QPCT Human

Glutaminyl-Peptide Cyclotransferase Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to QPCT Human

QPCT Human (glutaminyl-peptide cyclotransferase) is a zinc-dependent metalloenzyme encoded by the QPCT gene located on chromosome 2 (2p25.3) . It catalyzes the conversion of N-terminal glutaminyl or glutamyl residues in peptides and proteins to pyroglutamyl (pGlu) groups, a post-translational modification critical for peptide stability, receptor binding, and pathogenic amyloid formation .

Molecular Structure and Expression

AttributeDetailsSource
GeneQPCT (2p25.3)
Protein Size~38–39 kDa (Ala33-Leu361 isoform with N-terminal 6-His tag)
Expression HostsSpodoptera frugiperda (Sf9) cells via baculovirus system
Tissue DistributionPituitary, adrenal glands, brain (hippocampus, striatum), and B-lymphocytes
Purity>95% (SDS-PAGE validated)

QPCT is a glycosylated protein with a zinc-binding domain essential for catalytic activity . Its structure includes an N-terminal signal peptide and a C-terminal domain critical for substrate recognition .

Enzymatic Function and Mechanism

QPCT modifies peptides by eliminating ammonia from N-terminal glutaminyl residues, forming pyroglutamyl groups. This reaction:

  • Enhances peptide stability: Resists proteolytic degradation (e.g., amyloid-β peptides in Alzheimer’s disease) .

  • Alters receptor interactions: Modifies neuroendocrine hormones (e.g., thyrotropin-releasing hormone) .

  • Promotes amyloid formation: Facilitates aggregation of neurotoxic peptides linked to neurodegenerative diseases .

Key Substrates

SubstrateApplicationReference
Q-AMC (Bachem I-1175)Fluorescent enzymatic assay for QC activity quantification
[Glu-3]-β-amyloidPyroglutamate formation in Alzheimer’s pathology
PIK3CAStabilizes oncogenic PIK3CA in renal cell carcinoma (RCC)

Disease Associations and Pathological Roles

DiseaseMechanismKey FindingsSource
Alzheimer’s DiseasePyroglutamate formation in amyloid-β peptidesQPCT inhibitors reduce amyloid aggregation (e.g., indazole-based compounds)
Renal Cell CarcinomaStabilizes PIK3CA, promoting sunitinib resistanceHigh QPCT expression correlates with poor therapeutic response in RCC patients
Hepatitis BUpregulates HBV replication and viral protein expressionQPCT serum levels are elevated in HBV-infected patients; QPCT overexpression enhances HBV pgRNA/DNA
Thyroid TumorsDiagnostic marker for thyroid neoplasiaQPCT expression distinguishes thyroid cancers from benign lesions

QPCT Inhibitors

Compound ClassTargetEfficacyStudy
Indazole derivativesQPCT enzymatic activityIC₅₀ values in low nM range; reduces amyloid-β aggregation in vitroVan Manh et al. (2022)
PBD150QC activityReverses pyroglutamate-mediated neurotoxicity in Alzheimer’s modelsBusby et al. (1987)

QPCT in Cancer

  • RCC: QPCT overexpression enhances PIK3CA stability via reduced ubiquitination, driving sunitinib resistance .

  • Melanoma: QPCT promotes tumor aggressiveness through post-translational modifications of oncogenic proteins .

HBV Pathogenesis

  • Mechanism: HBV upregulates QPCT transcription, increasing viral replication and secretion of HBsAg/HBeAg .

  • Biomarker Potential: Elevated serum QPCT levels in HBV-infected patients correlate with active replication .

Applications in Research and Industry

ApplicationTools/MethodsSupplier
Enzymatic AssaysQ-AMC substrate, fluorescent plate readers (e.g., SpectraMax Gemini EM)R&D Systems
Antibody DetectionIHC/Western blotting (e.g., HPA008406, AF6368)Sigma-Aldrich , R&D Systems
Recombinant ProteinBaculovirus-expressed QPCT (His-tagged, >95% purity)Novoprolabs , ProSpec

Industrial Use: Recombinant QPCT is used in drug discovery (e.g., inhibitor screening) and diagnostic assays .

