PPP1CA Human

Protein Phosphatase 1, Catalytic Subunit Alpha Human Recombinant
Shipped with Ice Packs
In Stock

Description

Overview of PPP1CA Human

PPP1CA (Protein Phosphatase 1 Catalytic Subunit Alpha) is a human gene encoding the alpha isoform of the catalytic subunit of Protein Phosphatase 1 (PP1), a serine/threonine phosphatase critical for regulating diverse cellular processes . The PPP1CA protein forms holoenzymes by associating with over 200 regulatory subunits, enabling substrate-specific dephosphorylation . It is ubiquitously expressed and plays essential roles in glycogen metabolism, cell cycle progression, muscle contraction, synaptic plasticity, and circadian rhythm regulation .

Key Protein Features

PropertyDetail
Gene ID5499 (NCBI Gene)
UniProt IDP62136
Amino Acid Length330 residues
Molecular Weight~37.5–39.7 kDa (varies by isoform and expression system)
Enzyme ClassEC 3.1.3.16 (Protein serine/threonine phosphatase)
Key DomainsPP1 phosphatase catalytic domain (residues 43–297)

Three alternatively spliced transcript variants encode distinct isoforms, with isoform 1 being the canonical form . Post-translational modifications include phosphorylation at Thr-320, which regulates activity .

Recombinant PPP1CA Protein Production

Recombinant human PPP1CA is widely used in biochemical studies. Key commercial variants include:

VendorExpression SystemPuritySpecific ActivityApplications
AbcamBaculovirus/Sf9>75%228–250 nmol/min/mg WB, phosphatase assays
Prospec BioE. coli>80%>3,000 units/mg Enzyme kinetics, autophagy studies

Note: Activity assays typically measure hydrolysis of p-nitrophenyl phosphate (pNPP) .

Biological Functions and Mechanisms

PPP1CA regulates:

  • Cell Division: Controls mitotic exit via chromatin structure modulation and centrosome dynamics .

  • Metabolism: Dephosphorylates glycogen synthase to promote glycogen storage .

  • Neuronal Function: Modulates long-term synaptic plasticity by dephosphorylating Ca²⁺/calmodulin-dependent kinase II .

  • Circadian Rhythms: Balances phosphorylation of PER1/PER2 with CSNK1D/E to determine period length .

Pathophysiological Roles

Disease/ConditionMechanismCitation
HypertensionDephosphorylates NCC cotransporter in response to dietary potassium, reducing blood pressure
Heart FailureOverexpression in mice induces premature cardiac dysfunction
Intervertebral Disc DegenerationSUV39H2-mediated lysine methylation disrupts TFEB activation, impairing autophagy
Rheumatoid ArthritisInactivates FOXP3 in Treg cells via dephosphorylation, promoting immune dysfunction

Key Findings (2023–2025)

  1. Dietary Potassium Signaling
    High potassium intake suppresses Ppp1r1a expression, activating PPP1CA to dephosphorylate NCC in renal cells, offering mechanistic insights into salt-sensitive hypertension .

  2. Autophagy and Senescence
    Lysine 141 methylation by SUV39H2 disrupts PPP1CA-PPP1R9B interaction, preventing TFEB dephosphorylation and nuclear translocation in intervertebral disc degeneration .

  3. Aging and Cognitive Decline
    PP1 inhibition preserves memory in aged mice, implicating PPP1CA in cognitive aging .

Interaction Network

PPP1CA partners with diverse regulators and substrates:

  • Regulatory Subunits: PPP1R1A, PPP1R9B, and PPP1R8 .

  • Kinase Cross-Talk: Antagonizes WNK/SPAK signaling in renal ion transport .

  • Disease-Linked Proteins: BRCA1, TP53, and BCL2 .

Therapeutic Potential

Targeting PPP1CA modulation is being explored for:

  • Hypertension: Enhancing NCC dephosphorylation via potassium-rich diets .

  • Neurodegeneration: Inhibiting PP1 to mitigate age-related memory loss .

  • Cancer: Exploiting PP1’s role in oncogene-induced senescence .

Challenges and Future Directions

Current limitations include PPP1CA’s broad substrate specificity and pleiotropic effects. Emerging strategies focus on:

  • Developing subunit-specific PP1 modulators.

  • Investigating tissue-specific isoforms in disease models .

