PPP1R1A Human

Protein Phosphatase Inhibitor-1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Biochemical Properties of PPP1R1A Human

PPP1R1A is produced as a recombinant protein in Escherichia coli with a C-terminal His-tag. Key characteristics include:

PropertyDetails
Molecular Weight20 kDa
Amino Acid Sequence179 residues (MEQDNSPRKI...VLEHHHHHH)
Purity>90% (SDS-PAGE, RP-HPLC)
Formulation50 mM Tris (pH 8), 0.1 mM PMSF, 1 mM EDTA, 1 mM DTT, 10% glycerol
StabilityStore at -18°C with carrier protein (e.g., 0.1% HSA/BSA); avoid freeze-thaw cycles

Functional Roles of PPP1R1A

PPP1R1A modulates PP1 activity, impacting diverse cellular pathways:

  • Glycogen Metabolism: Acts as an inhibitor of PP1, enabling cAMP-dependent hormonal control of glycogen breakdown .

  • Calcium Signaling: Inactivated by calcineurin (PP2B) during calcium surges, linking it to neuronal and muscle signaling .

  • β-Cell Function:

    • Regulates insulin secretion and glucose uptake in pancreatic β-cells .

    • Silencing PPP1R1A reduces expression of Ins1, Pdx1, and Glut2, impairing β-cell function .

  • Biomarker Potential: Released into plasma during β-cell injury, serving as a real-time indicator of pancreatic damage .

3.1. Biomarker of β-Cell Injury

  • In Vitro Studies: Rat and human islets exposed to cytotoxic agents (e.g., streptozotocin, H₂O₂) showed PPP1R1A discharge proportional to β-cell death .

  • In Vivo Validation: Streptozotocin-treated rats exhibited a plasma PPP1R1A surge (peak at 4 hours) coinciding with insulin discharge and hyperglycemia .

  • Clinical Relevance: Detected in plasma of patients post-islet transplantation, suggesting utility in monitoring graft survival .

3.2. Genetic and Transcriptional Regulation

  • PDX1 Dependency: PDX1 silencing in INS-1 cells downregulates PPP1R1A, indicating transcriptional control .

  • Drug Modulation: Rosiglitazone upregulates PPP1R1A expression, while metformin and insulin show no effect .

3.3. Protein Interaction Network

PPP1R1A interacts with:

  • PP1 Catalytic Subunits (PPP1CC, PPP1CA, PPP1CB) .

  • Calcium-Dependent Enzymes (PPP3CA, PRKCA) .

  • Exocytosis Machinery (e.g., SNAP25, VAMP2) .

Clinical Implications

  • Diagnostic Use: PPP1R1A’s rapid clearance (half-life ~15 minutes) and β-cell selectivity make it a promising biomarker for acute β-cell injury, outperforming GAD65 in sensitivity .

  • Therapeutic Target: Overexpression in human islets enhances insulin secretion and upregulates key proteins (MAFA, GLUT1), suggesting potential for diabetes interventions .

Disease Associations

  • Diabetes: Linked to impaired insulin secretion and glucose homeostasis .

  • Neurological Disorders: Expressed in brain tissues, with potential roles in synaptic plasticity .

Future Directions

  • Assay Development: High-throughput methods are needed to detect PPP1R1A at picomolar sensitivity for clinical applications .

  • Mechanistic Studies: Further exploration of PPP1R1A’s role in autoimmune diabetes and its interplay with PP1 isoforms .