Product Specs

Introduction
Glutaminyl-Peptide Cyclotransferase, also known as QPCT, is an enzyme that plays a crucial role in the biosynthesis of pyroglutamyl peptides. This enzyme exhibits specificity towards acidic and tryptophan residues located adjacent to the N-terminal glutaminyl residue. Notably, the length of the peptide chain beyond the second residue does not significantly impact QPCT activity. In addition to its in vivo function, QPCT can catalyze the formation of pyroglutamate in vitro, including the N-terminal pyroglutamate formation in N-terminally truncated forms of APP amyloid-beta peptides like [Glu-3]-beta-amyloid.
Description
QPCT, produced using Sf9 Baculovirus cells, is a single, glycosylated polypeptide chain with a molecular weight of 38.7kDa. This protein consists of 339 amino acids, spanning from residue 29 to 361. The recombinant QPCT is engineered with a 6-amino acid His tag at its C-terminus. Purification is achieved through proprietary chromatographic techniques. On SDS-PAGE analysis, the molecular size of QPCT is expected to appear between 28-40kDa.
Physical Appearance
A clear, colorless solution that has been sterilized by filtration.
Formulation
The QPCT protein solution is provided at a concentration of 0.5mg/ml. The buffer consists of Phosphate Buffered Saline (pH 7.4) with 10% glycerol.
Stability
For short-term storage (2-4 weeks), the product can be stored at 4°C. For extended storage, it is recommended to freeze the product at -20°C. To ensure optimal stability during long-term storage, adding a carrier protein such as HSA or BSA (0.1%) is advised. It is crucial to avoid repeated freeze-thaw cycles to maintain product integrity.
Purity
The purity of QPCT is determined to be greater than 90% using SDS-PAGE analysis.
Synonyms
Glutaminyl-Peptide Cyclotransferase, Glutaminyl Cyclase, QC, Glutaminyl-TRNA Cyclotransferase, Glutamyl Cyclase, EC 2.3.2.5, SQC, EC, GCT, Glutaminyl-peptide cyclotransferase.
Source
Sf9, Baculovirus cells.
Amino Acid Sequence
VSPSASAWPE EKNYHQPAIL NSSALRQIAE GTSISEMWQN DLQPLLIERY PGSPGSYAAR QHIMQRIQRL QADWVLEIDT FLSQTPYGYR SFSNIISTLN PTAKRHLVLA CHYDSKYFSH WNNRVFVGAT DSAVPCAMML ELARALDKKL LSLKTVSDSK PDLSLQLIFF DGEEAFLHWS PQDSLYGSRH LAAKMASTPH PPGARGTSQL HGMDLLVLLD LIGAPNPTFP NFFPNSARWF ERLQAIEHEL HELGLLKDHS LEGRYFQNYS YGGVIQDDHI PFLRRGVPVL HLIPSPFPEV WHTMDDNEEN LDESTIDNLN KILQVFVLEY LHLHHHHHH.

Q&A

What is human QPCT and what is its primary biological function?

Human glutaminyl cyclase (QPCT) is an enzyme responsible for catalyzing the modification of N-terminal residues of glutamine or glutamate into an N-terminal 5-oxoproline or pyroglutamate (pE) . This post-translational modification occurs on numerous secretory peptides and proteins, altering their stability, bioactivity, and resistance to proteolytic degradation. The enzyme plays important physiological roles in stabilizing hormone and neuropeptide structures, with its activity identified across diverse mammalian tissues including the pituitary, hypothalamus, and peripheral tissues .

