PNP Human

Purine Nucleoside Phosphorylase Human Recombinant
Shipped with Ice Packs
In Stock

Description

Role in Immune Regulation and Disease

PNP is essential for lymphocyte function, with deficiencies linked to:

  • T-cell lymphopenia: Impaired dGTP clearance disrupts ribonucleotide reductase, halting DNA synthesis .

  • Neurological complications: PNP-deficient neurons exhibit reduced soma size, mitochondrial dysfunction, and p53-mediated apoptosis .

  • Autoimmune and hyperuricemic disorders: Elevated uric acid levels correlate with gout pathogenesis .

Therapeutic Targeting of PNP

PNP inhibitors are explored for T-cell malignancies, autoimmune diseases, and antimicrobial therapies. Notable developments:

Table 1: Inhibitor Activity Against Human PNP

Compound ClassIC₅₀ (Human PNP)Selectivity NotesSource
Acyclic nucleoside phosphonates19 nM60× selectivity for M. tuberculosis PNP
8-AminoinosineKᵢ = 0.45 μMCytotoxicity in T-leukemia cells (CCRF-CEM)
Immucillin-H (DADMe-ImmH)Kᵢ = 58 pMTransition-state analog with picomolar affinity

Key mechanisms:

  • Transition-state analogs: Immucillins mimic ribocation intermediates, achieving sub-nanomolar inhibition .

  • Fleximer bases: Modified purine analogs disrupt substrate binding via π-π stacking and hydrogen bonding .

Diagnostic and Research Tools

Table 2: PNP Levels in Human Biological Samples

Sample TypeMean PNP Concentration (pg/mL)
Serum1,435.64
Citrate Plasma4,419.64
Heparin Plasma2,483.57
EDTA Plasma1,750.71
Data from ELISA measurements

Emerging Applications and Challenges

  • Adjuvant immunotherapy: Low-dose PNP inhibitors enhance TLR-mediated immune activation .

  • Antimicrobial synergy: Co-administration with nucleoside analogs (e.g., acyclovir) reduces drug metabolism .

  • Blood-brain barrier penetration: Current inhibitors show limited efficacy in neurological PNP deficiency models .

Product Specs

Introduction
Purine nucleoside phosphorylase (PNP) is a key enzyme in purine metabolism, catalyzing the reversible phosphorolysis of ribonucleosides and 2'-deoxyribonucleosides. Its deficiency leads to a buildup of deoxyguanosine, primarily affecting T-cell development and function, resulting in immune deficiency.
Description
Recombinant human PNP, expressed in E. coli, is a 34.2 kDa monomeric protein containing a 20 amino acid His-tag at the N-terminus. This non-glycosylated protein, spanning amino acids 1-289, is purified using proprietary chromatographic techniques.
Physical Appearance
Clear and colorless solution, sterilized by filtration.
Formulation
The provided solution contains PNP at a concentration of 1 mg/ml in a buffer composed of 20mM Tris-HCl (pH 8.0), 0.1M NaCl, 2mM DTT, and 10% glycerol.
Stability
For short-term storage (up to 4 weeks), the PNP solution can be kept at 4°C. For extended storage, it is recommended to freeze the solution at -20°C. The addition of a carrier protein like HSA or BSA (0.1%) is advisable for long-term storage. Repeated freezing and thawing should be avoided.
Purity
The purity of PNP is greater than 90% as determined by SDS-PAGE analysis.
Synonyms
Purine nucleoside phosphorylase, Inosine phosphorylase, NP, PUNP, PRO1837, FLJ94043, FLJ97288, FLJ97312, MGC117396, MGC125915, MGC125916, PNP.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MENGYTYEDY KNTAEWLLSH TKHRPQVAII CGSGLGGLTD KLTQAQIFDY GEIPNFPRST VPGHAGRLVF GFLNGRACVM MQGRFHMYEG YPLWKVTFPV RVFHLLGVDT LVVTNAAGGL NPKFEVGDIM LIRDHINLPG FSGQNPLRGP NDERFGDRFP AMSDAYDRTM RQRALSTWKQ MGEQRELQEG TYVMVAGPSF ETVAECRVLQ KLGADAVGMS TVPEVIVARH CGLRVFGFSL ITNKVIMDYE SLEKANHEEV LAAGKQAAQK LEQFVSILMA SIPLPDKAS.

