PPP1CA Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to PPP1CA Monoclonal Antibody

The PPP1CA monoclonal antibody is a laboratory reagent designed to specifically detect and study the catalytic subunit alpha (PPP1CA) of protein phosphatase 1 (PP1), a serine/threonine phosphatase critical for cellular processes such as glycogen metabolism, cell division, and regulation of viral transcription (e.g., HIV-1) . PP1 complexes with regulatory proteins to form holoenzymes that dephosphorylate target substrates, modulating diverse pathways .

Structure and Function of PPP1CA

PPP1CA is one of three catalytic subunits (α, β/δ, γ) of PP1, with distinct isoform-specific roles. Key features include:

PropertyDetail
Molecular Weight38 kDa (calculated), 37 kDa (observed via Western blot)
Subcellular LocalizationPrimarily cytoplasmic and nuclear
Key InteractionsAssociates with >200 regulatory subunits (e.g., PPP1R1A, PPP1R3A) to form holoenzymes
Phosphorylation SitesThr320 (regulated by CDK2, AKT1, LMTK2)

PPP1CA activity is modulated by phosphorylation (e.g., Thr320) and binding to regulatory partners. Dysregulation is linked to heart failure, diabetes, and cancer .

Research Applications of PPP1CA Monoclonal Antibody

The antibody is employed in diverse experimental workflows:

3.1. Western Blot (WB)

  • Detection: Identifies PPP1CA in lysates from human, mouse, rat, pig, zebrafish, and rabbit tissues .

  • Dilution: 1:5,000–1:50,000 (optimized per protocol) .

  • Key Findings: Demonstrated PP1α’s role in centrosome splitting and DNA damage response .

3.2. Immunoprecipitation (IP)

  • Use: Isolates PPP1CA-protein complexes for downstream analysis (e.g., kinase assays) .

3.3. Immunofluorescence (IF)

  • Localization: Visualizes PPP1CA in subcellular compartments (e.g., apical membrane in renal cells) .

  • Dilution: 1:100–1:500 .

Antibody Characteristics: Cross-Reactivity and Specificity

Commercially available antibodies exhibit broad species reactivity and isoform specificity:

VendorCloneReactivityApplications
Prospec Bio P4G3ATHumanWB, ELISA, IHC
Proteintech 67070-1-IgHuman, mouse, rat, pig, zebrafish, rabbitWB, IP, ELISA
Boster Bio A02801Bovine, chicken, human, mouse, pig, ratWB, IF, ICC

5.1. Centrosome Regulation

  • PP1α Isoform-Specific Role: Knockdown of PPP1CA (but not β/δ or γ) disrupts centrosome splitting in response to DNA damage, implicating PP1α in mitotic regulation .

  • Mechanism: PP1α dephosphorylates Nek2 kinase, modulating centrosome dynamics .

5.2. Cardiac and Renal Pathways

  • Potassium Regulation: High dietary potassium increases PPP1CA expression in renal distal convoluted tubules (DCT), promoting NCC dephosphorylation and sodium reabsorption .

  • Cardiac Function: PP1 activity is elevated in heart failure, suggesting a role in disease progression .

5.3. Neural Development

  • Migration and Morphogenesis: PPP1CA regulates neural crest cell migration and optic fissure closure during embryogenesis .

Protocols and Optimization

ApplicationProtocol Highlights
WB1. Block membrane with 5% milk/TBST. 2. Incubate with antibody (1:10,000) overnight.
IF1. Fix cells with 4% PFA. 2. Permeabilize with 0.1% Triton X-100. 3. Probe with antibody (1:200) .

Clinical and Diagnostic Relevance

PPP1CA dysregulation is implicated in:

  • Cancer: Altered PP1 activity in colorectal and Parkinson’s disease models .

  • Metabolic Disorders: Links to glycogen metabolism and insulin signaling .

Product Specs

Form
Purified mouse monoclonal antibody in a buffer containing 0.1M Tris-Glycine (pH 7.4, 150 mM NaCl) with 0.2% sodium azide and 50% glycerol.
Lead Time
Typically, we can ship the products within 1-3 business days of receiving your order. Delivery times may vary depending on the purchasing method and location. Please contact your local distributor for specific delivery timelines.
Synonyms
PP-1A, PPP1A, PP1alpha

Q&A

What is PPP1CA and what cellular functions does it regulate?