Methodologically, researchers investigate QPCT function through a combination of enzymatic assays measuring the conversion of substrates like H-Glu-AMC into pyroGlu-AMC, structural biology approaches examining enzyme-substrate interactions, and functional studies in cellular and animal models . Recent advances in crystallography have provided detailed insights into the QPCT catalytic site, facilitating structure-based drug design approaches.

How does human QPCT differ from QPCT-like (QPCTL) proteins?

Human QPCT and QPCT-like (QPCTL) proteins share 51% sequence identity but exhibit distinct expression patterns and biological roles . The primary differences include:

FeatureHuman QPCTHuman QPCT-like (QPCTL)
Cellular localizationPredominantly secretory pathwayPrimarily Golgi apparatus
Expression profileBroad tissue expression, particularly in brainMore restricted expression pattern
Substrate preferenceHigher activity with glutamine substratesEqual activity with glutamine and glutamate
Inhibitor sensitivityGenerally more sensitive to specific inhibitorsOften requires different inhibitor profiles
Role in diseaseImplicated in Huntington's, Alzheimer'sLess characterized in pathological conditions

When designing experimental approaches, it's critical to use specific siRNAs that do not cross-target both proteins. In the studies examining QPCT effects on Huntington's disease mechanisms, researchers confirmed that their QPCT siRNAs did not target QPCT-like , highlighting the importance of target specificity in mechanistic investigations.

What experimental models are most appropriate for studying human QPCT function?

Selecting appropriate experimental models for human QPCT research requires consideration of the specific research question. The literature demonstrates successful application of several complementary approaches:

  • Cellular models: HEK293/T Rex cells and HeLa cells expressing QPCT have been successfully used to study enzyme function and effects on protein aggregation . These cell lines offer advantages including ease of genetic manipulation, high transfection efficiency, and suitability for high-throughput screening.

  • Primary neurons: Cortical neurons provide a physiologically relevant system for investigating QPCT function in neuronal contexts, particularly important for neurodegenerative disease research .

  • Animal models: Drosophila, zebrafish, and mouse models have demonstrated utility for validating QPCT modulation effects observed in cell systems . These models allow assessment of behavioral and pathological outcomes in complex organismal contexts.

When designing experiments, researchers should consider using multiple models to corroborate findings. For instance, initial discoveries from cell-based screens can be validated in primary neurons and subsequently confirmed in appropriate animal models, as demonstrated in studies of QPCT's role in Huntington's disease .

What are the most reliable methods for measuring human QPCT enzymatic activity?

Several methodological approaches have been validated for measuring human QPCT enzymatic activity, each with distinct advantages:

  • Fluorogenic substrate assay: The conversion of H-Glu-AMC fluorogenic substrate into pyroGlu-AMC provides a sensitive, quantitative measure of QPCT activity . This approach allows for high-throughput screening and kinetic analysis of enzyme function.

  • LC-MS/MS detection: Mass spectrometry-based detection of substrate-to-product conversion offers high specificity for detecting pyroglutamate formation on specific peptide substrates. This method is particularly valuable for confirming activity on novel substrates.

  • Activity-based protein profiling: Using activity-based probes that covalently bind to active QPCT allows visualization and quantification of active enzyme in complex biological samples.

For reliable results, researchers should consider several methodological controls:

  • Include catalytically inactive QPCT mutants (e.g., E201Q) as negative controls

  • Validate findings with both recombinant protein and endogenous enzyme sources

  • Confirm specificity using known QPCT inhibitors

  • Account for pH dependency of enzyme activity

How should researchers design siRNA experiments to specifically target QPCT without affecting QPCT-like?

Designing specific siRNA experiments for QPCT requires careful consideration of sequence homology and validation steps:

  • Sequence selection: Target regions with minimal homology between QPCT and QPCT-like (which share 51% sequence identity) . Utilize specialized siRNA design software that checks for off-target effects.

