ACHE Human

Acetylcholinesterase Human Recombinant
Shipped with Ice Packs
In Stock

Description

ACHE Human Recombinant produced in HEK cells is a single, glycosylated, polypeptide chain (32-614 a.a) containing a total of 592 amino acids, having a molecular mass of 65.6 kDa.
ACHE is fused to a 6 amino acid His-tag at C-terminus,and is purified by proprietary chromatographic techniques.

Product Specs

Introduction

Acetylcholinesterase (ACHE), a member of the type-B carboxylesterase/lipase family, is an enzyme responsible for breaking down the neurotransmitter acetylcholine, as well as other choline esters. In the process of neurotransmission, ACHE plays a crucial role in terminating the signal transmission. After acetylcholine is released from the presynaptic neuron into the synaptic cleft and binds to ACH receptors on the post-synaptic membrane to transmit the nerve signal, ACHE, located on the post-synaptic membrane, hydrolyzes ACH, thereby ending the signal transmission.

Description

Recombinant human ACHE, produced in HEK cells, is a single, glycosylated polypeptide chain with a molecular weight of 65.6 kDa. The chain consists of 592 amino acids (32-614 a.a). The ACHE protein has a 6 amino acid His-tag fused at its C-terminus and undergoes purification using proprietary chromatographic methods.

Physical Appearance

The product is a colorless solution that has been sterilized by filtration.

Formulation

The ACHE solution has a concentration of 0.25mg/ml and contains 10% Glycerol and Phosphate-Buffered Saline with a pH of 7.4.

Stability

For optimal storage, keep the ACHE vial at 4°C if you plan to use it within 2-4 weeks. For longer storage, it should be frozen at -20°C. To ensure stability during long-term storage, adding a carrier protein (0.1% HSA or BSA) is recommended. Avoid repeatedly freezing and thawing the product.

Purity

The purity of the ACHE is determined by SDS-PAGE analysis and is greater than 95.0%.

Biological Activity

The specific activity of ACHE is determined by measuring the amount of enzyme required to cleave 1 nmole of acetylthiocholine per minute at a pH of 7.5 and a temperature of 25°C. This activity is greater than 6,000 nmol/min/ug.

Synonyms

AChE, ACEE, ACES_HUMAN, Acetylcholinesterase, ACHE, ARACHE, N-ACHE, VT, Acetylcholinesterase isoform E4-E6

Source

HEK293 Cells.

Amino Acid Sequence

DGSEGREDAE LLVTVRGGRL RGIRLKTPGG PVSAFLGIPF AEPPMGPRRF LPPEPKQPWS GVVDATTFQS VCYQYVDTLY PGFEGTEMWN PNRELSEDCL YLNVWTPYPR PTSPTPVLVW IYGGGFYSGA SSLDVYDGRF LVQAERTVLV SMNYRVGAFG FLALPGSREA PGNVGLLDQR LALQWVQENV AAFGGDPTSV TLFGESAGAA SVGMHLLSPP SRGLFHRAVL QSGAPNGPWA TVGMGEARRR ATQLAHLVGC PPGGTGGNDT ELVACLRTRP AQVLVNHEWH VLPQESVFRF SFVPVVDGDF LSDTPEALIN AGDFHGLQVL VGVVKDEGSY FLVYGAPGFS KDNESLISRA EFLAGVRVGV PQVSDLAAEA VVLHYTDWLH PEDPARLREA LSDVVGDHNV VCPVAQLAGR LAAQGARVYA YVFEHRASTL SWPLWMGVPH GYEIEFIFGI PLDPSRNYTA EEKIFAQRLM RYWANFARTG DPNEPRDPKA PQWPPYTAGA QQYVSLDLRP LEVRRGLRAQ ACAFWNRFLP KLLSATDTLD EAERQWKAEF HRWSSYMVHW KNQFDHYSKQ DRCSDLHHHH HH 

Q&A

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

Human acetylcholinesterase (AChE) is a key regulatory enzyme of central and peripheral cholinergic function in the nervous system. Its primary biological function is to catalyze the hydrolysis of the neurotransmitter acetylcholine at cholinergic synapses, thereby terminating synaptic transmission. This enzyme plays a crucial role in regulating neural communication by controlling acetylcholine levels in the synaptic cleft, which directly affects the duration of signal transmission between neurons and from neurons to muscles. AChE is particularly essential for proper functioning of both the central and peripheral nervous systems, and alterations in its activity are associated with various neurological and neuromuscular disorders .

How should researchers approach developing a primary research question about human AChE?