Product Specs

Introduction
PPP1R1A, an inhibitor of protein-phosphatase 1, plays a crucial role in the hormonal regulation of glycogen metabolism. Increased levels of intracellular cAMP, often triggered by hormones, lead to elevated IPP-1 activity. This protein is involved in mediating cAMP's control over proteins not directly phosphorylated by PKA. Following a rise in intracellular calcium, IPP-1 is inactivated by calcineurin (PP2B). IPP-1 possesses multiple domains that target cellular PP1 complexes. Furthermore, PPP1R1A acts as a cellular regulator of eIF2 alpha phosphorylation.
Description
Recombinant human PPP1R1A, expressed in E. coli with a C-terminal His tag, is a single, non-glycosylated polypeptide chain. It comprises 179 amino acids, resulting in a molecular weight of 20 kDa. The purification of PPP1R1A is achieved using proprietary chromatographic techniques.
Physical Appearance
A clear, colorless solution that has been sterilized by filtration.
Formulation
The PPP1R1A solution is formulated in a buffer containing 50mM Tris (pH 8), 0.1mM PMSF, 1mM EDTA, 1mM DTT, and 10% glycerol.
Stability
For short-term storage (2-4 weeks), keep at 4°C. For extended storage, freeze at -20°C. Adding a carrier protein like 0.1% HSA or BSA is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
The purity is determined to be greater than 90.0% using SDS-PAGE analysis.
Synonyms
IPP1, IPP-1, I-1, PPP1R1A, PPI-1, Protein Phosphatase Inhibitor-1, Protein phosphatase 1 regulatory subunit 1A.
Source
Escherichia Coli.
Amino Acid Sequence
MEQDNSPRKI QFTVPLLEPH LDPEAAEQIR RRRPTPATLV LTSDQSSPEI DEDRIPNPHL KSTLAMSPRQ RKKMTRITPT MKELQMMVEH HLGQQQQGEE PEGAAESTGT QESRPPGIPD TEVESRLGTS GTAKKTAECI PKTHERGSKE PSTKEPSTHI PPLDSKGANS VLEHHHHHH.

Q&A

What is PPP1R1A and what is its primary function in human cells?

PPP1R1A (Protein Phosphatase 1 Regulatory Inhibitor Subunit 1A) functions as an inhibitor of protein phosphatase 1 (PP1). It plays a critical role in regulating phosphorylation-dependent cellular processes by inhibiting PP1 activity when phosphorylated at Thr-35. This inhibition is essential for hormonal control of various metabolic pathways, particularly in tissues where cAMP signaling is prominent. PPP1R1A serves as a regulatory node that can impose cAMP control over proteins that are not directly phosphorylated by PKA, effectively amplifying the cellular response to certain hormonal signals .

How is PPP1R1A activity regulated physiologically?

PPP1R1A activity is primarily regulated through phosphorylation and dephosphorylation cycles. Hormones that elevate intracellular cAMP levels increase PPP1R1A activity across many tissues by promoting its phosphorylation at Thr-35 through PKA. Conversely, when intracellular calcium levels rise, calcineurin (also known as PP2B) dephosphorylates and inactivates PPP1R1A. This dual regulation creates a sophisticated control mechanism that integrates both cAMP and calcium signaling pathways . Additionally, transcriptional regulation plays a role, as seen in tissues like pancreatic β-cells where the transcription factor MafA directly regulates PPP1R1A expression .

Which human tissues express significant levels of PPP1R1A?

PPP1R1A shows differential expression across human tissues. Based on data from multiple expression databases, PPP1R1A is expressed in brain tissues (as documented in the Allen Brain Atlas), pancreatic β-cells, cardiac tissue, and kidney tissues . The expression pattern varies significantly between tissues and can change during development or disease states. For instance, the Allen Brain Atlas datasets indicate specific expression patterns in both adult and developing human brain tissues, suggesting tissue-specific roles for this regulatory protein .

What role does PPP1R1A play in pancreatic β-cell function?

PPP1R1A serves as a critical mediator in glucose-stimulated insulin secretion (GSIS) and its amplification through incretin signaling in pancreatic β-cells. Research demonstrates that PPP1R1A expression is regulated by MafA, a central transcription factor for β-cell function. When PPP1R1A expression is reduced, as observed in dysfunctional β-cells lacking MafA, there is impaired GSIS amplification, decreased PKA-target protein phosphorylation, and reduced mitochondrial coupling efficiency. Furthermore, PPP1R1A silencing affects the expression of critical β-cell marker genes including MafA, Pdx1, NeuroD1, and Pax6, suggesting its role in maintaining β-cell identity and function .