  • Validation methodology:

    • Confirm knockdown efficiency at both mRNA level (using qPCR with gene-specific primers) and protein level (using specific antibodies)

    • Explicitly verify that QPCT siRNAs do not reduce QPCT-like expression

    • Include multiple independent siRNA sequences targeting different regions of QPCT to rule out off-target effects

  • Experimental controls:

    • Include non-targeting siRNA controls

    • Rescue experiments with siRNA-resistant QPCT expression constructs to confirm specificity

    • When examining phenotypic effects, include parallel experiments with QPCT-like knockdown to distinguish their functions

Research has demonstrated that properly designed QPCT siRNAs can achieve specific knockdown without affecting QPCT-like, enabling clear differentiation of their biological functions as demonstrated in studies examining QPCT's role in Huntington's disease models .

What are the recommended qPCR approaches for quantifying QPCT expression levels in human tissues?

Accurate quantification of QPCT expression using qPCR requires careful attention to experimental design and data analysis:

  • Experimental design considerations:

    • Include both biological replicates (typically three per condition) and technical replicates

    • Properly define experimental factors (e.g., tissue type, treatment conditions)

    • Select appropriate sample size based on expected variability

  • Reference gene selection:

    • Validate stability of candidate reference genes across all experimental conditions

    • Use multiple reference genes rather than relying on a single housekeeping gene

    • Consider geometric averaging of multiple reference genes for more robust normalization

  • Data analysis methodology:

    • Apply appropriate statistical methods based on data distribution and experimental design

    • Consider both parametric and non-parametric tests for differential expression analysis

    • For complex designs with multiple factors, use approaches like two-way ANOVA

  • Quality control steps:

    • Review amplification curves for abnormalities

    • Filter outliers using defined criteria

    • Flag genes with detection issues

    • Validate primers for specificity using melt curve analysis

Following these methodological recommendations ensures reliable quantification of QPCT expression levels across different experimental conditions, providing a foundation for understanding its regulation in various physiological and pathological contexts.

How does human QPCT contribute to protein aggregation in neurodegenerative diseases?

Human QPCT has emerged as a significant modulator of protein aggregation in neurodegenerative conditions through several mechanisms:

  • Modulation of aggregate-prone protein oligomerization: QPCT has been demonstrated to enhance the early stages of mutant huntingtin (HTT) oligomerization, increasing the formation of toxic oligomeric species . This effect appears independent of direct protein-protein interactions, as QPCT does not directly bind HTT .

  • Impact on multiple aggregation-prone proteins: QPCT's role extends beyond HTT, affecting the aggregation of diverse proteins including:

    • Polyglutamine expansion proteins fused to EGFP (Q57, Q81)

    • HTT exon 1 with 74 glutamines fused to HA

    • Polyalanine expansion proteins (expansion of 37 alanines)

  • Chaperone regulation: QPCT inhibition induces elevated levels of the molecular chaperone alpha B-crystallin, which may explain its broad effect on reducing aggregation of multiple protein types . This suggests QPCT influences protein quality control networks rather than directly modifying aggregation-prone proteins.

Importantly, the catalytic activity of QPCT is essential for this aggregation-enhancing effect, as demonstrated by experiments with the catalytically inactive E201Q mutant, which did not increase aggregation . This establishes QPCT enzymatic function as central to its role in protein aggregation dynamics.

What are the current approaches for developing human QPCT inhibitors as potential therapeutics?

The development of human QPCT inhibitors has followed a sophisticated multi-disciplinary approach:

  • Structure-based design strategies:

    • Generation of 3D pharmacophore models using the human QPCT X-ray structure (PDB ID: 2AFW)

    • Ensemble docking methodology to account for flexibility of the QPCT catalytic site

    • Integration of multiple X-ray structures (PDB IDs: 2AFW, 2AFX, 2AFZ, 3PBB, 3SI0) and protein conformations from molecular dynamics simulations

  • Compound selection and optimization:

    • Application of CNS filters to ensure blood-brain barrier penetration potential

    • Implementation of solubility models to predict bioavailability

    • Selection from both commercial compounds and specialized libraries

  • Screening cascade:

    • Primary functional assays assessing conversion of H-Glu-AMC fluorogenic substrate into pyroGlu-AMC

    • Selection of hits showing predicted robust binding

    • Iterative optimization based on physicochemical properties

    • Progressive evolution of the docking model during project development

  • Validation in disease models:

    • Cellular models including HEK293 and primary cortical neurons

    • Drosophila models of Huntington's disease

    • Zebrafish HD models

This comprehensive approach has yielded novel QPCT inhibitors capable of rescuing Huntington's disease-related phenotypes across multiple model systems, highlighting the promise of QPCT as a therapeutic target .

How does QPCT expression change in different disease states, and what are the methodological considerations for such analyses?

Analysis of QPCT expression across disease states requires specific methodological approaches to ensure accurate interpretation:

  • Tissue-specific expression patterns:

    • QPCT expression changes may be tissue-specific, requiring careful selection of appropriate control tissues

    • In Huntington's disease mouse models, QPCT mRNA levels were found to be reduced compared to wild-type littermates, suggesting possible compensatory mechanisms

  • Quantification methodologies:

    • qPCR analysis requires appropriate reference gene selection, which may vary by tissue and disease state

    • Statistical analysis should account for biological variability within groups

    • For two-factor analysis (e.g., tissue type and disease state), two-way ANOVA approaches are recommended

  • Expression vs. activity considerations:

    • QPCT enzymatic activity may not directly correlate with expression levels

    • Both parameters should be measured when possible

    • Research indicates that raised QPCT activity may not be a prerequisite for protein aggregation, but rather that normal QPCT activity can contribute to pathological processes

  • Compensatory mechanisms:

    • Decreased QPCT expression in disease models suggests possible compensatory downregulation

    • This has important implications for therapeutic approaches, indicating that QPCT inhibition aligns with natural compensatory mechanisms

These methodological considerations highlight the complexity of QPCT regulation in disease states and emphasize the importance of comprehensive analysis approaches.

How can researchers effectively utilize X-ray crystallography data for human QPCT in structure-based drug design?

Leveraging X-ray crystallography data for QPCT-targeted drug design requires sophisticated approaches that account for protein dynamics:

  • Ensemble-based methodologies:

    • Rather than relying on a single crystal structure, researchers should implement ensemble docking approaches that incorporate multiple QPCT conformations

    • The human QPCT catalytic site demonstrates significant flexibility that must be considered in structure-based design

    • Integration of X-ray structures (PDB IDs: 2AFW, 2AFX, 2AFZ, 3PBB, 3SI0) with molecular dynamics-derived conformations provides a more comprehensive structural landscape

  • Evolution of structural models:

    • Begin with available X-ray structures (initially 2AFW, 2AFX, 2AFZ)

    • Enhance with protein conformations generated through molecular dynamics simulations (100 ns timeframe)

    • Iteratively incorporate newly published structures (like 3PBB and 3SI0) to refine the model

    • Use clustering approaches to select representative conformations from molecular dynamics trajectories

  • Catalytic site analysis:

    • Focus on key residues involved in substrate recognition and catalysis

    • Pay special attention to the flexibility of these residues across different structures

    • Consider water-mediated interactions that may be critical for ligand binding

  • Validation approaches:

    • Cross-validate computational predictions with experimental binding data

    • Implement iterative cycles of prediction, synthesis, and testing

    • Use site-directed mutagenesis of key residues to confirm binding hypotheses

This sophisticated structural approach has proven successful in developing effective QPCT inhibitors that demonstrate activity in cellular and animal models of disease .

What methodological approaches can distinguish between direct and indirect effects of QPCT on protein aggregation?