Researchers should approach developing a primary research question about human AChE by following the PICOT framework (Population, Intervention, Comparator, Outcome, and Time frame). This systematic approach ensures that the research question is focused and addressable. For example, rather than asking a broad question like "How does cyclophosphamide affect AChE?" researchers should narrow it down to something more specific: "Among patients with neurological disorders (P), how does cyclophosphamide treatment (I) compared to standard therapies (C) affect human brain AChE activity (O) over a six-month period (T)?" .

Additionally, researchers should conduct a thorough literature review to identify gaps in existing knowledge about human AChE. This process may reveal areas where evidence is scarce, where literature yields conflicting results, or where methodologies could be improved. It's estimated that up to one-third of the time in a research project could be invested in finding the right primary study question, making this a critical step in the research process .

What are the most common methodological approaches for studying human AChE activity?

The most common methodological approaches for studying human AChE activity include:

  • Spectrophotometric assays: These are widely used to measure AChE activity by monitoring the rate of substrate hydrolysis.

  • Molecular modeling and docking studies: This approach involves creating a computational model of human AChE and studying its interactions with various ligands. For example, researchers have used Swiss Model Workspace to model human AChE structure and Autodock4.2 for docking studies with compounds like cyclophosphamide .

  • X-ray crystallography: While not always available, this technique provides detailed structural information when crystals of human AChE can be obtained.

  • Recombinant expression systems: These allow for the production of human AChE in controlled laboratory conditions for detailed biochemical and functional studies.

  • In vitro inhibition assays: These are used to evaluate how different compounds interact with and potentially inhibit AChE function.

When selecting a methodological approach, researchers should consider the specific research question being addressed and choose techniques that will provide the most relevant and reliable data .

How can researchers optimize experimental design when studying human AChE inhibitors?

Researchers can optimize experimental design when studying human AChE inhibitors by carefully applying the FINER criteria (Feasible, Interesting, Novel, Ethical, and Relevant) to their research questions. A well-designed study should be achievable from ethical and practical perspectives, interesting to the field, contribute new knowledge, comply with ethical standards, and provide clinically or scientifically relevant information .

For human AChE inhibitor studies specifically, researchers should:

  • Ensure adequate research design: Define clear experimental and control groups, with appropriate sample sizes determined through power analysis.

  • Standardize measurement protocols: Establish consistent protocols for measuring AChE activity and inhibition to ensure reproducibility.

  • Select appropriate model systems: Choose between in vitro systems (using recombinant human AChE), ex vivo samples (from human tissues where available), or in silico approaches (computational modeling).

  • Account for potential confounders: Control for factors that might influence AChE activity, such as age, sex, genetic variations, and concurrent medications.

  • Plan for statistical analysis: Determine in advance which statistical tests will be appropriate for the data generated and account for multiple comparisons if necessary .

Optimizing these aspects of experimental design will lead to more robust and meaningful research outcomes when studying human AChE inhibitors.

What computational modeling approaches are most effective for studying human AChE interactions?

The most effective computational modeling approaches for studying human AChE interactions incorporate multiple complementary methods to ensure accuracy and reliability. Based on current research, a comprehensive computational approach should include:

  • Homology modeling: When X-ray crystallographic data is unavailable, as is often the case with human AChE variants, homology modeling using tools like Swiss Model Workspace can effectively predict protein structure. For example, in studies of human brain AChE, models based on the amino acid sequence (NCBI Accession No: AAI05061.1) have been successfully created and validated .

  • Model verification: Before proceeding to interaction studies, models should be rigorously verified using multiple assessment tools. The QMEAN scoring function provides a composite score that reflects the quality of the model compared to experimental structures of similar size. A Z-score QMEAN near zero (e.g., -0.58) indicates good quality. Additional verification should include analysis of Z-scores for Cβ interaction, all-atom interaction, solvation, and torsion .

  • Molecular docking: Software like Autodock4.2 can be used to predict binding modes and affinities between human AChE and potential inhibitors or substrates. This approach has been successfully applied to study interactions between human AChE and compounds like cyclophosphamide .

  • Energy minimization: Before docking studies, ligand molecules should undergo energy minimization using appropriate force fields (e.g., MMFF94) to ensure they are in their most stable conformation .

  • Molecular dynamics simulations: These can provide insights into the dynamic behavior of AChE-ligand complexes over time, revealing conformational changes that may not be apparent from static models.

By combining these approaches, researchers can develop robust computational models for studying human AChE interactions, which can guide subsequent experimental work and help identify key residues involved in substrate or inhibitor binding.

How should researchers interpret contradictory data in human AChE inhibition studies?