Q&A

What is the structural composition of human PNP?

Human Purine Nucleoside Phosphorylase is a homotrimer containing three non-conserved tryptophan residues at positions 16, 94, and 178, all located remote from the catalytic site . The trimeric structure is essential for the enzyme's function, with each monomer containing a binding site for substrates. The active site includes key residues such as Glu201, Asn243, and Phe200 that are crucial for substrate binding and catalysis . The three-dimensional structure has been extensively studied through X-ray crystallography, providing valuable insights into the spatial arrangement of the enzyme's components.

To study PNP structure-function relationships, researchers have developed modified versions of the enzyme, such as Tryptophan-free PNP (Leuko-PNP), where the tryptophan residues are replaced with tyrosine. This modification maintains near-normal kinetic properties while providing a valuable research tool for spectroscopic studies . The crystallographic structure of human PNP has been resolved at 2.6Å resolution using synchrotron radiation, revealing specific binding interactions with inhibitors like Immucillin-H .

What role does human PNP play in purine metabolism?

Human PNP catalyzes the reversible phosphorolysis of purine nucleosides, converting them to their respective purine bases and ribose-1-phosphate. This reaction is critical in the purine salvage pathway, which allows the recycling of purine bases rather than their de novo synthesis, a much more energy-intensive process. The enzyme demonstrates specificity for various purine nucleosides including inosine, guanosine, and their deoxyforms .

While some organisms can produce purines through de novo pathways, the salvage pathway is energetically favorable. Interestingly, some parasites like Schistosoma mansoni entirely lack the de novo pathway for purine biosynthesis and depend exclusively on salvage pathways for their purine requirements, making PNP an attractive target for antiparasitic drug design . In human physiology, proper PNP function is essential for normal T-cell development and function, as PNP deficiency leads to impaired T-cell immunity while leaving B-cell function relatively intact.

How do researchers identify and validate potential binding sites in human PNP?

The computational phase involves molecular modeling software such as HyperChem for structural optimizations and quantum chemical analysis. Docking experiments are then performed in a limited area (typically a volume of 12Å × 10Å × 10Å) sufficient to accommodate the ligand inside the receptor . After clustering, the resulting ligand-protein complexes are ranked according to binding energies, with the lowest energy states and specific conservative ligand-protein interactions selected as potentially viable. These interactions are measured using specialized software like BIOVIA Discovery Studio Visualizer.

Validation of predicted binding sites requires experimental approaches including:

  • Site-directed mutagenesis of key residues

  • Enzyme kinetics with wild-type and mutant enzymes

  • Binding studies using isothermal titration calorimetry or surface plasmon resonance

  • Structural confirmation through X-ray crystallography of complexes

What experimental methods are used to assess PNP inhibition?

Assessment of PNP inhibition employs multiple complementary methodologies to provide a comprehensive understanding of inhibitor mechanisms and potency. The primary approach involves enzyme kinetic assays that measure reaction rates in the presence of varying inhibitor concentrations. These assays typically monitor the spectrophotometric changes associated with the phosphorolysis reaction (often at 293 nm) or employ coupled enzyme assays for increased sensitivity .

Researchers determine inhibition constants (Ki) through systematic analysis of reaction velocities at different substrate and inhibitor concentrations, applying appropriate kinetic models (competitive, uncompetitive, or mixed inhibition). For example, with compound inhibitor 4-Amino-5-(1H-pyrrol-3-yl)pyridine, researchers observed a Ki value of 0.70 mM, indicating significant inhibitory potential .