PPP1CA is one of three catalytic subunits of protein phosphatase 1 (PP1), a serine/threonine-specific protein phosphatase with wide-ranging regulatory functions. In humans, the canonical protein consists of 330 amino acid residues with a molecular mass of 37.5 kDa and localizes to both nuclear and cytoplasmic compartments . As a member of the PPP phosphatase family, PPP1CA associates with over 200 regulatory proteins to form highly specific holoenzymes that dephosphorylate hundreds of biological targets .

PPP1CA regulates numerous cellular processes, including:

  • Cell division

  • Glycogen metabolism

  • Muscle contractility

  • Protein synthesis

  • HIV-1 viral transcription

  • Cardiac function

  • Learning and memory processes

Research in both human and mouse models indicates that PP1 functions as a significant regulator of cardiac function, with increased PP1 activity detected in end-stage heart failure . Additionally, mouse studies suggest PP1 serves as a suppressor of learning and memory processes .

What experimental applications are most suitable for PPP1CA monoclonal antibodies?

PPP1CA monoclonal antibodies are versatile research tools that have been cited in over 190 scientific publications . The most common applications include:

ApplicationSuitabilityKey Considerations
Western BlotHighly suitableMost widely used application for detection of PPP1CA
ELISASuitableEffective for quantitative analysis
ImmunohistochemistrySuitableEnables tissue localization studies
ImmunoprecipitationVariableDependent on epitope accessibility
ImmunofluorescenceVariableMay require optimization for subcellular localization studies

When selecting a PPP1CA antibody, researchers should consider the specific isoform they wish to detect, as up to three different isoforms have been reported for this protein .

How do I optimize Western blot protocols for PPP1CA detection?

For optimal detection of PPP1CA using Western blot techniques, researchers should consider the following methodological approach:

  • Sample preparation: Include phosphatase inhibitors in lysis buffers to preserve the native phosphorylation state.

  • Protein loading: Load 10-30 μg of total protein per lane for cell lysates.

  • SDS-PAGE separation: Use 10-12% polyacrylamide gels for optimal resolution of the 37.5 kDa PPP1CA protein.

  • Transfer conditions: Transfer to PVDF membranes at 100V for 1 hour or 30V overnight.

  • Blocking: Use 5% non-fat dry milk in TBST (more effective than BSA for PPP1CA detection).

  • Antibody dilution: Typically 1:1000 to 1:2000 for primary antibody incubation.

  • Incubation time: Overnight at 4°C for primary antibody provides optimal signal-to-noise ratio.

  • Washing: Perform at least 3-5 washes with TBST before secondary antibody application.

Detection of specific isoforms may require careful selection of antibody clones like P4G3AT that recognize unique epitopes .

What cross-reactivity should be expected with PPP1CA antibodies across species?

PPP1CA is highly conserved across species, with orthologs reported in mouse, rat, bovine, chimpanzee, and chicken . This conservation enables many PPP1CA antibodies to cross-react across multiple species, which can be advantageous for comparative studies.

Expected cross-reactivity pattern for typical anti-PPP1CA antibodies:

  • Human: Strong reactivity (primary target)

  • Mouse: Strong reactivity due to high sequence homology

  • Rat: Strong reactivity due to high sequence homology

  • Bovine: Moderate to strong reactivity

  • Chimpanzee: Strong reactivity (nearly identical to human)

  • Chicken: Variable reactivity depending on epitope conservation

When working with unconventional model organisms, researchers should validate antibody specificity through Western blot analysis prior to conducting more complex experiments.

How can I distinguish between different PPP1CA isoforms using monoclonal antibodies?

The three known isoforms of PPP1CA present challenges for isoform-specific detection. For successful isoform discrimination:

  • Epitope selection: Choose antibodies raised against regions that differ between isoforms.

  • Molecular weight comparison: Use high-resolution gels (12-15%) to separate closely migrating isoforms.

  • Isoform-specific knockdown: Implement siRNA targeting specific isoforms as controls.

  • 2D gel electrophoresis: Separate isoforms based on both molecular weight and isoelectric point differences.

  • Mass spectrometry validation: Confirm antibody specificity for each isoform using immunoprecipitation followed by mass spectrometry analysis.

For definitive isoform identification, researchers should consider using recombinant isoform standards as positive controls and conduct parallel blots with multiple antibodies targeting different epitopes.

What approaches are most effective for studying PPP1CA interactions with regulatory proteins?