Product Specs

Introduction
Serine/Threonine-protein phosphatase PP1-alpha catalytic subunit (PPP1CA) is essential for cell division and plays a role in regulating glycogen metabolism, muscle contraction, and protein synthesis. PPP1CA may be important in dephosphorylating substrates like the postsynaptic density-associated Ca2+/calmodulin-dependent protein kinase II.
Description
Recombinant human PPP1CA, expressed in E. coli, is a single, non-glycosylated polypeptide chain consisting of 350 amino acids (residues 1-330) with a molecular weight of 39.7 kDa. This protein contains a 20 amino acid His-tag at the N-terminus and is purified using proprietary chromatographic techniques.
Physical Appearance
A clear, yellowish solution, sterile-filtered.
Formulation
The PPP1CA protein solution (0.2 mg/ml) is supplied in a buffer containing 50 mM Tris-HCl (pH 8.5), 0.2 M NaCl, 1 mM DTT, 0.1 mM PMSF, 1 mM MnCl2, and 50% glycerol.
Stability
For short-term storage (up to 4 weeks), keep at 4°C. For extended storage, freeze at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freezing and thawing.
Purity
Purity is determined to be greater than 80.0% by SDS-PAGE analysis.
Biological Activity
The specific activity of this protein is greater than 3,000 units/mg. Enzymatic activity was confirmed by determining the enzyme's ability to hydrolyze p-nitrophenyl phosphate (pNPP). One unit is defined as the amount of enzyme that hydrolyzes 1 nanomole of pNPP per minute at 37°C, pH 7.5, using a 30 mM substrate concentration.
Synonyms
Serine/threonine-protein phosphatase PP1-alpha catalytic subunit, PP-1A, PPP1CA, PPP1A, PP1alpha.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MSDSEKLNLD SIIGRLLEVQ GSRPGKNVQL TENEIRGLCL KSREIFLSQP ILLELEAPLK ICGDIHGQYY DLLRLFEYGG FPPESNYLFL GDYVDRGKQS LETICLLLAY KIKYPENFFL LRGNHECASI NRIYGFYDEC KRRYNIKLWK TFTDCFNCLP IAAIVDEKIF CCHGGLSPDL QSMEQIRRIM RPTDVPDQGL LCDLLWSDPD KDVQGWGEND RGVSFTFGAE VVAKFLHKHD LDLICRAHQV VEDGYEFFAK RQLVTLFSAP NYCGEFDNAG AMMSVDETLM CSFQILKPAD KNKGKYGQFS GLNPGGRPIT PPRNSAKAKK.

Q&A

What is PPP1CA and what is its primary function in human cells?

PPP1CA (Protein Phosphatase 1 Catalytic Subunit Alpha) is one of the catalytic subunits of Protein Phosphatase 1 (PP1), an abundant and ubiquitously expressed eukaryotic serine-threonine phosphatase. PP1 plays crucial roles in regulating diverse cellular processes including actomyosin contractility, glycogen metabolism, cell cycle progression, gene expression, protein synthesis, and neuronal signaling .

A particularly important function of PPP1CA occurs in complex with regulatory subunits of the PPP1R15 family (GADD34 and CReP), which direct the dephosphorylation of eukaryotic initiation factor 2-alpha (eIF2α) during cellular stress responses . This action plays a critical role in restoring protein synthesis following stress-induced translational attenuation.

How does PPP1CA differ from other PP1 isoforms in humans?

In mammals, there are three genes encoding different PP1 isoforms: PP1α (encoded by PPP1CA), PP1β, and PP1γ. These isoforms share high sequence similarity but exhibit subtle functional differences. All three isoforms have nearly ubiquitous expression across tissues, and their amino acid sequences have diverged relatively little during evolution, although important splice variants have been identified .

Despite their similarities, research indicates that these isoforms are not completely functionally redundant. The specific differences between PPP1CA and other PP1 isoforms include:

These differences, though subtle, can result in isoform-specific functions in particular cellular contexts, making PPP1CA research distinct from studies of other PP1 isoforms .

What are the major protein domains and structural features of PPP1CA?

PPP1CA consists of several key structural domains that contribute to its catalytic function and ability to interact with regulatory proteins:

  • Catalytic core domain: Contains the active site with metal ion cofactors (typically manganese or iron) essential for catalysis

  • C-terminal domain: Contains regulatory elements involved in substrate recognition

  • Hydrophobic groove: A surface feature that serves as a binding site for many regulatory subunits

  • Acidic groove: Another surface feature involved in protein-protein interactions

  • RVxF-binding pocket: A critical site for interaction with most regulatory subunits that contain the consensus RVxF motif

  • SILK-binding region: A secondary interaction site for some regulatory partners

  • MyPhoNE-binding site: An interaction surface for myosin phosphatase N-terminal element

These structural features enable PPP1CA to form specific complexes with various regulatory subunits, which in turn determine the subcellular localization, substrate specificity, and regulation of the phosphatase activity. The three-dimensional arrangement of these domains creates a sophisticated platform for controlled dephosphorylation activities within different cellular contexts.

