PGP Human, Active

Phosphoglycolate Phosphatase Human Recombinant, Active
Shipped with Ice Packs
In Stock

Description

PGP Human Recombinant produced in E.coli is a single, non-glycosylated polypeptide chain containing 345 amino acids (1-321a.a) and having a molecular mass of 36.5kDa.
PGP is fused to a 24 amino acid His-tag at N-terminus & purified by proprietary chromatographic techniques.

Product Specs

Introduction
Phosphoglycolate phosphatase (PGP) is found in all tissues, including red blood cells, lymphocytes, and cultured fibroblasts (at the protein level). It exhibits the highest activity in skeletal and cardiac muscle. PGP catalyzes the conversion of 2-phosphoglycolate and water into glycolate and phosphate. Notably, PGP has been associated with diseases such as tardive dyskinesia and polycystic kidney disease.
Description
Recombinant human PGP, expressed in E. coli, is a single, non-glycosylated polypeptide chain comprising 345 amino acids (1-321a.a) with a molecular weight of 36.5 kDa. It includes a 24-amino acid His-tag at the N-terminus and is purified using proprietary chromatographic techniques.
Physical Appearance
A clear, sterile-filtered solution.
Formulation
The PGP protein solution is provided at a concentration of 0.5 mg/ml in a buffer consisting of 20 mM Tris-HCl (pH 8.0), 0.15 M NaCl, 10% glycerol, and 1 mM DTT.
Stability
For short-term storage (2-4 weeks), the product should be kept at 4°C. For extended periods, store frozen at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Repeated freezing and thawing should be avoided.
Purity
The purity is determined to be greater than 95% by SDS-PAGE analysis.
Biological Activity
The specific activity, exceeding 3,000 units/mg, is defined as the amount of enzyme required to hydrolyze 1.0 nanomole of p-nitrophenyl phosphate (pNPP) per minute at pH 7.5 and a temperature of 37°C.
Synonyms

Glycerol-3-phosphate phosphatase, G3PP, Aspartate-based ubiquitous Mg(2+)-dependent phosphatase, AUM, Phosphoglycolate phosphatase, PGP.

Source
Escherichia Coli.
Amino Acid Sequence

MGSSHHHHHH SSGLVPRGSH MGSHMAAAEA GGDDARCVRL SAERAQALLA DVDTLLFDCD GVLWRGETAV PGAPEALRAL RARGKRLGFI TNNSSKTRAA YAEKLRRLGF GGPAGPGASL EVFGTAYCTA LYLRQRLAGA PAPKAYVLGS PALAAELEAV GVASVGVGPE PLQGEGPGDW LHAPLEPDVR AVVVGFDPHF SYMKLTKALR YLQQPGCLLV GTNMDNRLPL ENGRFIAGTG CLVRAVEMAA QRQADIIGKP SRFIFDCVSQ EYGINPERTV MVGDRLDTDI LLGATCGLKT ILTLTGVSTL GDVKNNQESD CVSKKKMVPD FYVDSIADLL PALQG.

Q&A

What is the structural composition of human PGP protein?

Human PGP (P-glycoprotein) is a transmembrane unidirectional efflux pump that belongs to the ATP-binding cassette (ABC) transporter superfamily. The commercially available recombinant human PGP protein typically consists of 321-345 amino acids with a molecular mass of approximately 36.5 kDa . The protein contains multiple functional domains, including transmembrane domains that form the substrate binding site and nucleotide binding domains (NBDs) responsible for ATP hydrolysis that powers the transport mechanism .

The full-length recombinant protein expressed in Escherichia coli systems typically includes a histidine tag for purification purposes and demonstrates >95% purity when analyzed by SDS-PAGE and mass spectrometry techniques . The amino acid sequence includes specific regions that are crucial for its function as a glycerol-3-phosphate phosphatase.

What are the primary physiological functions of PGP in human cells?

PGP serves multiple crucial physiological functions in human cells:

  • Xenobiotic protection: PGP functions as a protective barrier by actively transporting various substrates including drugs, metabolic products, and xenobiotics across cellular membranes, essentially protecting tissues from potential toxins .