Researchers should approach contradictory data in human AChE inhibition studies systematically through a multi-step process:

  • Evaluate methodological differences: First, examine differences in experimental procedures that might explain contradictory results. This includes:

    • Assay conditions (pH, temperature, buffer composition)

    • Enzyme source (recombinant vs. tissue-derived)

    • Substrate concentrations

    • Inhibitor preparation methods

  • Consider sample characteristics: Differences in sample populations can contribute to contradictory findings. Researchers should analyze:

    • Genetic variations in the AChE enzyme

    • Age and sex distribution of sample donors

    • Health status and medication use of donors

    • Tissue-specific expression patterns of AChE variants

  • Assess statistical power: Insufficient sample sizes may lead to false positives or negatives. Researchers should:

    • Calculate post-hoc power analyses

    • Consider the possibility of Type I and Type II errors

    • Evaluate statistical methods used for data analysis

  • Conduct meta-analyses or systematic reviews: When multiple contradictory studies exist, formal meta-analyses can help reconcile differences and identify patterns across studies.

  • Design validation studies: When contradictions persist, researchers should design new studies specifically to address the contradictions, using standardized methodologies and transparent reporting of all variables.

When interpreting contradictory findings, researchers should remember that biological systems are complex, and apparently contradictory results may reflect genuine biological variability rather than methodological errors. Acknowledging this complexity in research publications is essential for advancing the field .

What are the key structural features of human AChE that influence inhibitor binding?

The key structural features of human AChE that influence inhibitor binding include several specialized regions that have evolved for specific functional purposes:

  • Active site gorge: Human AChE features a deep, narrow gorge approximately 20Å long that leads to the catalytic site. This gorge creates a confined space that influences the binding kinetics and orientation of potential inhibitors. The architecture of this gorge acts as a physical filter, allowing only molecules of appropriate size and shape to access the catalytic site .

  • Catalytic triad: At the bottom of the active site gorge, the catalytic triad consists of three amino acid residues: serine, histidine, and glutamate. These residues work together to catalyze the hydrolysis of acetylcholine. Inhibitors often interact directly with these residues, particularly the nucleophilic serine, to form covalent or non-covalent complexes that prevent normal enzyme function .

  • Peripheral anionic site (PAS): Located at the entrance of the active site gorge, the PAS consists of aromatic and acidic residues that create a negatively charged surface. This site serves as an initial binding site for inhibitors and substrates before they enter the gorge. Binding to the PAS can cause allosteric changes that affect catalytic activity even without direct interaction with the catalytic triad .

  • Acyl pocket: This hydrophobic pocket accommodates the methyl group of acetylcholine during normal catalysis. The size and hydrophobicity of this pocket influence inhibitor specificity, particularly for bulkier compounds.

  • Oxyanion hole: This structural feature stabilizes the tetrahedral intermediate formed during acetylcholine hydrolysis. Certain inhibitors exploit this feature to increase binding affinity.

Understanding these structural elements is crucial for designing selective inhibitors and interpreting molecular docking studies. For example, research has shown that cyclophosphamide interacts with specific residues in the human AChE structure, which may explain its inhibitory effects observed in various models .

How do genetic variations in human AChE affect inhibitor sensitivity and experimental outcomes?

Genetic variations in human AChE can significantly influence inhibitor sensitivity and experimental outcomes through multiple mechanisms:

  • Single nucleotide polymorphisms (SNPs): Various SNPs in the ACHE gene can alter amino acid sequences, potentially changing:

    • The conformation of the active site

    • The electrostatic properties of binding pockets

    • Protein stability and expression levels

    • Post-translational modifications

  • Splice variants: Human AChE exists in multiple splice variants (including AChE-T, AChE-H, and AChE-R) with different C-terminal sequences. These variants show differential sensitivity to inhibitors and are expressed at varying levels in different tissues. Research designs must account for these variants, especially when studying tissue-specific effects.

  • Quantitative trait loci (QTLs): Regulatory regions affecting AChE expression levels can influence the apparent potency of inhibitors in different individuals or populations.

  • Pharmacogenetic implications: When studying AChE inhibitors as potential therapeutics, researchers must consider how genetic variations might lead to:

    • Variable drug response

    • Different side effect profiles

    • Altered pharmacokinetics or pharmacodynamics

To address these challenges, researchers should:

  • Sequence the ACHE gene in experimental samples when possible

  • Report which splice variants were studied

  • Include genetic background as a variable in statistical analyses

  • Consider using stratified analysis based on genetic profiles

  • Develop experimental protocols that account for individual variations

By accounting for genetic variations, researchers can better understand the sometimes contradictory results between studies and develop more personalized approaches to AChE-targeting therapeutics.