How does PPP1CA contribute to the Integrated Stress Response (ISR)?

PPP1CA plays a central role in the Integrated Stress Response (ISR) by mediating the dephosphorylation of eukaryotic initiation factor 2-alpha (eIF2α). The ISR is a cellular adaptive mechanism that responds to various stressors including ER stress, amino acid deprivation, viral infection, and oxidative stress .

The process involves several key steps:

  • Stress detection: Various cellular stressors activate specific eIF2α kinases (including PERK, PKR, GCN2, and HRI)

  • Translation attenuation: These kinases phosphorylate eIF2α at Ser51, which inhibits GDP-GTP exchange required for translation initiation, leading to reduced global protein synthesis

  • Selective translation: Despite global translation suppression, certain mRNAs with specific features (like upstream open reading frames) are preferentially translated, including those encoding stress response proteins like ATF4

  • Recovery phase: PPP1CA, in complex with regulatory subunits from the PPP1R15 family (GADD34 and CReP), dephosphorylates eIF2α to restore normal translation

The timing of PPP1CA-mediated dephosphorylation is critical for cell fate decisions. Early in the stress response (30 minutes to 1 hour), eIF2α phosphorylation levels remain high as GADD34 and CReP levels are low. In the later phase (around 4 hours post-stress), GADD34 expression increases, enhancing PPP1CA-mediated eIF2α dephosphorylation, which is essential for the return of normal protein synthesis and cellular recovery .

This regulated balance between phosphorylation and dephosphorylation determines whether cells adapt to stress or undergo apoptosis, making PPP1CA a crucial determinant in cell survival decisions during stress conditions.

What is the relationship between PPP1CA and the PPP1R15 regulatory subunits?

PPP1CA forms functional complexes with regulatory subunits from the PPP1R15 family, particularly GADD34 (encoded by PPP1R15A) and CReP (Constitutive Repressor of eIF2α Phosphorylation, encoded by PPP1R15B). These interactions are fundamental to the regulation of the integrated stress response and unfolded protein response .

Key aspects of these relationships include:

  • Complex formation: Both GADD34 and CReP function as targeting/regulatory subunits that bind to PPP1CA and direct it toward specific substrates, particularly phosphorylated eIF2α

  • Differential expression patterns:

    • CReP is constitutively expressed and maintains basal eIF2α dephosphorylation

    • GADD34 is stress-inducible, with expression increasing several hours after stress initiation

  • Binding mechanisms: Both regulatory subunits contain:

    • PP1-binding motifs (including the RVxF motif) that enable direct interaction with PPP1CA

    • C-terminal regions that facilitate substrate (eIF2α) recruitment to the phosphatase complex

  • Functional consequences:

    • The PPP1CA-CReP complex maintains low levels of eIF2α phosphorylation under normal conditions

    • The PPP1CA-GADD34 complex is crucial for recovery from stress-induced translational arrest, with GADD34 expression increasing around 4 hours post-stress to restore protein synthesis

  • Regulatory feedback loop: GADD34 expression is induced by ATF4 and CHOP, which are themselves upregulated during eIF2α phosphorylation, creating a negative feedback loop to restore protein synthesis

This intricate relationship between PPP1CA and its PPP1R15 regulatory partners enables precise temporal control of eIF2α phosphorylation status, allowing cells to modulate protein synthesis in response to various stress conditions and ultimately determining cell fate decisions between adaptation and apoptosis.

How does PPP1CA activity influence the Unfolded Protein Response (UPR)?

PPP1CA plays a critical role in regulating the Unfolded Protein Response (UPR), particularly through its involvement in the PERK pathway. The UPR consists of three main signaling branches (IRE1-XBP1, ATF6, and PERK-eIF2α), all of which respond to endoplasmic reticulum (ER) stress caused by the accumulation of misfolded proteins .

PPP1CA's influence on the UPR includes:

The timing and extent of PPP1CA activity therefore serve as critical determinants in the UPR, balancing the need to restore protein synthesis against the risk of premature translation resumption during unresolved ER stress, which could lead to the production of pro-apoptotic factors and cell death .

What are the most effective approaches for studying PPP1CA-specific functions versus general PP1 activities?