How does PPP1R1A connect to type 2 diabetes pathophysiology?

PPP1R1A expression is significantly reduced in type 2 diabetic islets, with a strong positive correlation between PPP1R1A mRNA levels and GLP1-mediated GSIS amplification. This reduction contributes to β-cell dedifferentiation, a characteristic feature of type 2 diabetes. The impaired PPP1R1A function may explain why some type 2 diabetic patients show unresponsiveness to GLP1R-based treatments, as PPP1R1A is required for proper GLP1R-mediated amplification of insulin secretion. This connection provides a molecular explanation for treatment resistance and identifies PPP1R1A as a potential therapeutic target for improving β-cell function in diabetes .

What is the relationship between PPP1R1A and glycogen metabolism?

PPP1R1A plays an important role in the hormonal control of glycogen metabolism. As an inhibitor of protein phosphatase 1, which dephosphorylates enzymes involved in glycogen synthesis and breakdown, PPP1R1A can influence the phosphorylation status of key metabolic enzymes. When hormones elevate intracellular cAMP, increased PPP1R1A activity maintains the phosphorylated (and thus active) state of enzymes that promote glycogen breakdown while keeping glycogen synthesis enzymes phosphorylated and inactive. This regulatory mechanism allows for rapid adaptation of glycogen metabolism in response to hormonal cues, particularly in tissues like muscle and liver where glycogen storage is crucial for energy homeostasis .

What are the most effective methods for studying PPP1R1A phosphorylation status?

When studying PPP1R1A phosphorylation status, researchers should combine multiple complementary approaches. Western blotting with phospho-specific antibodies targeting Thr-35 remains the gold standard for direct quantification. This should be complemented with mass spectrometry for comprehensive phosphosite mapping, particularly when investigating novel regulatory phosphorylation sites. For dynamic studies, researchers can employ phosphorylation-specific FRET sensors or BRET-based approaches to monitor PPP1R1A phosphorylation in real-time within living cells. Additionally, in vitro kinase assays using purified components can determine direct phosphorylation by candidate kinases, while phosphomimetic (T35D) and phospho-deficient (T35A) mutants provide valuable tools for functional studies. When designing experiments, researchers should account for the rapid turnover of the phosphorylated form by including phosphatase inhibitors during sample preparation .

How can researchers effectively silence or overexpress PPP1R1A in cellular models?

For effective manipulation of PPP1R1A expression, researchers should select methods based on their experimental timeline and model system. For transient knockdown, siRNA or shRNA approaches targeting PPP1R1A mRNA have proven effective in β-cell lines, with validation of knockdown efficiency through qPCR and western blot . For stable knockdown or knockout, CRISPR-Cas9 editing offers greater specificity and completeness. When overexpressing PPP1R1A, researchers should consider using inducible expression systems (like Tet-On/Off) to control expression levels and timing, as constitutive overexpression may disrupt cellular phosphorylation networks. Expression constructs should include appropriate tags (His, FLAG, etc.) for purification and detection while ensuring the tag doesn't interfere with protein function. For physiologically relevant studies, researchers should aim to achieve expression levels within the range observed in target tissues and verify functionality through phosphatase inhibition assays .

What cell lines are most appropriate for studying PPP1R1A function in different contexts?

The selection of appropriate cell lines depends on the specific aspect of PPP1R1A function under investigation. For β-cell studies, INS1 (832/13) cells have been successfully used to study PPP1R1A's role in insulin secretion and incretin signaling . For cardiac research, human induced pluripotent stem (iPS) cell-derived cardiomyocytes provide a physiologically relevant model that captures the relationship between PPP1R1A and cardiac hypertrophy . When studying renal functions, kidney-derived cell lines that express the sodium chloride cotransporter (NCC) would be most appropriate given PPP1R1A's role in potassium-regulated NCC dephosphorylation . Researchers should verify endogenous PPP1R1A expression levels in their chosen cell line and consider how well the cell line recapitulates the signaling networks present in the tissue of interest. Primary cells, while more challenging to work with, often provide the most physiologically relevant results, especially when studying tissue-specific regulatory mechanisms .