Structural studies complement kinetic analyses, with crystallography providing atomic-level details of inhibitor-enzyme interactions. Temperature-jump perturbation and fluorescence spectroscopy offer insights into binding dynamics and conformational changes. In particular, the use of Tryptophan-free PNP (Leuko-PNP) has enabled precise fluorescence-based binding studies without interference from native tryptophan residues . Pre-steady-state kinetics can further elucidate the mechanism and rate-limiting steps of inhibitor binding and action.

How do the thermodynamics of substrate binding differ between wild-type PNP and engineered variants?

The thermodynamic profiles of substrate binding show notable differences between wild-type human PNP and engineered variants such as Leuko-PNP (Tryptophan-free PNP) and Y249W-Leuko-PNP. In wild-type PNP, binding of substrates like guanine and phosphate occurs in a random order mechanism, but with decreased affinity for the formation of ternary complexes . This negative cooperativity is preserved in Leuko-PNP, suggesting that the tryptophan residues aren't critical for this thermodynamic property.

For researchers investigating new PNP variants, it's essential to characterize both binary (enzyme-substrate) and ternary (enzyme-substrate-phosphate) complexes to fully understand the thermodynamic consequences of protein engineering. The introduction of specific tryptophan residues as catalytic site probes, as in Y249W-Leuko-PNP, provides versatile protein scaffolds for such investigations while maintaining catalytic activity .

What structural differences between human PNP and parasitic PNPs can be exploited for selective drug design?

Comparative structural analysis between human PNP and parasitic PNPs, such as that from Schistosoma mansoni (SmPNP), reveals significant differences that can be exploited for selective drug design. While both enzymes catalyze the same reaction, their active site architectures exhibit distinct features . The atomic coordinates of human PNP complexed with inosine have been used as templates for modeling SmPNP-inosine complexes, highlighting crucial differences in binding affinity and specificity.

The differential affinity for inosine between human and S. mansoni PNPs can be attributed to specific amino acid variations in the binding pocket . These differences create opportunities for designing selective inhibitors that preferentially target the parasitic enzyme while minimizing interactions with human PNP. This selectivity is particularly important since schistosomiasis, caused by S. mansoni, ranks second after malaria in terms of social and economic impact among parasitic diseases .

Successful selective inhibitor design requires:

  • Identification of unique structural features in the parasite PNP binding site

  • Structure-based virtual screening against these specific targets

  • Synthesis and testing of promising candidates

  • Optimization of lead compounds for improved selectivity and drug-like properties

The fact that parasites like S. mansoni lack the de novo pathway for purine biosynthesis and depend entirely on salvage pathways makes PNP an even more attractive target, as inhibition could potentially be lethal to the parasite while merely modulating immune function in humans .

How does tautomerism of purine bases influence the binding and catalytic mechanism of human PNP?

The tautomeric form of purine bases plays a crucial role in determining binding affinity and catalytic efficiency in human PNP. Research using Leuko-PNP has provided significant insights into this phenomenon, particularly regarding guanine binding. Spectroscopic studies, including 13C NMR of guanine bound to Leuko-PNP, combined with fluorescence properties and molecular orbital electronic transition analysis, have established that the fluorescence of bound guanine originates from the lowest singlet excited state of the N1H, 6-keto, N7H guanine tautomer .

This specific tautomeric preference has profound implications for the catalytic mechanism. The ionization state of purine bases affects their hydrogen-bonding capabilities and electronic distribution, directly influencing interactions with key catalytic residues. For instance, spectral changes of Leuko-PNP upon phosphate binding establish that the hydroxyl of Tyr88 is not ionized to the phenolate anion when phosphate is bound, contrary to some mechanistic hypotheses .

Computational studies using density functional theory (DFT) calculations at the B3LYP/6–311++G(d,p) level have further elucidated the Jablonski diagrams of singlet excited-state transitions of different N1-ionization states of N7H guanine, both in gas phase and bound to Leuko-PNP . These detailed electronic analyses provide crucial information for understanding the precise orientation and electronic requirements for optimal substrate binding and catalysis.