  • Metabolic regulation: As a glycerol-3-phosphate phosphatase, PGP hydrolyzes glycerol-3-phosphate into glycerol, thereby regulating cellular levels of glycerol-3-phosphate, which is a metabolic intermediate in glucose, lipid, and energy metabolism .

  • Barrier function: PGP is an essential component of the blood-brain barrier (BBB), where it is expressed on the apical surface of capillary endothelial cells and restricts the entry of potentially toxic substances into the brain .

  • Immune system modulation: PGP plays crucial roles in the innate immune response, including elevation of type I interferon responses and influencing the activation and migration of dendritic cells from the periphery to lymph nodes .

  • T-cell function: PGP expression has been observed in various T cells including CD4+ T regulatory cells, T effector cells, CD8+ cytotoxic T cells, and natural killer cells, where it contributes to normal development and protection from oxidative stress .

How can researchers accurately measure PGP-mediated efflux activity in experimental settings?

When measuring PGP-mediated efflux activity, researchers should employ a multi-faceted approach:

  • Calcein-AM efflux assay: This is one of the most widely used methods for assessing PGP activity. Calcein-AM is a PGP substrate that becomes fluorescent once cleaved by intracellular esterases. In cells with active PGP, calcein-AM is pumped out before it can be cleaved, resulting in lower fluorescence. This assay can be used to screen potential PGP inhibitors .

  • ATPase activity assays: Since PGP functions as an ATP-dependent pump, measuring its ATPase activity provides direct insight into its transport function. Verapamil-stimulated ATPase activity is commonly used to confirm PGP inhibition by test compounds .

  • Radioactive substrate accumulation: Using radiolabeled PGP substrates (such as [³H]-digoxin or [¹⁴C]-doxorubicin) allows researchers to quantify intracellular drug accumulation in the presence or absence of PGP inhibitors.

  • Flow cytometry: This technique can be used with fluorescent PGP substrates to assess efflux activity at the single-cell level, allowing for the identification of heterogeneous PGP expression within cell populations.

Methodological considerations: When conducting these assays, researchers should include appropriate positive controls (known PGP inhibitors like verapamil or tariquidar) and negative controls. Additionally, using multiple complementary assays provides more robust evidence of PGP inhibition or activity modulation.

What computational approaches are most effective for identifying novel PGP inhibitors?

Advanced computational approaches for identifying novel PGP inhibitors include:

  • Machine learning-guided molecular docking: This combined approach has proven successful in screening large compound libraries. A recent study screened 2.6 billion synthesizable molecules using this method to identify potential inhibitors targeting the nucleotide binding domains (NBDs) of PGP .

  • Molecular dynamics (MD) simulations: These simulations provide valuable insights into the dynamic interactions between PGP and potential inhibitors. MD allows researchers to observe how compounds interact with specific binding sites and how these interactions might affect the conformational changes necessary for PGP function .

  • Structure-based virtual screening: With the availability of high-resolution PGP structures, this approach has become increasingly powerful. It involves docking potential inhibitors to specific binding sites on PGP and scoring them based on predicted binding affinity and interaction patterns.

Methodological workflow for computational screening:

StepApproachOutput
1Initial screeningLarge compound library filtered by physicochemical properties
2Machine learning pre-selectionCompounds ranked by predicted binding affinity
3Molecular dockingTop compounds docked to target site (NBD or DBD)
4Molecular dynamicsAssessment of binding stability over time
5Selection for testingDiverse set of high-scoring compounds

When implementing this approach, researchers should focus on both traditional drug binding domain (DBD) inhibitors and emerging strategies targeting the nucleotide binding domains, as the latter may offer novel mechanisms to overcome PGP-mediated drug resistance .

How do post-translational modifications regulate PGP function and drug efflux activity?

Post-translational modifications (PTMs) critically influence PGP function through multiple mechanisms:

  • Phosphorylation: Several kinases regulate PGP activity through phosphorylation. For example, activation of protein kinase C (PKC) pathways can increase PGP phosphorylation, enhancing its efflux activity. Conversely, inhibition of these pathways can reduce PGP function.

  • Glycosylation: PGP contains N-linked glycosylation sites that affect its proper folding, membrane localization, and stability. Research has shown that altered glycosylation patterns can impact substrate recognition and transport efficiency.