What are the current methodological challenges in translating in vitro human AChE findings to in vivo applications?

Translating in vitro human AChE findings to in vivo applications presents several methodological challenges that researchers must address:

  • Microenvironment differences: The controlled environment of in vitro studies differs significantly from the complex biological milieu in vivo. Factors such as pH, ion concentrations, protein-protein interactions, and cellular compartmentalization can all affect AChE function and inhibitor interactions. Researchers should develop experimental designs that account for these differences, perhaps by using more complex in vitro systems that better mimic physiological conditions .

  • Pharmacokinetic considerations: In vitro studies typically expose AChE directly to inhibitors at fixed concentrations, while in vivo applications must account for:

    • Absorption and distribution of compounds

    • Metabolism and potential conversion to active/inactive metabolites

    • Clearance rates and routes

    • Blood-brain barrier penetration for CNS applications

  • Isoform differences and tissue specificity: Human AChE exists in multiple molecular forms with tissue-specific expression patterns. For example, the tetrameric G4 form predominates in the brain and muscles, while other forms are more common in other tissues. In vitro studies often use a single recombinant form, which may not accurately represent the diverse forms found in vivo .

  • Species differences: Many preliminary in vivo studies use animal models, but human AChE differs from that of other species in subtle but important ways. These differences can affect inhibitor binding and efficacy. Computational modeling approaches like those used in the cyclophosphamide-AChE interaction studies can help predict human-specific effects, but verification remains challenging .

  • Long-term effects and adaptations: In vitro studies typically examine acute effects, while in vivo applications must consider long-term consequences, including:

    • Compensatory changes in acetylcholine receptors

    • Altered expression of AChE and related proteins

    • Potential development of tolerance

    • Secondary physiological adaptations

Researchers can address these challenges by employing a staged research approach that progresses from simple in vitro systems to more complex models before moving to in vivo applications. This might include using:

  • Human tissue slices that maintain cellular architecture

  • Organoid cultures that recapitulate tissue organization

  • Humanized animal models

  • Advanced computational models that incorporate physiological parameters

How can researchers effectively use molecular docking studies to predict human AChE inhibitor efficacy?

Researchers can effectively use molecular docking studies to predict human AChE inhibitor efficacy by following a systematic approach that combines computational modeling with experimental validation:

  • Obtain a high-quality human AChE structure: Either retrieve a validated crystal structure from the Protein Data Bank or create a homology model using tools like Swiss Model Workspace. When using homology models, researchers should verify quality through multiple assessment methods. For example, when modeling human brain AChE, researchers should evaluate parameters such as QMEAN scores (-0.58 indicates good quality) and Z-scores for various structural features (Cβ interaction, all-atom interaction, solvation, and torsion) .

  • Prepare ligands properly: Potential inhibitors should undergo energy minimization using appropriate force fields (e.g., MMFF94) before docking. All relevant chemical states should be considered, including tautomers, ionization states at physiological pH, and realistic conformations .

  • Select appropriate docking algorithms: Different algorithms (such as those in Autodock4.2) have strengths and weaknesses. Researchers should consider:

    • Rigid receptor vs. flexible receptor docking

    • Treatment of water molecules

    • Scoring functions appropriate for the chemical classes being studied

  • Establish validation protocols: Before studying novel compounds, researchers should validate their docking protocol using known AChE inhibitors with experimentally determined binding modes and affinities. The protocol should reliably reproduce known binding constants and poses.

  • Analyze interaction patterns: Beyond simple binding energy scores, researchers should analyze:

    • Hydrogen bonding networks

    • Hydrophobic interactions

    • π-stacking with aromatic residues

    • Water-mediated interactions

    • Interactions with specific regions (catalytic triad, peripheral anionic site)

  • Consider active site dynamics: Static docking may miss important dynamic effects. Molecular dynamics simulations following initial docking can provide insights into stability of binding modes and induced-fit effects.

  • Correlate computational predictions with experimental data: To establish predictive value, researchers should correlate docking scores with experimental inhibition constants using regression analysis. This allows for calibration of the computational model.

By following these approaches, researchers can develop docking protocols that provide valuable predictions about potential AChE inhibitors before committing resources to chemical synthesis and experimental testing, as demonstrated in studies of the interaction between human AChE and compounds like cyclophosphamide .

What is the relationship between human AChE inhibition and cyclophosphamide treatment in research models?

The relationship between human AChE inhibition and cyclophosphamide (CP) treatment in research models reveals a complex interaction with potential clinical implications. Studies have demonstrated that CP can inhibit brain and retinal AChE enzymatic activity in several animal models, though the exact mechanism and relevance to human enzyme function remained unclear until recent computational studies .