What conformational changes occur in human PNP upon substrate binding, and how do they influence catalysis?

A particularly significant conformational change involves a loop region (residues 243–266) near the purine base, which becomes highly ordered upon substrate or inhibitor binding . This loop closure effectively sequesters the active site from bulk solvent, creating the appropriate microenvironment for catalysis. To investigate the role of this catalytic loop, researchers have introduced single tryptophan residues into this region of Leuko-PNP (Y249W-Leuko-PNP), creating a fluorescence probe for monitoring loop dynamics .

The binding of phosphate induces additional conformational adjustments that position the nucleophile optimally for attack on the ribosidic bond. Although Y249W-Leuko-PNP is highly fluorescent and catalytically active, substrate binding did not perturb its fluorescence, suggesting that the introduced tryptophan may not directly report on substrate-induced conformational changes . This highlights the complexity of protein dynamics and the need for multiple complementary approaches to fully characterize the conformational landscape of enzymatic reactions.

What is the structural basis for the high specificity of Immucillin-H for human PNP?

Immucillin-H (ImmH) exhibits remarkably high specificity for human PNP, making it a valuable tool for studying T-cell immune modulation. The crystallographic structure of the human PNP-Immucillin-H complex solved at 2.6Å resolution using synchrotron radiation has provided critical insights into this specificity . This structure represents the first detailed report of human PNP complexed with Immucillin-H, offering valuable information for understanding the molecular basis of its potent inhibition.

The high specificity arises from multiple precise interactions between ImmH and the enzyme's active site. ImmH is a transition state analog that mimics the geometry and charge distribution of the reaction's transition state, explaining its extraordinary binding affinity. Comparison of the human PNP-ImmH complex with other crystallographic structures of human PNP reveals the structural features that contribute to this specificity .

The biological effects of ImmH are significant, as it inhibits the growth of malignant T-cell lines in the presence of deoxyguanosine without affecting non-T-cell tumor lines. It also inhibits activated normal human T cells after antigenic stimulation in vitro . These selective effects on T-cell function while sparing other cell types highlight the potential utility of ImmH and related compounds in treating human diseases characterized by abnormal T-cell growth or activation.

What are the most effective expression systems for producing recombinant human PNP for structural studies?

For high-quality structural studies of human PNP, the selection of an appropriate expression system is critical. Bacterial expression in Escherichia coli remains the most widely used approach due to its simplicity, cost-effectiveness, and high yield. Typically, researchers use BL21(DE3) or similar strains with pET vector systems containing the human PNP gene optimized for bacterial expression . This system allows for inducible expression using IPTG (isopropyl β-D-1-thiogalactopyranoside), with yields often exceeding 50 mg of pure protein per liter of culture.

For studies requiring post-translational modifications or when bacterial expression results in insoluble protein, eukaryotic expression systems may be preferable. Insect cell expression using baculovirus vectors provides a compromise between bacterial simplicity and mammalian authenticity. For absolute fidelity to the native human enzyme, mammalian expression in CHO or HEK293 cells can be employed, though with significantly lower yields.

Purification typically involves a combination of:

  • Affinity chromatography (often using His-tags)

  • Ion-exchange chromatography

  • Size-exclusion chromatography

For crystallographic studies, additional steps to ensure homogeneity and remove the affinity tag may be necessary. The Trp-free variant (Leuko-PNP) has been successfully expressed in bacterial systems with similar yields to wild-type, demonstrating that the tryptophan replacements do not significantly impact protein folding or stability .

How can molecular docking be optimized for predicting binding modes of novel PNP inhibitors?

Optimizing molecular docking for PNP inhibitor prediction requires a multi-faceted approach that builds upon established protocols while addressing the specific challenges of this enzyme system. Based on successful research strategies, the following methodology has proven effective for predicting binding modes of novel PNP inhibitors:

First, a comprehensive analysis of existing crystallographic structures is essential to identify conserved binding interactions. Researchers should examine complexes with natural substrates and known inhibitors from the Protein Database, using specialized tools like ProteinsPlus web service and PoseView to create two-dimensional diagrams of the selected complexes . This foundation provides crucial insights into the most frequent atomic interactions (hydrogen bonds, π–π stacking) that should be preserved in novel inhibitor designs.