Differentiating direct from indirect effects of QPCT on protein aggregation requires carefully designed experimental approaches:

  • Domain-specific mutation studies:

    • Compare effects of catalytically inactive mutants (E201Q) to wild-type QPCT

    • The observation that E201Q mutants fail to increase aggregation suggests dependency on enzymatic activity rather than direct protein-protein interactions

  • Substrate diversity analysis:

    • Examine QPCT effects on diverse aggregation-prone proteins:

      • Polyglutamine proteins (Q57, Q81 fused to EGFP)

      • HTT exon 1 with expanded polyglutamine tract

      • Polyalanine expansions (37 alanines)

    • The broad effect across different aggregate-prone proteins suggests an indirect mechanism

  • Interaction studies:

    • Co-immunoprecipitation experiments between QPCT and aggregation-prone proteins

    • The absence of direct interaction (as seen with HTT) points toward indirect mechanisms

  • Chaperone network analysis:

    • Examination of molecular chaperone levels (specifically alpha B-crystallin) following QPCT inhibition

    • The observation that QPCT inhibition increases alpha B-crystallin levels suggests QPCT influences aggregation through modulation of protein quality control mechanisms

  • Mechanistic validation:

    • Alpha B-crystallin knockdown/overexpression studies to determine if it mediates QPCT effects

    • Examination of additional chaperones to identify broader effects on proteostasis networks

These methodological approaches have established that QPCT likely influences protein aggregation indirectly through effects on cellular protein quality control systems rather than direct modification of aggregation-prone proteins .

How can researchers accurately model the relationship between QPCT enzymatic activity and its effects on cellular proteostasis?

Developing accurate models of QPCT's influence on proteostasis networks requires integration of quantitative approaches:

  • Dose-response relationships:

    • Establish quantitative relationships between:

      • QPCT expression/activity levels

      • Alpha B-crystallin induction

      • Aggregation outcomes

    • Compare effects of genetic modulation (siRNA, overexpression) with pharmacological inhibition

  • Temporal dynamics analysis:

    • Determine the time course of changes in:

      • QPCT inhibition/activation

      • Chaperone induction

      • Protein aggregation

    • This helps establish causality and feedback relationships

  • Network modeling approaches:

    • Integrate QPCT into broader proteostasis network models

    • Consider upstream regulators and downstream effectors

    • Account for compensatory mechanisms, as suggested by reduced QPCT expression in HD mouse models

  • Integration of structural and functional data:

    • Relate structure-based insights about QPCT catalytic activity to cellular outcomes

    • Develop predictive models connecting inhibitor binding modes to downstream effects on proteostasis

  • Validation methodology:

    • Test model predictions with targeted experiments

    • Use combination approaches (e.g., QPCT inhibition + chaperone modulation)

    • Validate across multiple model systems from cells to organisms

This integrated modeling approach would advance understanding beyond the current observation that QPCT inhibition increases alpha B-crystallin levels , providing a comprehensive framework for understanding how QPCT enzymatic activity influences cellular proteostasis networks.

What statistical approaches are most appropriate for analyzing complex QPCT experimental designs?

QPCT research often involves complex experimental designs with multiple factors, requiring sophisticated statistical approaches:

  • Multi-factor experimental designs:

    • Two-way ANOVA is recommended for designs with two experimental factors

    • For example, when examining QPCT expression or activity across:

      • Different tissues (e.g., control vs. target tissue)

      • Different treatment conditions (e.g., untreated vs. low dose vs. high dose)

    • This approach allows evaluation of:

      • Main effects of each factor independently

      • Interaction effects between factors

  • Sample size and replication considerations:

    • Three biological replicates per condition represents a minimum standard

    • Power analysis should ideally inform sample size selection

    • Technical replicates should not be confused with biological replicates

  • Data transformation and normalization:

    • Ct values from qPCR should undergo appropriate normalization

    • For gene expression studies, relative quantification using reference genes is standard

    • Data distribution should be examined to determine if parametric tests are appropriate

  • Advanced statistical approaches:

    • Parametric, non-parametric, and paired tests for relative quantification

    • Multiple testing correction when examining numerous endpoints

    • Correlation analyses to examine relationships between QPCT activity and downstream effects

How should researchers interpret contradictory findings about QPCT's role across different model systems?