Molecular docking studies using models of human brain AChE have provided insights into the potential mechanisms of this inhibition. The interaction between human AChE and CP appears to involve specific amino acid residues that may explain the observed inhibitory effects. These computational models were created using the Swiss Model Workspace based on the human AChE amino acid sequence (NCBI Accession No: AAI05061.1) and validated through quality assessment measures .

The docking studies revealed:

  • CP can bind to specific sites on the human AChE enzyme

  • This binding may interfere with the normal catalytic function of AChE

  • The interaction appears to be specific rather than random, suggesting a direct inhibitory mechanism

This relationship has several research implications:

  • Neurological side effects: The inhibition of AChE by CP may contribute to some of the neurological side effects observed in patients receiving this chemotherapeutic agent, which warrants further investigation.

  • Dual-action potential: Understanding this interaction may lead to the development of new compounds that combine anticancer properties with controlled AChE inhibition for specific therapeutic applications.

  • Structure-activity relationships: The insights from these docking studies could guide the design of more selective AChE inhibitors by identifying key structural features that determine binding affinity and specificity .

Further research is needed to fully characterize this relationship and its clinical significance, particularly in human subjects, as most current evidence comes from animal models and computational studies.

How can researchers implement the PICOT and FINER frameworks to develop better human AChE research questions?

Researchers can implement the PICOT and FINER frameworks to develop better human AChE research questions through a structured approach that ensures both methodological rigor and scientific relevance:

Implementing the PICOT Framework:

  • Population (P): Clearly define the study population for human AChE research.

    • Example: "In adult patients with Alzheimer's disease" rather than "In patients"

    • AChE-specific consideration: Specify genetic variants, age groups, or disease states that might affect AChE expression or function

  • Intervention (I): Precisely describe the intervention being studied.

    • Example: "Does administration of a specific AChE inhibitor at 5mg/kg twice daily" rather than "Does medication"

    • AChE-specific consideration: Include dosage, route, duration, and chemical characteristics of inhibitors

  • Comparator (C): Identify appropriate control or comparison groups.

    • Example: "Compared to the current standard donepezil treatment" rather than "Compared to other treatments"

    • AChE-specific consideration: Consider including multiple comparators, such as different classes of AChE inhibitors

  • Outcome (O): Define specific, measurable outcomes.

    • Example: "Improve cognitive function as measured by ADAS-Cog scores" rather than "Improve symptoms"

    • AChE-specific consideration: Include both biochemical measures (e.g., AChE activity levels) and functional outcomes

  • Time frame (T): Specify the study duration and assessment points.

    • Example: "Over a 12-month period with assessments at 3, 6, and 12 months" rather than "Over time"

    • AChE-specific consideration: Account for potential adaptation and compensatory mechanisms that may develop over time

Implementing the FINER Framework:

  • Feasible (F): Ensure the study is practically achievable.

    • Key questions: Is the required technology available? Can adequate samples be obtained? Is funding sufficient?

    • AChE-specific example: A study requiring fresh human brain tissue may not be feasible, but cerebrospinal fluid samples might be obtainable

  • Interesting (I): The question should engage the scientific community.

    • Key questions: Does it address a gap in knowledge? Will results advance the field?

    • AChE-specific example: Investigating novel binding sites on human AChE that could lead to more selective inhibitors

  • Novel (N): The research should add new information.

    • Key questions: Has this been studied before? Does it extend previous work in meaningful ways?

    • AChE-specific example: Examining how specific genetic polymorphisms affect response to AChE inhibitors in different populations

  • Ethical (E): The research must adhere to ethical standards.

    • Key questions: Are risks minimized? Is informed consent possible? Are vulnerable populations protected?

    • AChE-specific example: Using computational modeling and in vitro studies before human trials of new AChE inhibitors

  • Relevant (R): Results should matter to science, clinical practice, or public health.

    • Key questions: Will findings change practice? Do they address a significant health problem?

    • AChE-specific example: Studying how AChE inhibitors might be repurposed for emerging neurodegenerative conditions

By systematically applying these frameworks, researchers can develop human AChE research questions that are both scientifically sound and practically answerable, leading to more impactful research outcomes.