  • Ubiquitination: This modification regulates PGP degradation and turnover. Changes in ubiquitination can affect PGP half-life and consequently its abundance at the cell membrane.

Methodological approaches to study PGP PTMs:

  • Site-directed mutagenesis: Mutating specific residues prone to PTMs allows researchers to assess their functional significance in cell-based assays.

  • Mass spectrometry: This technique enables precise identification and quantification of PTMs on purified PGP.

  • Phospho-specific antibodies: These can be used to detect and quantify phosphorylation at specific sites.

  • Pharmacological modulators: Using kinase inhibitors, glycosylation inhibitors, or proteasome inhibitors to manipulate specific PTM pathways and observe effects on PGP function.

When investigating PTMs, researchers should consider the cellular context, as regulation may differ between tissue types and disease states. Additionally, integrating multiple techniques provides a more comprehensive understanding of how PTMs regulate PGP function.

What is the relationship between PGP expression and immune system function in various disease states?

The relationship between PGP and immune function is complex and context-dependent:

  • Innate immunity: PGP is essential for the type I interferon response to viral infection. Research has shown that PGP expression elevates the type I interferon response, while PGP blockade reduces the expression of interferon-beta (IFN-β) in infected cells .

  • Dendritic cell function: PGP is crucial for the activation and migration of dendritic cells from the periphery to lymph nodes. Inhibition of PGP by compounds like venlafaxine prevents dendritic cell differentiation and diminishes T cell polarization, proliferation, and cytokine production .

  • T cell development and function: PGP is essential for normal T regulatory cell development and protects T helper cells (Th1 and Th17) from bile acid-driven oxidative stress in the small intestine. PGP-expressing CD8+ T cells exhibit greater effector memory phenotypes compared to naïve cells lacking PGP .

  • Autoimmune conditions: In lupus nephritis, increased expression of PGP has been observed in CD4+ cells in peripheral blood and renal tissue. Additionally, PGP knockout animals have shown impaired dendritic cell maturation and decreased T cell stimulation, exhibiting symptoms of experimental autoimmune encephalomyelitis, suggesting a possible role for PGP in multiple sclerosis .

Research methodologies for studying PGP-immune interactions:

  • Flow cytometric analysis: To quantify PGP expression in specific immune cell populations

  • Knockout/knockdown models: To assess the functional consequences of PGP loss in specific immune contexts

  • Pharmacological inhibition: Using selective PGP inhibitors to determine the effects on immune cell function

  • Ex vivo cell culture systems: To study isolated immune cell populations under controlled conditions

Researchers investigating these interactions should employ multiple complementary techniques and consider the specific disease context, as PGP's role may differ significantly between different immune-mediated conditions.

How does PGP contribute to pharmacoresistant epilepsy and what research methods best elucidate this mechanism?

PGP plays a significant role in pharmacoresistant epilepsy through several interconnected mechanisms:

  • Overexpression at the blood-brain barrier: Multiple animal models and human studies have demonstrated that pharmacoresistant epilepsy is associated with overexpression of PGP at the blood-brain barrier (BBB) . This increased expression reduces the brain penetration of anti-epileptic drugs.

  • Seizure-induced upregulation: Recurrent seizure activity leads to additional extracellular glutamate release, which activates NMDA receptors and elevates cyclooxygenase-2 (COX-2) levels . This glutamate-NMDA-COX-2 pathway has been established as a key regulator of PGP expression.

  • COX-2 mediated induction: Elevated COX-2 levels increase PGP expression, resulting in enhanced efflux of anti-epileptic drugs from the brain . This creates a feedback loop where seizures induce PGP expression, which reduces drug efficacy, potentially leading to more seizures.