How can PPP1R1A function be studied in cardiac hypertrophy models?

To study PPP1R1A in cardiac hypertrophy, researchers can utilize human iPS cell-derived cardiomyocytes as demonstrated in previous studies. This model involves culturing cardiomyocytes to confluence, starving them for 48 hours, and then stimulating them with adrenergic agonists (angiotensin II, endothelin-1, isoproterenol) for up to 72 hours. The hypertrophic response can be quantified through multiple methodological approaches: 1) cell surface area measurements, 2) immunofluorescence visualization of hypertrophy markers like C-FOS and C-JUN, and 3) microarray or RNA-seq analysis to identify differentially expressed genes. This model has successfully demonstrated that PPP1R1A is among the genes affected during hypertrophy induction, suggesting its potential role in both pathological and physiological hypertrophy mechanisms .

What approaches can be used to study PPP1R1A's role in β-cell dysfunction in diabetes?

To investigate PPP1R1A's role in β-cell dysfunction, researchers should employ a multi-faceted approach. Primary islets from diabetic and non-diabetic donors can be compared for PPP1R1A expression levels using qPCR and immunoblotting. Functional studies should measure glucose-stimulated insulin secretion (GSIS) and its amplification by GLP1 agonists in models with PPP1R1A knockdown or overexpression. For mechanistic insights, researchers should assess PKA-target protein phosphorylation status, mitochondrial coupling efficiency using respirometry, and expression of β-cell identity genes (MafA, Pdx1, NeuroD1, Pax6). Single-cell transcriptomics can reveal heterogeneity in PPP1R1A expression across β-cell populations. Additionally, co-immunoprecipitation studies can identify PPP1R1A interaction partners in normal versus diabetic conditions, providing insights into how its regulatory network changes during disease progression .

How does dietary potassium modulation affect PPP1R1A expression in kidney models?

Dietary potassium modulation significantly impacts PPP1R1A expression in kidney tissues, creating an important experimental paradigm for renal physiology research. PPP1R1A has been identified as a potassium-suppressed gene in the kidney, meaning its expression decreases when dietary potassium is elevated. This regulation has functional consequences for sodium chloride cotransporter (NCC) activity, which is critical for renal electrolyte handling. To study this phenomenon, researchers can utilize both in vivo models with controlled dietary potassium manipulation and in vitro models with varied extracellular potassium concentrations. Key experimental approaches include measuring PPP1R1A expression changes (mRNA and protein levels) in response to different potassium diets, assessing its phosphorylation status, and examining functional outcomes on NCC phosphorylation and activity. For mechanistic studies, genetic models that constitutively activate the NCC-regulatory kinase SPAK can help isolate the effects of the PPP1R1A/PP1A pathway from the WNK/SPAK kinase cascade .

How do conflicting results in PPP1R1A studies across different tissues get reconciled?

Reconciling conflicting results in PPP1R1A studies requires a systematic approach that accounts for tissue-specific differences in regulation and function. Researchers should first consider that PPP1R1A may have distinct roles in different cellular contexts—serving as a crucial regulator of insulin secretion in β-cells , contributing to cardiac hypertrophy pathways , and participating in renal electrolyte handling . To address discrepancies, investigators should:

  • Carefully examine experimental conditions, including cell types, animal models, and methodological approaches

  • Consider tissue-specific expression levels and isoform differences

  • Analyze the complete signaling networks in which PPP1R1A functions in each tissue

  • Evaluate post-translational modifications that may differ between tissues

  • Use tissue-specific conditional knockout models to isolate PPP1R1A functions

Meta-analysis of published data, collaborative cross-laboratory validation studies, and systematic review of methodological differences are essential approaches for reconciling apparently contradictory findings .

What are the challenges in translating PPP1R1A findings from animal models to human applications?

