing techniques offer significant advantages for comprehensive phosphorylation analysis:

  • Simultaneous detection of multiple phosphorylation states: Fluorescent multiplexing allows researchers to simultaneously measure phosphorylated and total protein levels in a single experiment, eliminating the variability introduced by stripping and reprobing membranes .

  • Pathway-level analysis: By detecting multiple phosphorylation events within the same signaling pathway simultaneously, researchers can obtain a more complete picture of signaling dynamics and cross-talk between pathways .

  • Implementation strategies: For western blotting, use primary antibodies from different host species (e.g., rabbit and mouse) paired with species-specific secondary antibodies conjugated to different fluorophores . For microscopy or flow cytometry, select fluorophores with minimal spectral overlap.

  • Data normalization: Multiplexing facilitates more accurate normalization, as the ratio of phosphorylated to total protein can be determined from the same sample region, eliminating spatial variation effects .

  • High-throughput screening applications: Reverse Phase Protein Array (RPPA) methods combined with multiplexed detection enable simultaneous profiling of dozens to hundreds of phosphorylation events across multiple samples. This approach has been successfully used for clinical applications, showing significant correlation with pathological markers in both melanoma and lung cancer FFPE samples .

In clinical proteomics, multiplexed RPPA profiling has demonstrated great interexperimental reproducibility and significant correlation with pathological markers, generating meaningful data that match clinical features and setting benchmarks for efficient workflows compatible with precision oncology applications .

What control experiments are essential when publishing research using phospho-antibodies?

When publishing research involving phospho-antibodies, the following control experiments should be considered essential for ensuring reliability and reproducibility:

  • Specificity controls:

    • Demonstrate antibody specificity through dephosphorylation experiments (e.g., alkaline phosphatase treatment)

    • Include knockout/knockdown samples when available, or use siRNA/shRNA to reduce target protein expression

    • Show peptide competition results with phosphorylated and non-phosphorylated peptides

  • Validation across methods:

    • Confirm key findings with at least two independent methods (e.g., western blot and immunofluorescence)

    • For critical findings, consider validation with a second phospho-antibody targeting the same site but from a different commercial source

  • Quantification controls:

    • Always include loading controls and normalize phospho-signals to total protein levels

    • Include positive control samples with known phosphorylation status

    • Demonstrate dose-response relationships with activators/inhibitors of the relevant pathway

  • Technical validation:

    • Report antibody catalog numbers, lot numbers, and complete experimental conditions

    • Provide full-length blot images including molecular weight markers

    • Demonstrate reproducibility across multiple experimental replicates

  • Biological validation:

    • Show consistency of findings across relevant cell types or tissues

    • Validate with phosphomimetic or phospho-dead mutants when feasible

    • Correlate phosphorylation changes with functional outcomes

Research has demonstrated that phospho-antibodies vary significantly in their specificity and reliability. For example, studies of PP2Ac Tyr307 phospho-specific antibodies (clones E155 and F-8) found they could not reliably differentiate between phosphorylated and unphosphorylated forms of PP2Ac , highlighting the importance of rigorous controls.

How can I standardize phospho-protein quantification across different experiments?

Standardizing phospho-protein quantification requires systematic approaches to minimize technical and biological variability:

For high-throughput applications like RPPA, researchers have developed quantitative capability evaluations using pathological markers. Studies have found that certain markers like human epidermal growth factor receptor 2 (HER2) exhibit nearly 100% concordance between immunohistochemistry and RPPA based on signal intensity cutoffs, while other markers like estrogen receptor and progesterone receptor show less consistent results .

What are the current limitations of phospho-antibodies and how might they be overcome?

Current limitations of phospho-antibodies include:

  • Cross-reactivity issues: Many phospho-antibodies show cross-reactivity with other proteins, particularly those with similar phosphorylation motifs. This problem is especially pronounced when proteins of interest are expressed at low levels .

    Potential solutions:

    • Development of more stringent validation pipelines incorporating multiple controls

    • Use of complementary mass spectrometry-based phosphoproteomic approaches

    • Implementation of CRISPR-based tagging of endogenous proteins for antibody-independent detection

  • Context-dependent performance: Antibody performance often varies between applications (western blot, immunohistochemistry, flow cytometry) and between sample types (cell lines, tissues) .

    Potential solutions:

    • Application-specific validation of each antibody

    • Development of universal epitope tags that can be inserted at phosphorylation sites

    • Creation of comprehensive databases documenting antibody performance across applications

  • Sensitivity to neighboring modifications: Many phospho-antibodies are sensitive to modifications at adjacent amino acids, complicating interpretation when multiple sites are modified simultaneously .

    Potential solutions:

    • Development of antibodies insensitive to neighboring post-translational modifications

    • Combined approaches that detect multiple modifications simultaneously

    • Computational modeling to predict modification interactions

  • Temporal resolution limitations: Traditional antibody-based methods provide static snapshots rather than dynamic views of phosphorylation events.

    Potential solutions:

    • Development of biosensors for real-time monitoring of phosphorylation in living cells

    • Integration with optogenetic tools for precise temporal control

    • Advanced microscopy techniques with improved temporal resolution

Research on phospho-S129 alpha-synuclein antibodies has underscored the need for more antibodies that are not sensitive to neighboring post-translational modifications and more thorough characterization and validation of existing and new antibodies . Similarly, studies have shown that antibodies targeting phosphorylated PP2Ac at Tyr307 have been problematic, necessitating reinterpretation of previous research findings .

How are phospho-antibodies being integrated with emerging technologies for enhanced phosphorylation analysis?

Phospho-antibodies are increasingly being integrated with cutting-edge technologies to advance phosphorylation analysis:

  • Single-cell phosphoproteomics:

    • Phospho-antibodies coupled with mass cytometry (CyTOF) allow simultaneous measurement of multiple phosphorylation events at the single-cell level

    • Microfluidic platforms combined with phospho-antibodies enable analysis of signaling heterogeneity within cell populations

    • Single-cell western blotting technologies are being adapted for phosphoprotein detection

  • Spatial phosphoproteomics:

    • Multiplexed immunofluorescence with phospho-antibodies preserves spatial information about phosphorylation events within tissues

    • Digital spatial profiling combines phospho-antibodies with precise spatial resolution

    • Spatial transcriptomics integration allows correlation between phosphorylation states and gene expression patterns

  • High-throughput screening platforms:

    • Reverse Phase Protein Array (RPPA) approaches allow for efficient phospho-antibody characterization and high-throughput screening

    • Antibody microarrays enable simultaneous detection of hundreds of phosphorylation events

    • Automated liquid handling systems increase reproducibility of phospho-antibody experiments

  • Computational integration:

    • Machine learning algorithms help predict antibody specificity and cross-reactivity

    • Network analysis tools integrate phosphoproteomic data with other omics datasets

    • Pathway modeling incorporates quantitative phosphorylation data to predict cellular responses

In clinical proteomics, RPPA-based phospho-antibody methodologies have shown significant potential for precision oncology applications. Studies have demonstrated strong correlations between RPPA results and clinical features in melanoma and lung cancer samples, setting benchmarks for efficient workflows compatible with high-throughput clinical proteomics .

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.