When faced with contradictory findings about QPCT across different models, researchers should implement a systematic interpretive framework:

  • Model-specific context evaluation:

    • Consider inherent differences between models:

      • Cellular models may lack complex intercellular interactions

      • Animal models may have species-specific QPCT functions

      • Disease models may represent different stages of pathology

    • The observation that QPCT expression is reduced in HD mouse models but its inhibition is still beneficial exemplifies model-specific nuances

  • Methodological reconciliation:

    • Examine differences in:

      • QPCT manipulation approaches (genetic vs. pharmacological)

      • Readout measurements (direct vs. indirect)

      • Temporal aspects of intervention

    • Standardize methodologies where possible to enable direct comparisons

  • Integration of conflicting data:

    • Develop conceptual frameworks that might explain apparent contradictions

    • Consider biphasic responses, compensatory mechanisms, or context-dependent functions

    • For example, reduced QPCT expression in disease models may represent a compensatory response that is insufficient to fully counteract pathology

  • Hierarchy of evidence approach:

    • Weigh evidence based on methodological rigor

    • Prioritize findings replicated across multiple models

    • Consider physiological relevance of different model systems

This systematic approach to interpreting contradictory findings advances understanding beyond simplistic views of QPCT function, allowing for nuanced appreciation of its context-dependent roles in normal physiology and disease states.

What are the recommended approaches for validating novel QPCT substrates in research settings?

Validation of novel QPCT substrates requires a multi-faceted approach combining biochemical, structural, and functional methods:

  • In vitro enzymatic validation:

    • Direct enzymatic assays with recombinant QPCT and candidate substrates

    • Mass spectrometry confirmation of pyroglutamate formation

    • Kinetic characterization to determine substrate efficiency (kcat/Km values)

    • Comparison with known QPCT substrates as positive controls

  • Structural validation:

    • Molecular docking studies using ensemble models of QPCT

    • Analysis of key binding interactions in the QPCT catalytic site

    • Comparison with binding modes of established substrates

    • Mutagenesis of key residues to confirm computational predictions

  • Cellular validation approaches:

    • Examining substrate modification in cells with normal vs. reduced QPCT activity

    • Use of QPCT inhibitors to confirm enzyme-dependent modification

    • Comparison between wild-type QPCT and catalytically inactive E201Q mutant effects

  • Functional significance assessment:

    • Determine biological consequences of substrate modification

    • Compare properties of modified vs. unmodified substrate forms

    • Establish relevance to physiological or pathological processes

    • Connect to broader effects, such as protein aggregation in the case of HTT

These methodological approaches provide a comprehensive framework for validating and characterizing novel QPCT substrates, ensuring both biochemical confirmation and biological relevance.

What emerging technologies hold the most promise for advancing human QPCT research?

As QPCT research progresses, several cutting-edge technologies offer significant potential for advancing our understanding:

  • Cryo-electron microscopy:

    • Beyond X-ray crystallography, cryo-EM can capture QPCT in multiple conformational states

    • Particularly valuable for visualizing QPCT in complex with larger substrates or interacting proteins

    • May provide insights into dynamic aspects of catalysis not captured in crystal structures

  • Proteomics-based substrate identification:

    • Unbiased approaches to identify the complete "QPCTome" of modified proteins

    • Targeted proteomics to quantify pyroglutamate formation on specific substrates

    • Integrative proteomics to map QPCT's position in broader proteostasis networks

  • CRISPR-based technologies:

    • Precise genome editing to create improved cellular and animal models

    • CRISPRi/CRISPRa approaches for graded modulation of QPCT expression

    • Base editing to introduce specific catalytic site mutations

  • Advanced computational approaches:

    • Machine learning for prediction of QPCT substrates and inhibitors

    • Enhanced molecular dynamics simulations to capture long-timescale conformational changes

    • Integration of structural, functional, and -omics data into comprehensive QPCT activity models

These emerging technologies will complement existing approaches in QPCT research, providing deeper mechanistic insights and accelerating translational applications.