Studying PPP1CA-specific functions presents unique challenges due to the high sequence similarity between PP1 isoforms and their overlapping activities. Researchers can employ several strategies to distinguish PPP1CA-specific functions from general PP1 activities:

  • Isoform-specific genetic manipulation:

    • CRISPR/Cas9-mediated knockout or knockin of PPP1CA with minimal off-target effects

    • Isoform-specific siRNA or shRNA with carefully validated specificity

    • Conditional knockout models (e.g., floxed PPP1CA alleles with tissue-specific Cre expression)

    • Rescue experiments with wild-type PPP1CA to confirm specificity of observed phenotypes

  • Protein interaction studies:

    • Immunoprecipitation with validated PPP1CA-specific antibodies

    • Proximity labeling approaches (BioID, APEX) with PPP1CA as the bait protein

    • Yeast two-hybrid or mammalian two-hybrid screening with PPP1CA-specific constructs

    • Peptide arrays to identify isoform-specific binding partners

  • Structural biology approaches:

    • X-ray crystallography or cryo-EM of PPP1CA with various binding partners

    • Hydrogen-deuterium exchange mass spectrometry to identify interaction surfaces

    • NMR studies of PPP1CA-specific protein interactions

  • Phosphatase activity assays:

    • In vitro phosphatase assays using purified PPP1CA versus other isoforms

    • Phospho-specific antibodies to monitor dephosphorylation of candidate substrates

    • Phosphoproteomics following PPP1CA manipulation versus other PP1 isoforms

    • Development of isoform-specific inhibitors or activity-based probes

  • Subcellular localization studies:

    • Immunofluorescence with isoform-specific antibodies

    • Live-cell imaging with fluorescently-tagged PPP1CA constructs

    • Subcellular fractionation followed by western blotting

    • Super-resolution microscopy to detect subtle differences in localization patterns

These methodological approaches, when carefully implemented with appropriate controls, can help researchers distinguish isoform-specific functions and advance our understanding of PPP1CA's unique roles in cellular physiology and pathology.

What experimental systems are optimal for investigating PPP1CA interactions with the PPP1R15 family?

Investigating the interactions between PPP1CA and PPP1R15 family members (GADD34 and CReP) requires careful selection of experimental systems that recapitulate physiologically relevant conditions. The following systems are particularly valuable for such studies:

  • Cell-based stress response models:

    • ER stress models using tunicamycin, thapsigargin, or DTT treatment

    • Integrated stress response activation via amino acid deprivation, oxidative stress, or viral infection

    • Time-course experiments capturing both early (0-1h) and late (4-24h) stress responses

    • Cell types with physiologically relevant stress sensitivity (e.g., pancreatic β cells, neurons)

  • Biochemical interaction assays:

    • Co-immunoprecipitation of endogenous complexes under various stress conditions

    • GST pulldown assays with recombinant proteins to map interaction domains

    • Surface plasmon resonance or isothermal titration calorimetry for binding kinetics

    • Size exclusion chromatography to analyze complex formation

    • Cross-linking mass spectrometry to identify interaction interfaces

  • Structural biology approaches:

    • X-ray crystallography of PPP1CA-GADD34/CReP complexes

    • Cryo-EM analysis of holophosphatase complexes with substrate (eIF2α)

    • NMR studies of dynamic interactions

    • Hydrogen-deuterium exchange mass spectrometry

  • Live-cell imaging techniques:

    • FRET/BRET biosensors to monitor real-time interactions

    • BiFC (Bimolecular Fluorescence Complementation) for visualizing complex formation

    • Fluorescence correlation spectroscopy to measure complex dynamics

    • Optogenetic approaches to temporally control complex formation

  • Genetic models:

    • CRISPR/Cas9-engineered cell lines with tagged endogenous proteins

    • Conditional knockout mouse models for PPP1CA or PPP1R15 family members

    • Domain mutation knock-in models to disrupt specific interactions

    • Patient-derived cells carrying mutations in PPP1R15A/B

  • Pharmacological approaches:

    • Using selective inhibitors of GADD34-PP1 interactions (e.g., Sephin1, Guanabenz)

    • Small molecule modulators of stress pathways to examine context-dependent interactions

    • Chemical genetics approaches with engineered PPP1CA variants

When selecting experimental systems, researchers should consider the temporal dynamics of the stress response, as the PPP1CA-GADD34 interaction is highly regulated and changes significantly between early and late stress phases. Additionally, the constitutive nature of CReP expression versus the stress-inducible nature of GADD34 requires experimental designs that can distinguish between these different regulatory mechanisms.

What are the technical challenges in measuring PPP1CA-specific phosphatase activity?