PPP1CA forms holoenzymes with over 200 regulatory proteins, creating complexes with distinct functions and substrate specificities . To study these interactions effectively:

  • Co-immunoprecipitation: Use anti-PPP1CA antibodies to pull down complexes, followed by identification of interacting partners. This approach has revealed important interactions, such as the Phactr1/PP1 complex .

  • Proximity ligation assays: Detect protein-protein interactions in situ with high sensitivity.

  • BiFC (Bimolecular Fluorescence Complementation): Visualize interactions in living cells.

  • Pull-down assays with RVxF motif peptides: Many PPP1CA-interacting proteins utilize RVxF motifs for binding, as demonstrated in studies of Phactr1 .

  • Structural analysis: Crystallography studies have revealed how partners like Phactr1 remodel the PP1 substrate-binding grooves to influence substrate recognition .

The Phactr1/PP1 complex demonstrates how partner binding can dramatically alter substrate specificity, enhancing catalytic efficiency up to 400-fold for specific substrates compared to apo-PP1 .

How does substrate recognition by PPP1CA complexes determine dephosphorylation specificity?

PPP1CA substrate specificity is largely determined by its regulatory binding partners. The Phactr1/PP1 complex provides an excellent model for understanding this mechanism:

  • Surface remodeling: Phactr1 binding creates a composite hydrophobic pocket and adjacent amphipathic cavity topped by a basic rim that profoundly transforms the surface of PP1 adjacent to its catalytic site .

  • Recognition motifs: Phactr1/PP1 substrates can be defined by a hydrophobic doublet at position +4/+5 or +3/+4 relative to the dephosphorylation site, with leucine preferred at the distal position, embedded within acidic sequences (core recognition sequence: S/T-x(2-3)-φ-L) .

  • Binding efficiency: Substrate interactions with the modified hydrophobic pocket provide additional binding energy that enhances catalytic rates compared to apo-PP1 or other PP1 complexes .

  • Substrate orientation: Structural studies show that Phactr1/PP1 substrates dock with the catalytic cleft in a specific orientation that differs from other phosphatase-substrate complexes .

To experimentally identify specific substrates for PPP1CA complexes, researchers can use immunoprecipitation followed by phosphoproteomic analysis, or employ substrate-trapping mutants that bind but do not dephosphorylate targets.

What techniques can be used to study the role of PPP1CA in cardiac function and heart failure?

Given that increased PP1 activity is detected in end-stage heart failure , researchers can employ the following approaches:

  • Tissue-specific expression analysis: Compare PPP1CA levels in healthy versus diseased cardiac tissue using immunohistochemistry with monoclonal antibodies.

  • Substrate profiling: Identify cardiac-specific substrates using phosphoproteomic approaches combined with PPP1CA inhibition or depletion.

  • Animal models: Analyze cardiac phenotypes in PPP1CA knockout or overexpression mouse models.

  • Patient sample analysis: Quantify PPP1CA levels and activity in human cardiac biopsy samples using activity assays and immunoblotting.

  • Structure-function studies: Investigate how PPP1CA forms complexes with cardiac-specific regulatory proteins using co-immunoprecipitation and structural biology approaches.

These techniques can help elucidate how PPP1CA dysregulation contributes to pathological cardiac remodeling and identify potential therapeutic targets for heart failure.

What are common causes of non-specific binding with PPP1CA antibodies and how can they be minimized?

Non-specific binding is a frequent challenge when working with PPP1CA antibodies due to the high conservation of phosphatase catalytic domains. To minimize these issues:

  • Antibody selection: Choose monoclonal antibodies like clone P4G3AT that target unique epitopes .

  • Blocking optimization:

    • Test different blocking agents (BSA, non-fat milk, commercial blockers)

    • Extend blocking time to 2 hours at room temperature

    • Include 0.1-0.3% Tween-20 in blocking solutions

  • Dilution optimization: Titrate antibody concentrations to determine optimal signal-to-noise ratio.

  • Cross-adsorption: Pre-incubate antibodies with lysates from cells lacking PPP1CA to remove antibodies that bind to non-specific epitopes.

  • Validation with knockdown/knockout controls: Include samples with reduced or absent PPP1CA expression to verify specificity.

  • Sample preparation: Include phosphatase inhibitors and reduce proteolytic degradation by using fresh samples and appropriate protease inhibitors.

How can I validate the specificity of PPP1CA antibodies in my experimental system?