Optimal research methodologies to investigate PGP in epilepsy:

MethodologyApplicationKey Insights
PET imaging with PGP substratesIn vivo assessment of PGP functionQuantitative measurement of drug transport across the BBB
MicrodialysisDirect measurement of drug concentrationsLocal drug levels in epileptic foci vs. normal brain regions
ImmunohistochemistryTissue-specific PGP expressionRegional differences in PGP upregulation
Pharmacological interventionTesting of pathway inhibitorsEffects of COX-2 inhibitors or NMDA antagonists on PGP expression
Gene expression analysisTranscriptional regulationIdentification of regulatory elements controlling PGP expression

When designing studies to investigate PGP's role in epilepsy, researchers should consider using multiple complementary techniques and incorporate both animal models and, where ethically possible, human tissue samples from pharmacoresistant epilepsy patients. The glutamate-NMDA-COX-2-PGP pathway provides multiple potential intervention points that could be targeted to overcome drug resistance.

What molecular mechanisms link PGP expression to neurodegenerative disorders?

PGP has been implicated in several neurodegenerative disorders through distinct molecular mechanisms:

  • Parkinson's disease (PD): Lower expression levels of PGP due to genetic variants of the ABCB1 gene have been associated with increased risk of Parkinson's disease . PET scans and biochemical studies have demonstrated significant reduction in PGP activity in PD patients compared to controls . This reduced activity may impair the clearance of neurotoxins that contribute to neurodegeneration.

  • Amyotrophic Lateral Sclerosis (ALS): Elevated PGP levels have been identified in ALS . In some forms of ALS, sporadic astrocytes that modulate PGP expression through NMDA receptors have been observed . This dysregulation may contribute to neuroinflammation and disease progression.

  • Blood-brain barrier dysfunction: As a key component of the BBB, alterations in PGP function can affect the brain's protection against neurotoxic compounds. This dysfunction may contribute to the accumulation of toxic substances that promote neurodegeneration.

  • Oxidative stress modulation: Normal-functioning PGP helps ensure cell survival by suppressing oxidative stress and preserving mitochondrial function . Dysregulation of this protective function could contribute to the increased oxidative damage observed in many neurodegenerative conditions.

Research approaches for investigating PGP in neurodegeneration:

  • Genetic association studies: Identifying ABCB1 gene variants associated with disease risk or progression

  • Functional imaging: Using PET with radiolabeled PGP substrates to quantify transport activity in patients

  • Post-mortem tissue analysis: Examining PGP expression and localization in affected brain regions

  • Cell culture models: Studying how disease-relevant stressors affect PGP expression and function in neurons and glial cells

  • Animal models: Using PGP knockout or transgenic models to investigate the consequences of altered PGP function in neurodegeneration

Understanding the bidirectional relationship between PGP and neurodegeneration—where PGP dysfunction may contribute to disease and disease processes may alter PGP function—is crucial for developing potential therapeutic strategies targeting this transport protein.

What strategies can overcome the limitations of traditional PGP inhibitors that target the drug binding domain?

Traditional PGP inhibitors targeting the drug binding domain (DBD) have faced significant challenges in clinical development. Several alternative strategies show promise for overcoming these limitations:

  • Nucleotide binding domain (NBD) inhibitors: Targeting the NBDs represents a novel approach, as demonstrated by recent computational screening of 2.6 billion synthesizable molecules that identified compounds capable of inhibiting PGP-mediated calcein-AM efflux and decreasing verapamil-stimulated ATPase activity . This approach targets the energy source of PGP rather than competing with substrates.

  • Allosteric inhibitors: Compounds that bind to sites distinct from both the DBD and NBDs may alter PGP conformation and function without directly competing with transported drugs. These may offer improved specificity.

  • Drug structure modification: Rather than inhibiting PGP, modifying drugs to reduce their affinity for PGP represents an alternative approach. For instance, adding polar moieties or polar side chains to otherwise hydrophobic compounds could potentially reduce the efflux function of PGP .

  • Repurposing FDA-approved drugs: Many approved medications have been found to modulate PGP function. Repurposing these drugs as PGP inhibitors or adjuvants could accelerate clinical development by avoiding extensive preclinical toxicity testing .

  • Conformation-locking approaches: Strategies that restrict the PGP structure to either inward- or outward-facing conformations could potentially reduce the frequency of substrate transport, providing another mechanism to overcome drug resistance .

  • CRISPR/Cas9-based approaches: Genome editing technologies offer the potential to directly modify ABCB1 expression in specific tissues. Research has shown that CRISPR/Cas9 knockout of ABCB1, the gene encoding PGP, significantly improved sensitivity to rhodamine 123 and doxorubicin in cancer cells .