Measuring PPP1CA-specific phosphatase activity presents several technical challenges that researchers must address to obtain reliable and physiologically relevant results:

  • Isoform specificity challenges:

    • High sequence similarity between PP1 isoforms (PPP1CA, PPP1CB, PPP1CC)

    • Limited availability of truly isoform-specific antibodies or inhibitors

    • Overlapping substrate preferences among isoforms

  • Holoenzyme complexity issues:

    • PPP1CA rarely functions alone but as part of diverse holoenzyme complexes

    • Regulatory subunits dramatically alter substrate specificity and activity

    • In vitro assays with purified catalytic subunit may not reflect physiological activity

  • Technical assay limitations:

    • Traditional colorimetric assays (e.g., pNPP) lack substrate specificity

    • Phosphopeptide-based assays may not recapitulate native protein substrate conformations

    • Maintaining enzyme stability and activity during purification

    • Background phosphatase activity from contaminating enzymes

  • Methodological solutions:

    • Immunodepletion approaches: Sequential immunoprecipitation to remove specific PP1 isoforms

    • Engineered sensitivity: Creating analog-sensitive PPP1CA variants that can be selectively inhibited

    • Holoenzyme reconstitution: Assembling defined complexes with specific regulatory subunits

    • Substrate trapping: Using catalytically inactive mutants to capture physiological substrates

    • FRET-based sensors: Developing specific sensors for real-time activity monitoring

    • Mass spectrometry: Quantitative phosphoproteomics following acute PPP1CA manipulation

    • Activity-based probes: Development of chemical tools to label active PPP1CA

  • Experimental design considerations:

    • Careful selection of buffer conditions that maintain physiological regulation

    • Use of metal cofactors that reflect cellular conditions (Mn²⁺/Fe²⁺)

    • Inclusion of appropriate inhibitor controls (e.g., okadaic acid, calyculin A)

    • Analysis of activity across subcellular fractions to account for localization

    • Time-resolved measurements to capture dynamic regulation

  • Validation strategies:

    • Parallel genetic approaches (siRNA, CRISPR) to confirm specificity

    • Rescue experiments with wild-type versus catalytically inactive PPP1CA

    • Comparison of results across multiple assay platforms

    • Correlation of in vitro findings with cellular phenotypes

By addressing these challenges through careful experimental design and validation, researchers can more accurately measure PPP1CA-specific phosphatase activity and gain insights into its distinctive roles in cellular signaling pathways.

How is PPP1CA dysregulation implicated in neurodegenerative diseases?

PPP1CA dysregulation has emerged as a significant factor in various neurodegenerative disorders, particularly through its role in regulating the integrated stress response (ISR) and unfolded protein response (UPR). The relationship between PPP1CA activity and neurodegeneration is complex and involves several mechanisms:

  • Altered eIF2α phosphorylation dynamics:

    • Persistent eIF2α phosphorylation can lead to sustained translational attenuation

    • Conversely, premature dephosphorylation may allow accumulation of misfolded proteins

    • Both scenarios can be detrimental to neuronal health and function

  • Disease-specific implications:

    • Alzheimer's disease: Dysregulated PPP1CA activity affects tau phosphorylation and amyloid-β processing

    • Parkinson's disease: ER stress and impaired UPR are linked to α-synuclein aggregation

    • Amyotrophic Lateral Sclerosis (ALS): TDP-43 aggregates trigger ER stress and ISR activation

    • Prion diseases: PrP^Sc accumulation activates PERK and sustained eIF2α phosphorylation

  • Therapeutic targeting:

    • GADD34-PPP1CA inhibitors (like Sephin1, Guanabenz) have shown neuroprotective effects in various models

    • These compounds delay UPR progression by maintaining eIF2α phosphorylation, which:

      • Reduces the translation of proapoptotic factors like CHOP

      • Extends the prosurvival phase of the UPR

      • Provides additional time for clearance of misfolded proteins

  • Cellular stress integration:

    • Neurons are particularly vulnerable to protein homeostasis disruptions

    • PPP1CA activity influences multiple stress response pathways relevant to neurodegeneration:

      • Oxidative stress

      • Mitochondrial dysfunction

      • Inflammatory responses

      • Autophagy regulation

  • Age-related vulnerabilities:

    • Aging neurons show decreased capacity to handle proteostatic stress

    • PPP1CA-mediated stress response modulation becomes increasingly critical with age

    • Chronic low-level stress may gradually overwhelm compensatory mechanisms

Understanding the precise role of PPP1CA in different neurodegenerative contexts requires further investigation but represents a promising avenue for therapeutic development. The temporal dynamics of PPP1CA activity appear particularly important, with the potential for both beneficial and detrimental effects depending on the specific disease context and stage.