Translating PPP1R1A findings from animal models to human applications presents several significant challenges. First, there may be species-specific differences in PPP1R1A regulation, expression patterns, and interaction partners. Human PPP1R1A may have subtle structural or functional differences from its rodent counterparts, affecting its inhibitory potency or regulation. Second, the complex signaling networks in which PPP1R1A operates may differ between species, particularly in specialized tissues like pancreatic β-cells. Third, human genetic diversity introduces variability not captured in inbred animal models. To address these challenges, researchers should validate findings in human tissues and cell lines whenever possible, utilize humanized animal models, and consider population-specific genetic variations in PPP1R1A. Additionally, combining data from multiple model systems (rodents, human cell lines, primary human tissues) provides stronger translational evidence than relying on a single model system .

How might PPP1R1A function be affected by aging and how should researchers account for this variable?

The impact of aging on PPP1R1A function represents an important but understudied area. Age-related changes in PPP1R1A expression, phosphorylation, or regulatory networks could contribute to age-associated pathologies in metabolism, cardiac function, and kidney physiology. When designing studies investigating PPP1R1A in age-related contexts, researchers should:

  • Include age-matched controls and multiple age groups (young, middle-aged, elderly) in experimental designs

  • Analyze age-dependent changes in PPP1R1A expression across tissues of interest

  • Investigate potential alterations in post-translational modifications and protein stability with aging

  • Assess age-related changes in upstream regulators (like MafA in β-cells) and downstream effectors

  • Consider how aging affects the broader signaling networks in which PPP1R1A operates

Longitudinal studies in animal models and cross-sectional analyses of human samples across different age groups can provide valuable insights into how PPP1R1A function evolves throughout the lifespan and potentially contributes to age-related disease processes .

What are the best approaches for purifying recombinant PPP1R1A for in vitro studies?

Purification of recombinant PPP1R1A for in vitro studies requires careful consideration of expression systems and purification strategies. The most effective approach involves bacterial expression (E. coli) of human full-length PPP1R1A with an appropriate tag (His-tag is commonly used) for affinity purification. The protein should be expressed with >90% purity and validated for proper folding and functionality. Key methodological considerations include:

  • Selection of expression vector with inducible promoter to control expression levels

  • Optimization of induction conditions (temperature, IPTG concentration, duration)

  • Lysis under conditions that preserve protein structure and function

  • Multi-step purification combining affinity chromatography with size exclusion

  • Validation of protein functionality through phosphatase inhibition assays

  • Consideration of whether the purified protein should be pre-phosphorylated at Thr-35 for activity studies

For studies requiring phosphorylated PPP1R1A, in vitro phosphorylation with purified PKA followed by separation of phosphorylated from non-phosphorylated forms is recommended .

How should researchers design experiments to study PPP1R1A interactions with other proteins?

Designing experiments to study PPP1R1A protein interactions requires a multi-technique approach. Co-immunoprecipitation (Co-IP) serves as an initial method to identify interactions in cellular contexts, using either endogenous proteins or tagged overexpression systems. This should be followed by proximity ligation assays to visualize interactions in situ and FRET/BRET approaches for live-cell interaction dynamics. For direct binding studies, researchers should employ surface plasmon resonance (SPR) or isothermal titration calorimetry (ITC) with purified components to determine binding kinetics and affinity constants. Crosslinking mass spectrometry can identify specific interaction interfaces. When studying PPP1R1A interactions with PP1, researchers must consider the phosphorylation state of PPP1R1A at Thr-35, as this dramatically affects binding affinity. For comprehensive interaction mapping, BioID or proximity labeling approaches can identify the complete PPP1R1A interactome under different cellular conditions .

What statistical approaches are most appropriate for analyzing PPP1R1A expression data in clinical samples?

When analyzing PPP1R1A expression data in clinical samples, researchers should employ robust statistical approaches that account for biological variability and potential confounding factors. For comparing expression between groups (e.g., diabetic vs. non-diabetic), appropriate tests include t-tests for normally distributed data or non-parametric alternatives (Mann-Whitney U) when normality cannot be assumed. ANOVA with post-hoc tests should be used for multi-group comparisons. For correlation analyses, as seen in studies examining the relationship between PPP1R1A expression and functional outcomes like GLP1-mediated GSIS amplification, Pearson's or Spearman's correlation coefficients should be calculated based on data distribution .