When evaluating these approaches, researchers should consider not only efficacy in inhibiting PGP but also specificity, potential for adverse effects on physiological PGP functions, and practical aspects of drug delivery in relevant disease contexts.

How can CRISPR/Cas9 genome editing be effectively employed to study PGP function or reverse multidrug resistance?

CRISPR/Cas9 technology offers powerful approaches for both studying PGP function and potentially reversing PGP-mediated multidrug resistance:

  • Knockout studies: Complete knockout of the ABCB1 gene can reveal the physiological roles of PGP and its contribution to drug resistance. As reported, CRISPR/Cas9 knockout of ABCB1 significantly improved sensitivity to rhodamine 123 and doxorubicin with a marked increase in intracellular accumulation of these drugs in cancer cells .

  • Targeted promoter modification: Instead of complete gene knockout, modifying regulatory regions can provide more nuanced control over PGP expression levels, potentially reducing overexpression while maintaining basal physiological function.

  • Single nucleotide modifications: Introducing specific mutations can help researchers understand how genetic variants affect PGP function. This approach can be used to study naturally occurring polymorphisms associated with altered drug response or disease risk.

  • Reporter gene knock-in: Inserting fluorescent reporters under the control of the endogenous ABCB1 promoter enables real-time monitoring of PGP expression in response to various stimuli or therapeutic interventions.

Methodological considerations for CRISPR/Cas9 PGP studies:

  • Guide RNA design: Design multiple guide RNAs targeting different regions of the ABCB1 gene or its regulatory sequences to optimize editing efficiency and specificity.

  • Delivery systems: For in vivo applications, appropriate delivery vectors (viral or non-viral) must be selected based on tissue specificity and efficiency.

  • Off-target analysis: Comprehensive assessment of potential off-target effects is crucial, particularly when targeting a gene involved in multiple physiological processes.

  • Functional validation: Following genetic modification, thorough assessment of PGP expression, localization, and function using techniques such as western blotting, immunofluorescence, and transport assays is essential.

  • Phenotypic characterization: In both in vitro and in vivo models, careful evaluation of physiological consequences beyond drug sensitivity is important to understand PGP's broader roles.

By strategically applying CRISPR/Cas9 technology, researchers can not only advance our understanding of PGP biology but also develop potential therapeutic strategies to overcome multidrug resistance in cancer and other diseases.

What are the current contradictions in PGP research data and how might these be resolved methodologically?

Several contradictions and challenges exist in PGP research that require methodological solutions:

  • Variability in screening results: Different laboratories report conflicting results when screening for PGP substrates and inhibitors . This variability creates setbacks in drug development processes and complicates the interpretation of research findings.

  • Tissue-specific functions: PGP appears to have distinct roles in different tissues, complicating the development of therapeutic approaches that target PGP without disrupting its essential physiological functions.

  • Dual roles in disease: In some conditions (like cancer), PGP overexpression contributes to pathology, while in others (like Parkinson's disease), reduced PGP function is associated with disease risk .

  • Model system limitations: Cell lines commonly used for PGP studies may not fully recapitulate the complex physiological regulation of PGP in vivo.

Methodological approaches to resolve these contradictions:

  • Standardized assay protocols: Developing and adopting consensus guidelines for PGP assays, including standardized cell lines, substrates, and experimental conditions.

  • Multi-assay validation: Employing multiple complementary techniques to characterize PGP substrates and inhibitors, rather than relying on a single assay.

  • Tissue-specific models: Using organoid cultures, tissue slices, or conditional knockout animals to better understand tissue-specific PGP functions.

  • Systems biology approaches: Integrating transcriptomic, proteomic, and metabolomic data to develop comprehensive models of PGP regulation and function across different tissues and disease states.

  • Improved structural understanding: Leveraging recent advances in cryo-electron microscopy to generate more accurate structural models of PGP in different conformational states, improving structure-based drug design.

  • Collaborative research initiatives: Establishing multi-laboratory collaborative efforts to simultaneously test compounds using standardized protocols, reducing inter-laboratory variability.