Framework ComponentExamples for Human AChE ResearchCommon Pitfalls to Avoid
PopulationAlzheimer's patients with specific ApoE genotypesToo broad (e.g., "patients with cognitive decline")
InterventionReversible AChE inhibitor at specific dose and scheduleUndefined parameters (e.g., "AChE inhibitor treatment")
ComparatorStandard-of-care AChE inhibitor or placeboMissing appropriate controls
OutcomeChange in CSF AChE activity and ADAS-Cog scoresSingle outcome type or unmeasurable outcomes
Time frame6-month treatment with measurements at baseline, 3 and 6 monthsInadequate follow-up for observing meaningful changes
FeasibleCSF sampling at three timepointsRequiring samples that cannot be ethically obtained
InterestingNovel binding mechanisms for selective inhibitionQuestions already thoroughly answered
NovelFirst study of a new inhibitor class in humansReplication without innovation
EthicalIn silico and in vitro screening before human studiesUnnecessary risks to participants
RelevantAddressing treatment resistance in neurodegenerative diseaseInsights without practical applications

What analytical techniques provide the most reliable measurements of human AChE activity?

The most reliable analytical techniques for measuring human AChE activity include a combination of established biochemical assays and newer advanced methods, each with specific strengths and applications:

  • Ellman's Colorimetric Assay: This traditional method remains widely used due to its reliability and accessibility. It measures AChE activity by detecting the yellow product (5-thio-2-nitrobenzoic acid) formed when the enzyme hydrolyzes acetylthiocholine. For human AChE research, modifications to the original protocol can increase specificity:

    • Using specific inhibitors of butyrylcholinesterase to isolate AChE activity

    • Optimizing substrate concentrations for human enzyme kinetics

    • Controlling reaction conditions (pH 8.0, 25°C) for consistent results

    • Including appropriate controls to account for non-enzymatic hydrolysis

  • Radiometric Assays: These provide excellent sensitivity and specificity by using radiolabeled substrates (typically 3H or 14C-labeled acetylcholine). The radioactive product (acetate) is separated and quantified, offering several advantages:

    • Lower detection limits than colorimetric methods

    • Fewer interference issues from sample components

    • Direct measurement of the natural substrate

  • Fluorometric Methods: These utilize fluorogenic substrates that generate fluorescent products upon hydrolysis by AChE. Modern variants include:

    • Fluorescein diacetate assays

    • Amplified fluorescence techniques

    • Continuous monitoring capabilities for kinetic studies

  • Mass Spectrometry-Based Approaches: LC-MS/MS methods allow direct quantification of acetylcholine and its metabolites in complex biological samples, offering:

    • Superior specificity without interference from other esterases

    • Ability to measure activity in complex biological matrices

    • Simultaneous analysis of multiple cholinergic markers

    • Detection of acetylcholine and choline with excellent sensitivity

  • Electrochemical Techniques: These methods monitor the electroactive species produced during ACh hydrolysis and are particularly valuable for:

    • Real-time monitoring of AChE activity

    • Biosensor applications

    • Field-portable testing systems

For optimal reliability in human AChE research, the following methodological considerations are essential:

  • Tissue-specific optimization: Assay conditions should be optimized for the specific human tissue being studied, as AChE characteristics vary between brain, erythrocytes, and other tissues.

  • Selective inhibitors: Use of selective inhibitors helps distinguish AChE from butyrylcholinesterase activity in mixed samples.

  • Standardization: Reference standards and quality controls should be included in every assay run.

  • Multiple methodologies: When possible, combining two different analytical approaches provides greater confidence in results.

By selecting and optimizing the appropriate analytical technique for specific research questions, investigators can ensure reliable measurements of human AChE activity across various experimental conditions.

How should researchers account for individual variability in human AChE studies?

Researchers should implement a comprehensive strategy to account for individual variability in human AChE studies, addressing biological, methodological, and statistical considerations:

Biological Sources of Variability:

  • Genetic factors: Researchers should:

    • Screen for known polymorphisms in the ACHE gene

    • Consider genotyping subjects for variants known to affect enzyme activity

    • Include genetic background as a covariate in statistical analyses

    • Consider stratified analysis based on genetic profiles

  • Age-related differences: AChE activity and distribution change throughout the lifespan. Studies should:

    • Establish narrow age ranges for participant inclusion

    • Create age-matched control groups

    • Include age as a covariate in statistical models

    • Report age-specific reference ranges

  • Sex differences: Hormonal influences can affect AChE expression. Researchers should:

    • Balance sex distribution in study samples

    • Analyze data for potential sex-specific effects

    • Consider hormonal status in female participants

Methodological Approaches:

  • Standardized protocols: To minimize technique-related variability:

    • Establish detailed standard operating procedures

    • Use the same lot of reagents throughout a study

    • Implement rigorous quality control measures

    • Include internal standards in each analytical run

  • Repeated measurements: When feasible:

    • Obtain multiple samples from each subject

    • Establish intra-individual variability metrics

    • Calculate coefficients of variation for each participant

  • Reference standards: To normalize between subjects:

    • Develop population-specific reference ranges

    • Use relative values (percent of control) when appropriate

    • Include positive and negative controls in each assay

Statistical Considerations:

  • Sample size determination: Properly powered studies require:

    • A priori power analysis based on expected effect sizes

    • Adjustment for anticipated variability

    • Consideration of multiple testing corrections

    • Planning for potential dropouts or technical failures

  • Advanced statistical methods:

    • Mixed-effects models to account for within-subject correlations

    • Bayesian approaches that can incorporate prior knowledge

    • Sensitivity analyses to assess robustness of findings

    • Analysis of outliers with biological justification

  • Reporting practices:

    • Transparent reporting of all variables collected

    • Clear description of inclusion/exclusion criteria

    • Publication of individual data points when possible

    • Discussion of limitations related to variability

Implementation Table:

Source of VariabilityAssessment MethodMitigation Strategy
Genetic polymorphismsGenotyping of ACHE geneStratification or covariate adjustment
Age-related differencesAge documentationAge-matching or statistical adjustment
Sex and hormonal factorsSex documentation, hormonal assaysBalanced recruitment, subgroup analysis
Diurnal variationsStandardized sampling timesConsistent sampling protocols
Disease statesClinical assessment, biomarkersClear case definitions, severity measures
Medication effectsMedication history, drug assaysExclusion criteria or washout periods
Technical variabilityReplicate measurementsQuality control systems, standardization

By systematically addressing these sources of variability, researchers can enhance the reliability and reproducibility of human AChE studies, leading to more robust and clinically relevant findings .

What are the best practices for designing inhibitor selectivity studies for human AChE?

Best practices for designing inhibitor selectivity studies for human AChE require a multi-faceted approach that addresses enzyme specificity, structural considerations, and methodological rigor:

Enzyme Preparation and Characterization

  • Source selection: Use recombinant human AChE expressed in mammalian cells rather than prokaryotic systems to ensure proper folding and post-translational modifications.

  • Isoform specificity: Clearly identify which splice variant(s) of human AChE are being studied (AChE-T, AChE-H, AChE-R) as they may have different inhibitor sensitivity profiles.

  • Purity verification: Employ multiple methods (SDS-PAGE, mass spectrometry) to confirm enzyme purity and identity before inhibition studies.

  • Activity standardization: Normalize enzyme preparations to consistent activity levels rather than protein concentration to account for variations in specific activity .

Counter-Screening Panel Development

  • Related cholinesterases: Always include human butyrylcholinesterase (BChE) in selectivity panels due to its structural similarity and overlapping substrate specificity with AChE.

  • Other esterases: Include carboxylesterases and other related enzymes that might interact with potential inhibitors.

  • Species orthologs: Test against rodent AChE to identify potential species differences that could affect preclinical to clinical translation.

  • Off-target proteins: Screen against common off-target proteins such as monoamine oxidases, cytochrome P450 enzymes, and transporters that might interact with the chemical scaffold of the inhibitor .

Assay Methodology

  • Multiple assay formats: Employ at least two different detection methods (e.g., Ellman's colorimetric assay and a fluorescence-based method) to confirm selectivity findings and rule out assay-specific artifacts.

  • Substrate considerations: Use both artificial substrates (acetylthiocholine) and natural substrate (acetylcholine) when possible, as inhibitor selectivity can sometimes be substrate-dependent.

  • Kinetic characterization: Determine full inhibition mechanisms (competitive, non-competitive, uncompetitive, or mixed) rather than just IC50 values.

  • Time-dependent effects: Assess both immediate inhibition and time-dependent effects to identify slow-binding or irreversible inhibitors .

Experimental Design Features

  • Concentration-response curves: Generate complete concentration-response curves rather than testing at single concentrations.

  • Replication requirements: Perform experiments in triplicate at minimum, with at least three independent enzyme preparations.

  • Reference compounds: Include established selective and non-selective inhibitors as benchmarks (e.g., donepezil for AChE selectivity, rivastigmine for dual AChE/BChE inhibition).

  • Blind testing: Conduct key selectivity experiments in a blinded fashion to eliminate investigator bias .

Advanced Characterization

  • Structural studies: Complement functional assays with structural studies (X-ray crystallography or molecular modeling) to understand the molecular basis of selectivity.

  • Cellular validation: Validate selectivity in cellular systems that express human AChE to account for cellular compartmentalization and protein-protein interactions.