What role does PPP1CA play in cancer development and progression?

PPP1CA's involvement in cancer biology is multifaceted, affecting key processes in tumor development, progression, and response to therapy:

  • Cell cycle regulation:

    • PPP1CA dephosphorylates critical cell cycle regulators including Rb, p53, and CDC25

    • Dysregulation can lead to aberrant cell cycle progression and genomic instability

    • Altered PPP1CA activity or expression has been observed in various cancer types

  • Apoptosis and survival signaling:

    • Through its role in the integrated stress response (ISR), PPP1CA influences the balance between pro-survival and pro-apoptotic signaling

    • Cancer cells often exhibit enhanced stress tolerance, potentially through altered PPP1CA function

    • Modulation of PPP1CA activity can influence tumor cell sensitivity to therapy-induced stress

  • Cancer-specific expression patterns:

    • Altered expression of PPP1CA has been reported in multiple cancer types

    • Changes in PPP1CA regulatory subunit expression can redirect phosphatase activity in cancer cells

    • The PPP1R15A/GADD34 regulatory subunit in particular shows complex roles in tumorigenesis

  • Invasion and metastasis:

    • PPP1CA regulates cytoskeletal dynamics through dephosphorylation of key substrates

    • Changes in PPP1CA activity can influence cell adhesion, migration, and invasive potential

    • Altered expression of PPP1CA or its regulatory partners may contribute to metastatic phenotypes

  • Therapeutic implications:

    • Modulating PPP1CA activity represents a potential therapeutic strategy

    • Cancer-specific vulnerabilities may exist due to heightened dependence on stress response pathways

    • Combination approaches targeting PPP1CA activity alongside conventional therapies show promise

  • Biomarker potential:

    • PPP1CA expression or activity patterns may serve as prognostic or predictive biomarkers

    • Phosphorylation status of PPP1CA substrates could indicate pathway activity

    • Analysis of PPP1CA regulatory networks may reveal patient-specific vulnerabilities

The contextual nature of PPP1CA function makes its role in cancer highly dependent on tissue type, genetic background, and tumor microenvironment. Further research is needed to fully elucidate the specific contexts in which PPP1CA acts as a tumor suppressor versus an oncogenic factor.

How can PPP1CA activity be targeted therapeutically?

Targeting PPP1CA activity for therapeutic purposes presents both opportunities and challenges due to its essential cellular functions and involvement in multiple signaling pathways. Several approaches have shown promise:

  • Targeting PPP1CA-regulatory subunit interactions:

    • Small molecules that disrupt specific PPP1CA-regulatory subunit interactions

    • Examples include Sephin1 and Guanabenz, which interfere with GADD34-PPP1CA interaction

    • These compounds have shown therapeutic potential in neurodegenerative disease models by modulating the integrated stress response

    • Advantage: Greater specificity than targeting the catalytic site directly

  • Substrate-specific approaches:

    • Design of molecules that block PPP1CA access to specific substrates

    • Development of substrate-mimetic peptides or peptidomimetics

    • Creation of bifunctional molecules that redirect PPP1CA to alternative substrates

  • Allosteric modulation:

    • Identification of allosteric sites unique to PPP1CA

    • Development of compounds that bind these sites to alter activity or substrate specificity

    • Screening for natural products with isoform-selective effects

  • Expression modulation:

    • RNA interference approaches targeting PPP1CA specifically

    • Antisense oligonucleotides to modify PPP1CA expression

    • Transcriptional regulators of PPP1CA expression

  • Localization-based strategies:

    • Targeting PPP1CA subcellular localization mechanisms

    • Disrupting or enhancing interaction with anchoring proteins

    • Compartment-specific activation or inhibition

  • Context-specific targeting:

    • Exploitation of disease-specific vulnerabilities

    • Stress-activated pro-drug approaches

    • Combination therapies targeting parallel pathways

  • Emerging therapeutic applications:

    • Neurodegeneration: GADD34-PPP1CA inhibitors have shown promise in models of ALS, prion disease, and other neurodegenerative conditions by extending the protective phase of the UPR

    • Cancer: Context-dependent approaches based on specific tumor vulnerabilities

    • Metabolic disorders: Targeting PPP1CA roles in insulin signaling and glucose metabolism

    • Inflammatory conditions: Modulating PPP1CA influence on inflammatory signaling pathways

Given the essential nature of PPP1CA functions, therapeutic approaches must maintain a careful balance between efficacy and toxicity. The most promising strategies typically focus on modulating specific PPP1CA functions rather than complete inhibition of all activities, often by targeting regulatory subunit interactions or specific subcellular pools of PPP1CA.