By addressing these methodological challenges, researchers can build a more coherent understanding of PGP biology and develop more effective therapeutic strategies that target PGP dysfunction in disease while preserving its essential physiological functions.

How are advances in structural biology transforming our understanding of PGP function and inhibitor design?

Recent advances in structural biology have revolutionized PGP research in several key ways:

  • High-resolution structures: The availability of detailed three-dimensional structures of PGP in various conformational states has provided unprecedented insights into how this transporter binds substrates and undergoes conformational changes during the transport cycle.

  • Binding site characterization: Structural studies have revealed the presence of multiple binding sites within the large drug-binding domain (DBD), explaining PGP's remarkable substrate promiscuity and providing opportunities for more targeted inhibitor design .

  • Nucleotide binding domain focus: Structural information about the nucleotide binding domains (NBDs) has enabled novel approaches to inhibitor design that target ATP binding and hydrolysis rather than substrate binding .

  • Conformational dynamics: Combining structural data with molecular dynamics simulations has improved our understanding of how PGP transitions between inward- and outward-facing conformations during the transport cycle, identifying potential points for intervention.

Impact on inhibitor design strategies:

The enhanced structural understanding of PGP has directly informed several promising approaches to inhibitor development:

  • Structure-based virtual screening: High-throughput computational screening of compound libraries based on structural data has identified novel inhibitor candidates targeting specific binding pockets .

  • Fragment-based drug design: Starting with small molecular fragments that bind to specific regions of PGP and optimizing them based on structural information.

  • Allosteric inhibitor development: Identifying binding sites distant from the primary substrate binding region that can be targeted to alter PGP conformational dynamics.

  • Conformational state stabilizers: Designing compounds that preferentially bind to and stabilize specific conformational states, preventing the completion of the transport cycle.

Future advances in time-resolved structural methods, such as time-resolved cryo-EM and X-ray free electron laser crystallography, may further enhance our understanding of PGP dynamics and provide additional insights for inhibitor design. The integration of structural biology with functional assays and computational methods represents a powerful approach for developing the next generation of PGP inhibitors.

Product Science Overview

Discovery and Function

Phosphoglycolate phosphatase was first studied and purified in plants, where it is involved in the photorespiratory pathway. This pathway is essential for photosynthesis, as it helps in the regeneration of 3-phosphoglycerate from 2-phosphoglycolate, which is a byproduct of the carboxylation reaction catalyzed by ribulose-1,5-bisphosphate carboxylase-oxygenase (rubisco) .

In humans, phosphoglycolate phosphatase has been identified in various tissues, including red blood cells, lymphocytes, and cultured fibroblasts. It is most active in skeletal muscle and cardiac muscle . The enzyme’s activity is crucial for the regulation of 2,3-diphosphoglycerate (2,3-DPG), a compound that affects hemoglobin’s oxygen-binding affinity .

Structural Characteristics

The structural characterization of phosphoglycolate phosphatase from Thermoplasma acidophilum revealed that the enzyme exists as a dimer. Each monomer consists of two distinct domains: a smaller cap domain and a larger core domain. The active site of the enzyme is a continuous tunnel lined with acidic residues, which is consistent with other acid phosphatases .

Human Recombinant Phosphoglycolate Phosphatase

Human recombinant phosphoglycolate phosphatase is produced using Escherichia coli expression systems. The recombinant enzyme is a single, non-glycosylated polypeptide chain containing 345 amino acids and has a molecular mass of approximately 36.5 kDa . It is purified using proprietary chromatographic techniques to ensure high purity and activity .

The recombinant enzyme is formulated in a buffer solution containing Tris-HCl, NaCl, glycerol, and dithiothreitol (DTT) to maintain its stability and activity. It is typically stored at 4°C for short-term use or frozen at -20°C for long-term storage .

Applications and Research

Phosphoglycolate phosphatase is used in various research applications, including studies on metabolic pathways, enzyme kinetics, and the regulation of 2,3-DPG in red blood cells. It is also used in the development of therapeutic strategies for diseases associated with phosphoglycolate metabolism, such as tardive dyskinesia and polycystic kidney disease .

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.