For the computational phase, a sequential approach is recommended:

  • Generate initial structures of potential inhibitors using software like HyperChem

  • Perform quantum chemical analysis to determine conformational preferences and electronic properties

  • Execute docking in a defined area (approximately 12Å × 10Å × 10Å) using algorithms that account for both rigid and flexible regions of the protein

  • Cluster resulting complexes and rank them according to binding energies

  • Select states with the lowest energy and specific conservative ligand-protein interactions for further analysis

Importantly, the flexibility of certain inhibitors (like flex-base compounds) must be considered, as they may adopt multiple conformations in the active site. For example, the ability of pyridine and pyrrole fragments to undergo rotation around the C-C bond allows for multiple binding modes, potentially providing several plausible bioactive conformations . This flexibility should be explicitly modeled during the docking process.

What spectroscopic methods are most informative for studying PNP-ligand interactions?

Multiple spectroscopic techniques provide complementary information about PNP-ligand interactions, with each method offering unique insights into binding mechanisms and structural changes. Fluorescence spectroscopy stands out as particularly valuable, especially when using engineered variants like Tryptophan-free PNP (Leuko-PNP) that eliminate background tryptophan signals . This approach has been instrumental in determining the tautomeric form of bound guanine and elucidating binding dynamics.

Nuclear Magnetic Resonance (NMR) spectroscopy provides atomic-level details of ligand binding. The 13C NMR spectrum of guanine bound to Leuko-PNP has helped establish the specific tautomeric form involved in enzyme interactions . For inhibitor design, NMR techniques like STD (Saturation Transfer Difference) can map binding epitopes, identifying which portions of the inhibitor make closest contact with the protein.

Additional valuable spectroscopic methods include:

  • Circular Dichroism (CD): Monitors protein secondary structure changes upon ligand binding

  • Infrared (IR) Spectroscopy: Provides information about hydrogen bonding networks

  • Temperature-jump perturbation combined with fluorescence detection: Reveals kinetic aspects of binding and conformational changes

  • Raman Spectroscopy: Offers vibrational information complementary to IR

For researchers designing new PNP inhibitors, combining multiple spectroscopic techniques with computational methods and X-ray crystallography provides the most comprehensive understanding of ligand-protein interactions.

How can site-directed mutagenesis be used to probe the catalytic mechanism of human PNP?

Site-directed mutagenesis represents a powerful approach for investigating the catalytic mechanism of human PNP by selectively modifying specific residues implicated in substrate binding and catalysis. This methodology allows researchers to test mechanistic hypotheses and establish structure-function relationships with precision.

A systematic approach to site-directed mutagenesis studies of PNP should target:

  • Residues directly involved in substrate binding (e.g., Glu201, Asn243, and Phe200)

  • Catalytic residues proposed to participate in chemical steps (e.g., Tyr88 implicated in phosphate activation)

  • Residues in flexible loops that undergo conformational changes (e.g., residues 243-266)

  • Interface residues important for trimer formation and stability

The creation of Tryptophan-free PNP (Leuko-PNP) exemplifies the successful application of multiple site-directed mutations, where all three non-conserved tryptophan residues were replaced with tyrosine . This variant maintained near-normal kinetic properties while enabling fluorescence-based studies without interference from intrinsic tryptophan signals.

For each mutant, comprehensive characterization should include:

  • Steady-state kinetics to determine changes in Km and kcat values

  • Pre-steady-state kinetics to identify altered steps in the reaction mechanism

  • Binding studies to quantify affinity changes for substrates and inhibitors

  • Structural analysis through crystallography or spectroscopic methods

The Y249W-Leuko-PNP variant demonstrates how strategically placed mutations can create useful spectroscopic probes while maintaining catalytic activity, providing a versatile protein scaffold for further mechanistic investigations .