How can researchers most effectively translate fundamental QPCT findings into therapeutic applications?

Translating basic QPCT research into therapeutic applications requires strategic approaches spanning the bench-to-bedside continuum:

  • Target validation strategies:

    • Validate QPCT's role across multiple disease models

    • Determine disease-specific vs. general mechanisms

    • Establish clear biological rationale for QPCT modulation in specific conditions

    • Consider compensatory mechanisms, as observed in HD models

  • Optimized inhibitor development pipeline:

    • Continue refinement of structure-based approaches using ensemble models

    • Prioritize CNS penetrance for neurodegenerative applications

    • Develop tissue-selective QPCT modulators for specific disease applications

    • Consider alternative approaches beyond competitive inhibitors

  • Biomarker development:

    • Identify measurable indicators of QPCT activity in accessible fluids

    • Develop assays for pyroglutamate-modified proteins as pharmacodynamic markers

    • Establish correlations between QPCT inhibition and disease-relevant outcomes

  • Strategic clinical translation:

    • Select optimal patient populations based on mechanistic understanding

    • Design proof-of-concept studies with meaningful endpoints

    • Consider combination approaches with other disease-modifying strategies

    • Implement adaptive trial designs informed by biomarker data

This comprehensive translational strategy builds upon the promising results already observed with QPCT inhibitors in preclinical models , creating a roadmap for advancing these findings toward clinical application.

Product Science Overview

Introduction

Glutaminyl-Peptide Cyclotransferase (QPCT) is an enzyme that plays a crucial role in the post-translational modification of proteins. It catalyzes the conversion of N-terminal glutaminyl residues into pyroglutamyl residues, a modification that is essential for the stability and function of various peptide hormones and neuropeptides. This enzyme is particularly significant in the pituitary and adrenal glands, where it is involved in the generation of N-terminal pyroglutamyl groups of peptide hormones such as neurotensin and thyrotropin-releasing hormone .

Recombinant Human QPCT

Recombinant human QPCT is a form of the enzyme that is produced using recombinant DNA technology. This involves inserting the gene that encodes QPCT into a suitable host cell, such as the insect cell line Spodoptera frugiperda (Sf21), which is then cultured to produce the enzyme. The recombinant enzyme is typically tagged with a histidine tag to facilitate purification and is supplied in a carrier-free form to avoid interference from other proteins .

Preparation Methods

The preparation of recombinant human QPCT involves several steps:

  1. Gene Cloning: The gene encoding human QPCT is cloned into an expression vector.
  2. Transformation: The expression vector is introduced into the host cells (e.g., Sf21 cells).
  3. Expression: The host cells are cultured under conditions that promote the expression of the QPCT enzyme.
  4. Purification: The enzyme is purified using affinity chromatography, taking advantage of the histidine tag. The purity of the enzyme is typically greater than 95%, as determined by SDS-PAGE and silver staining .
Chemical Reactions and Activity

The activity of recombinant human QPCT is measured by its ability to convert glutaminyl-AMC (a synthetic substrate) to pyroglutamyl-AMC. The specific activity of the enzyme is greater than 550 pmol/min/μg under the described conditions . This enzymatic activity is crucial for the formation of stable and functional peptide hormones and neuropeptides.

Applications and Importance

Recombinant human QPCT is used in various research applications, including:

  • Studying Post-Translational Modifications: Understanding the role of pyroglutamylation in protein stability and function.
  • Drug Development: Investigating the enzyme’s role in diseases and developing inhibitors as potential therapeutic agents.
  • Biochemical Assays: Serving as a standard in enzyme activity assays and other biochemical studies .

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.