For clinical datasets, researchers should employ multivariate analyses to adjust for confounding variables such as age, sex, BMI, medication use, and disease duration. Linear or logistic regression models can determine independent associations between PPP1R1A expression and clinical outcomes. For time-course or longitudinal data, mixed-effects models account for repeated measures. Power calculations should be performed a priori to ensure adequate sample sizes for detecting clinically meaningful differences. Lastly, machine learning approaches can identify complex patterns in large datasets, potentially revealing novel insights into how PPP1R1A expression patterns relate to disease phenotypes .

What are the most promising therapeutic strategies targeting PPP1R1A?

The most promising therapeutic strategies targeting PPP1R1A focus on tissue-specific modulation of its activity rather than global inhibition or activation. For diabetes applications, enhancing PPP1R1A expression or activity specifically in β-cells could improve GLP1R-mediated insulin secretion and potentially restore responsiveness to GLP1-based therapies in type 2 diabetic patients. Small molecules that stabilize the phosphorylated form of PPP1R1A or prevent its dephosphorylation by calcineurin represent one approach. For cardiac applications, targeted modulation of PPP1R1A in cardiomyocytes could potentially mitigate pathological hypertrophy while preserving physiological adaptations. Emerging gene therapy approaches using tissue-specific promoters could allow for selective restoration of PPP1R1A expression in tissues where it is downregulated in disease states. When developing such therapies, researchers must carefully consider the broader signaling networks influenced by PPP1R1A, as its inhibition of PP1 affects numerous downstream pathways .

How might single-cell approaches advance our understanding of PPP1R1A function?

Single-cell approaches offer transformative potential for understanding PPP1R1A function across heterogeneous cell populations. Single-cell RNA sequencing can reveal cell-specific expression patterns and co-expression networks, particularly valuable in tissues like pancreatic islets where multiple cell types interact. This approach could identify previously unrecognized cell populations where PPP1R1A plays crucial roles and reveal how its expression varies across different β-cell states. Single-cell proteomics and phosphoproteomics can map how PPP1R1A phosphorylation states and protein levels vary at the individual cell level, potentially uncovering subpopulations with distinct regulatory mechanisms. Live-cell imaging combined with FRET-based sensors for PPP1R1A activity could track real-time dynamics in individual cells responding to stimuli. Additionally, spatial transcriptomics approaches can map PPP1R1A expression within tissue architecture, revealing potential relationships between cellular location and function. These technologies will be particularly valuable for understanding how PPP1R1A dysfunction contributes to diseases characterized by cellular heterogeneity, such as diabetes and heart failure .

Product Science Overview

Discovery and Function

I-1 was the first endogenous molecule identified to specifically inhibit PP-1 . It is activated by cAMP-dependent protein kinase A (PKA), which prevents the dephosphorylation of target proteins by PP-1, thereby amplifying β-adrenoceptor (β-AR) signaling . This signaling pathway is vital for heart function, and dysregulation can lead to heart failure and arrhythmias .

Physiological and Pathological Roles

In physiological conditions, I-1 helps maintain proper cardiac function by regulating the phosphorylation state of various proteins involved in heart muscle contraction and relaxation . However, in pathological conditions such as heart failure and atrial fibrillation, I-1 is often down-regulated and hypo-phosphorylated, leading to impaired cardiac function . Conversely, in some cases of atrial fibrillation, I-1 can become hyperactive, contributing to the disease’s progression .

Therapeutic Potential

Given its critical role in cardiac function, I-1 has been suggested as a potential therapeutic target for heart diseases . Research involving genetic mouse models has provided insights into the therapeutic potential of modulating I-1 activity to treat heart failure and arrhythmias .

Recombinant I-1

Recombinant I-1 is produced using recombinant DNA technology, which involves inserting the gene encoding I-1 into a host organism, such as bacteria or yeast, to produce the protein in large quantities . This recombinant form is used in various research and therapeutic applications, including studies on cardiac function and potential treatments for heart diseases .

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.