How do PNP inhibitors affect T-cell function and immune response in various disease models?

PNP inhibitors demonstrate significant effects on T-cell function through a well-defined biochemical mechanism. By inhibiting PNP, these compounds cause accumulation of deoxyguanosine, which is subsequently phosphorylated to deoxyguanosine triphosphate (dGTP). Elevated dGTP levels are particularly toxic to T-cells, leading to selective T-cell depletion while largely sparing other cell types .

Research has shown that Immucillin-H (ImmH) specifically inhibits the growth of malignant T-cell lines in the presence of deoxyguanosine without affecting non-T-cell tumor lines. Furthermore, ImmH inhibits activated normal human T cells after antigenic stimulation in vitro . These selective effects make PNP inhibitors promising candidates for treating diseases characterized by abnormal T-cell proliferation or hyperactivation.

For researchers exploring therapeutic applications, it's important to note that PNP inhibition represents a targeted approach to immune modulation that may be particularly valuable in conditions where more selective intervention is desired than that offered by conventional immunosuppressants.

What are the challenges in developing PNP inhibitors as potential therapeutics for autoimmune disorders?

Developing PNP inhibitors as therapeutics for autoimmune disorders presents several significant challenges despite their promising mechanism. First, achieving sufficient selectivity remains problematic; while human PNP inhibitors like Immucillin-H show excellent enzyme selectivity, translating this to cellular and tissue selectivity in vivo requires extensive optimization. The differential sensitivity of T-cell subpopulations to PNP inhibition means that beneficial regulatory T-cells might be affected alongside pathogenic effector T-cells .

Pharmacokinetic hurdles include designing inhibitors with appropriate drug-like properties. Many potent PNP inhibitors, including nucleoside analogs, have limited oral bioavailability and short half-lives necessitating frequent dosing or alternative delivery strategies. Additionally, as nucleoside analogs, these compounds may interact with other enzymes in nucleoside metabolism, potentially leading to off-target effects.

The complex pathophysiology of autoimmune disorders presents another challenge. These conditions typically involve multiple immune and non-immune mechanisms, making it unlikely that PNP inhibition alone would provide complete disease control. Identifying the specific autoimmune conditions where PNP inhibition would have the greatest impact requires detailed understanding of disease mechanisms and careful patient selection.

Finally, the risk-benefit profile must be carefully assessed. While selective T-cell immunomodulation is less broad than conventional immunosuppression, it may still increase susceptibility to certain infections or impair vaccination responses. Long-term safety studies are essential, particularly given the potential need for chronic administration in autoimmune conditions.

Product Science Overview

Function and Mechanism

PNP catalyzes the reversible reaction where a purine nucleoside and phosphate are converted into a purine and alpha-D-ribose 1-phosphate . This enzyme is part of the glycosyltransferase family, specifically the pentosyltransferases . It is essential in the nucleotide salvage pathways, which allow cells to produce nucleotide monophosphates when the de novo synthesis pathway is interrupted or non-existent .

Clinical Significance

PNP deficiency is a rare autosomal recessive disorder that leads to severe combined immunodeficiency (SCID) and other immune abnormalities . This deficiency was first identified in the 1970s and is characterized by progressive immune abnormalities, neurological issues, and autoimmunity . Early detection and diagnosis are crucial to prevent life-threatening complications .

Recombinant Human PNP

Recombinant human PNP is expressed in Escherichia coli (E. coli) without tags . This recombinant enzyme is used in various biochemical applications, including the detection of inorganic phosphate from biochemical reactions . It is also a target for therapeutic intervention in T-cell immune response .

Applications

The recombinant form of PNP is particularly useful in research and clinical settings. It helps in understanding the enzyme’s role in purine metabolism and its implications in immune response. Additionally, it is used in therapeutic interventions aimed at modulating the immune system .

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.