What mechanisms control PPP1CA expression and localization in different cellular contexts?

PPP1CA expression and localization are regulated through multiple sophisticated mechanisms that ensure appropriate phosphatase activity in different cellular contexts:

  • Transcriptional regulation:

    • Tissue-specific transcription factors influencing PPP1CA expression

    • Stress-responsive elements in the PPP1CA promoter

    • Epigenetic modifications affecting chromatin accessibility

    • microRNA-mediated regulation of PPP1CA mRNA stability and translation

  • Post-translational modifications of PPP1CA:

    • Phosphorylation at specific residues affecting catalytic activity

    • Methylation influencing binding to certain regulatory partners

    • Oxidation of catalytic site residues as a redox-sensitive regulatory mechanism

    • Ubiquitination controlling protein turnover and degradation

  • Subcellular targeting mechanisms:

    • Interaction with targeting regulatory subunits containing localization signals

    • The PPP1R15 family (GADD34 and CReP) directs PPP1CA to the endoplasmic reticulum

    • Nuclear targeting through specific nuclear regulatory subunits

    • Cytoskeletal anchoring via actin or myosin-binding regulatory partners

    • Membrane association through lipid-binding domains of certain regulatory subunits

  • Context-dependent complex formation:

    • Competitive binding between different regulatory partners

    • Stress-induced changes in regulatory subunit availability

    • Cell cycle-dependent interactions

    • Developmental stage-specific regulatory partners

  • Regulation by inhibitory proteins:

    • Endogenous inhibitors like inhibitor-1, DARPP-32, and inhibitor-2

    • Stimulus-dependent activation/inactivation of these inhibitors

    • Compartment-specific inhibition patterns

  • Turnover and stability regulation:

    • Proteasomal degradation pathways

    • Chaperone interactions affecting stability

    • Autophagy-mediated turnover

    • Stress-induced stability changes

  • Interactome dynamics:

    • Changes in binding partner availability during development

    • Stress-induced remodeling of PPP1CA complexes

    • Disease-associated alterations in the PPP1CA interactome

How do post-translational modifications affect PPP1CA function?

Post-translational modifications (PTMs) of PPP1CA serve as critical mechanisms for fine-tuning its enzymatic activity, substrate specificity, localization, and interactions with regulatory partners. These modifications create an additional layer of regulation beyond the effects of regulatory subunits:

  • Phosphorylation:

    • Thr320 phosphorylation by CDK kinases inhibits PPP1CA activity during mitosis

    • Tyr phosphorylation can affect interaction with certain regulatory subunits

    • Sequential phosphorylation events can create switch-like behavior in PPP1CA activity

    • Phosphorylation may induce conformational changes affecting catalytic site accessibility

  • Methylation:

    • Methylation of the C-terminal leucine (Leu309) affects interaction with certain regulatory subunits

    • Carboxymethylation is catalyzed by leucine carboxyl methyltransferase 1 (LCMT1)

    • Demethylation by protein methylesterase 1 (PME1) creates dynamic regulation

    • Methylation status influences formation of specific holoenzyme complexes

  • Oxidation:

    • The catalytic site contains metal ions (Mn²⁺/Fe²⁺) susceptible to oxidation

    • Oxidative stress can temporarily inactivate PPP1CA through metal center oxidation

    • Redox-sensitive cysteine residues may act as cellular redox sensors

    • Reversible oxidation provides a mechanism linking oxidative stress to phosphatase inhibition

  • Ubiquitination and SUMOylation:

    • Polyubiquitination targets PPP1CA for proteasomal degradation

    • SUMOylation may affect nuclear localization and function

    • Deubiquitinating enzymes provide additional regulatory control

    • Stress conditions can alter ubiquitination patterns

  • PTM crosstalk:

    • Hierarchical modifications where one PTM influences the occurrence of others

    • Combinatorial PTM patterns creating specific "molecular barcodes"

    • Competition between different modifications for the same residues

    • Integration of multiple signaling inputs through different PTMs

  • PTM-dependent interactions:

    • Some regulatory subunits preferentially interact with modified or unmodified PPP1CA

    • PTMs can create or destroy binding interfaces for regulatory proteins

    • Modification-dependent interactions with chaperones affecting stability

  • Temporal dynamics:

    • Rapid and reversible modifications (like phosphorylation) for acute regulation

    • More stable modifications (like methylation) for sustained functional changes

    • Cell cycle-dependent modification patterns

    • Stress-responsive modification programs

Dysregulation of these PTMs can contribute to disease states by altering PPP1CA function. Therapeutic approaches targeting specific PPP1CA modifications or the enzymes responsible for them represent an emerging area of interest, potentially offering greater specificity than direct catalytic site inhibitors.