Rigorous validation of PPP1CA antibodies is essential for generating reliable research data. Implement the following validation strategy:

  • Positive controls: Include recombinant PPP1CA protein or lysates from cells with known PPP1CA expression.

  • Negative controls:

    • Use siRNA/shRNA knockdown of PPP1CA

    • When possible, include CRISPR-Cas9 knockout cell lines

    • Pre-absorb antibody with immunizing peptide/protein

  • Orthogonal detection methods: Compare results using alternative antibodies that recognize different epitopes of PPP1CA.

  • Immunoprecipitation followed by mass spectrometry: Confirm that the antibody is capturing the intended target.

  • Detection of expected interacting partners: Verify that the antibody can co-immunoprecipitate known PPP1CA binding partners such as those containing RVxF motifs .

  • Cross-reactivity assessment: Test for cross-reactivity with other PPP family members, particularly PPP1CB and PPP1CC catalytic subunits, which share high sequence homology.

How can PPP1CA monoclonal antibodies be used to study substrate-specific dephosphorylation in complex cellular environments?

Advanced studies of PPP1CA substrate specificity can employ the following methodological approaches:

  • Substrate-specific phospho-antibodies: Use in conjunction with PPP1CA inhibition to monitor dephosphorylation of specific substrates.

  • Proximity-dependent biotinylation (BioID): Identify substrates that physically interact with PPP1CA complexes in living cells.

  • Reconstitution experiments: Use purified PPP1CA holoenzymes and synthetic phosphopeptides to measure dephosphorylation kinetics in vitro, as demonstrated with the Phactr1/PP1 complex .

  • Structure-guided mutagenesis: Create mutations in the composite binding surfaces formed by PPP1CA and its regulatory partners to alter substrate specificity .

  • Phosphoproteomic analysis: Compare phosphorylation changes upon manipulation of specific PPP1CA holoenzymes.

The Phactr1/PP1 complex provides an excellent model system, as it exhibits 100- to 400-fold greater catalytic efficiency against specific substrates compared with apo-PP1 or other PP1 complexes . This approach could be extended to study other regulatory complexes.

What emerging technologies are advancing PPP1CA research beyond traditional antibody applications?

While monoclonal antibodies remain essential tools, several emerging technologies are expanding PPP1CA research capabilities:

  • CRISPR-Cas9 genome editing: Generate endogenously tagged PPP1CA to monitor dynamics in live cells without antibodies.

  • Single-molecule imaging: Track individual PPP1CA molecules and their interactions in living cells.

  • Optogenetic control of PPP1CA activity: Use light-controlled protein interaction domains to regulate PPP1CA complex formation.

  • PPP1CA-specific small molecule modulators: Develop compounds that target specific PPP1CA holoenzymes rather than the general phosphatase activity.

  • Cryo-EM analysis of PPP1CA complexes: Determine structures of PPP1CA bound to various regulatory partners and substrates.

  • PP1-fusion proteins: Create engineered PP1-regulatory protein fusions to study specific holoenzyme functions, as demonstrated with PP1-Phactr1 fusion proteins that retain substrate specificity similar to intact Phactr1/PP1 holoenzyme .

These technologies promise to provide more precise understanding of PPP1CA function with less reliance on antibody-based detection methods.

How can structural insights into PPP1CA complexes guide the development of specific inhibitors?

Structural studies of PPP1CA in complex with regulatory proteins reveal opportunities for developing specific inhibitors:

  • Targeting composite surfaces: The remodeled PP1 hydrophobic groove created by regulatory protein binding creates unique composite surfaces that could be targeted by small molecules .

  • Disrupting specific interactions: Small molecules could be designed to disrupt the interaction between PPP1CA and specific regulatory proteins.

  • Structure-guided drug design: Crystal structures of PPP1CA complexes, such as Phactr1/PP1, provide templates for virtual screening and rational design of inhibitors .

  • Pocket-specific inhibitors: The composite pocket formed by Phactr1 binding to PP1 increases binding efficiency and reactivity 100-fold compared to PP1 alone , suggesting that targeting such pockets could yield highly specific inhibitors.

  • Allosteric modulators: Identify compounds that bind to allosteric sites and alter the conformation or dynamics of PPP1CA complexes.

Current phosphatase inhibitors lack specificity for particular phosphatase complexes, but the composite surfaces formed by regulatory protein binding offer promising targets for developing complex-specific inhibitors .

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 2024 Thebiotek. All Rights Reserved.