What are the emerging research directions in PPP1CA biology?

The field of PPP1CA research is rapidly evolving, with several emerging directions that promise to expand our understanding of this essential phosphatase:

  • Single-cell analysis of PPP1CA function:

    • Single-cell phosphoproteomics to capture cell-to-cell variability in PPP1CA substrates

    • Single-cell transcriptomics to identify context-specific regulatory networks

    • Spatial proteomics to map subcellular PPP1CA activity patterns

    • Understanding how PPP1CA contributes to cellular heterogeneity in tissues

  • Advanced structural biology approaches:

    • Cryo-EM studies of diverse PPP1CA holoenzyme complexes

    • Hydrogen-deuterium exchange mass spectrometry to capture dynamic conformational changes

    • Integrative structural biology combining multiple techniques

    • Computational modeling of allostery and regulatory mechanisms

  • Systems biology perspectives:

    • Network modeling of PPP1CA-centered signaling hubs

    • Multi-omics integration to understand PPP1CA's global impact

    • Quantitative models of phosphorylation/dephosphorylation dynamics

    • Machine learning approaches to predict context-specific PPP1CA functions

  • Novel therapeutic strategies:

    • Development of holoenzyme-specific modulators

    • Targeted protein degradation approaches (PROTACs) for specific PPP1CA complexes

    • Gene therapy approaches for PPP1CA-related disorders

    • Combination therapies targeting stress response networks

  • Roles in non-canonical pathways:

    • PPP1CA functions beyond phosphatase activity (scaffolding, sequestration)

    • Non-protein substrates and novel enzymatic activities

    • Roles in RNA metabolism and regulation

    • Functions in extracellular vesicles and intercellular communication

  • Developmental and evolutionary perspectives:

    • Tissue-specific functions during development

    • Evolutionary conservation and divergence of PPP1CA regulatory mechanisms

    • Comparative analysis across model organisms

    • Ancient origins of phosphatase regulation

  • Innovative methodological approaches:

    • Optogenetic and chemogenetic tools to control PPP1CA activity with spatial and temporal precision

    • Biosensors for real-time monitoring of PPP1CA activity in living cells

    • Genome-wide CRISPR screens for synthetic interactions

    • Proximity labeling approaches to map dynamic interactomes

  • Translational research directions:

    • Biomarker development based on PPP1CA activity signatures

    • Personalized medicine approaches targeting specific PPP1CA vulnerabilities

    • Expanded therapeutic applications beyond neurodegeneration

    • Combination approaches with stress pathway modulators

These emerging directions reflect the central importance of PPP1CA in cellular physiology and the increasing recognition of its roles in health and disease. Advances in technology and interdisciplinary approaches are driving rapid progress in our understanding of this fascinating phosphatase system.

Product Science Overview

Gene and Protein Structure

The PPP1CA gene is located on chromosome 11 in humans . It encodes the alpha isoform of the catalytic subunit of PP1, which is one of the three catalytic subunits of the enzyme. The protein is broadly expressed and associates with over 200 regulatory proteins to form holoenzymes, which dephosphorylate their biological targets with high specificity .

Function and Biological Significance

PP1 is involved in the regulation of several critical cellular processes, including:

  • Cell Division: PP1 plays a role in the progression of the cell cycle and mitosis.
  • Glycogen Metabolism: It regulates glycogen synthesis and breakdown.
  • Muscle Contractility: PP1 is involved in the regulation of muscle contraction.
  • Protein Synthesis: It influences the translation of proteins.
  • HIV-1 Viral Transcription: PP1 is implicated in the transcription of HIV-1 .

Increased activity of PP1 has been observed in the end stage of heart failure, suggesting its importance in cardiac function. Deregulation of PP1 is also implicated in diabetes and multiple types of cancer .

Recombinant Protein

The recombinant form of Protein Phosphatase 1, Catalytic Subunit Alpha, is produced using recombinant DNA technology. This involves cloning the PPP1CA gene into an expression vector, which is then introduced into a host cell (such as E. coli) to produce the protein. The recombinant protein is then purified for use in research and therapeutic applications .

Clinical and Research Applications

Due to its involvement in various cellular processes, PP1 is a significant target for research in fields such as cancer, diabetes, and cardiovascular diseases. Understanding the regulation and function of PP1 can lead to the development of new therapeutic strategies for these conditions .

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.