  • Ex vivo confirmation: When possible, confirm selectivity findings using ex vivo human tissue samples with endogenous AChE expression .

Data Analysis and Reporting

  • Selectivity indices: Calculate and report formal selectivity indices (ratio of IC50 values) for all enzymes in the counter-screening panel.

  • Statistical rigor: Employ appropriate statistical methods for comparing potency across enzymes, including confidence intervals.

  • Transparent reporting: Fully disclose all experimental conditions, including buffer compositions, enzyme concentrations, and incubation times that might affect selectivity determinations .

By adhering to these best practices, researchers can develop robust and reproducible selectivity profiles for human AChE inhibitors, leading to better understanding of structure-activity relationships and more targeted therapeutic development.

What are the current gaps in human AChE research methodologies?

Current gaps in human AChE research methodologies span technical, translational, and conceptual domains that limit our understanding of this crucial enzyme system. These gaps represent important opportunities for methodological advancement:

  • Limited access to native human enzyme: Most studies rely on recombinant enzymes or animal models, which may not fully recapitulate the complexity of native human AChE in its physiological context. This gap is particularly problematic when studying:

    • Tissue-specific variants and post-translational modifications

    • Protein-protein interactions that may modify enzyme function

    • Compartmentalization effects within neurons and at synapses

  • Integration challenges across scales: There remains a significant disconnect between:

    • Molecular and structural studies (often in silico or with purified proteins)

    • Cellular and tissue-level investigations

    • Systems-level understanding of AChE function in complex networks

    • Translation to clinical observations and therapeutic applications

  • Temporal dynamics limitations: Current methodologies are often inadequate for capturing:

    • Real-time changes in AChE activity in living systems

    • Circadian and activity-dependent regulation

    • Adaptive responses to chronic inhibition

    • Developmental changes in enzyme distribution and function

  • Genetic and population diversity representation: Research frequently fails to account for:

    • Genetic polymorphisms affecting AChE function across different populations

    • Ethnic variations in drug response and AChE inhibitor sensitivity

    • Epigenetic influences on AChE expression

  • Technological limitations: Several technical challenges persist:

    • Lack of highly selective probes for imaging AChE in vivo

    • Insufficient sensitivity for measuring subtle changes in enzymatic activity

    • Difficulty in distinguishing between different molecular forms of AChE

    • Challenges in creating physiologically relevant in vitro models

  • Contextual understanding: Most assays measure AChE activity in isolation rather than:

    • Within the complete cholinergic signaling pathway

    • In the context of neuroinflammation and oxidative stress

    • As part of non-enzymatic functions of AChE (such as adhesion or development)

  • Standardization and reproducibility issues: The field suffers from:

    • Inconsistent methodologies between laboratories

    • Variable reporting of experimental conditions

    • Lack of standardized reference materials

    • Insufficient validation of computational models against experimental data

Addressing these methodological gaps requires interdisciplinary approaches combining advanced computational modeling, development of more sophisticated in vitro systems, and improved translational models that better connect molecular findings to clinical observations. Particular emphasis should be placed on developing technologies that can study AChE in its native state within functioning neural circuits.

How can researchers contribute to improving human AChE research standards?

Researchers can contribute to improving human AChE research standards through systematic efforts in methodology, reporting, collaboration, and education:

Methodology Enhancement:

  • Standardize experimental protocols: Develop and publish detailed protocols for AChE activity assays that specify:

    • Precise buffer compositions and pH conditions

    • Enzyme source and preparation methods

    • Substrate concentrations and incubation times

    • Temperature control parameters

    • Quality control measures and acceptance criteria

  • Establish reference materials: Create and validate standard reference materials for:

    • Recombinant human AChE calibration

    • Known inhibitor compounds with established potency values

    • Control samples with defined activity levels

    • Certified reference materials for instrument validation

  • Improve computational model validation: When conducting molecular modeling studies:

    • Validate computational predictions against experimental data

    • Report model quality metrics (e.g., QMEAN scores, Z-scores)

    • Make models publicly available in repositories

    • Clearly describe all parameters and constraints used

Reporting Practices:

  • Implement comprehensive reporting guidelines: Follow and promote structured reporting that includes:

    • Complete methods sections that enable experimental reproduction

    • Detailed characterization of enzyme sources

    • Raw data availability when possible

    • Transparent reporting of both positive and negative results

  • Standardize data presentation: Adopt consistent formats for:

    • Inhibition constants (IC50, Ki) with specified conditions

    • Kinetic parameters (Km, Vmax) with standard units

    • Dose-response curves with appropriate statistical analyses

    • Selectivity indices with defined calculation methods

Collaborative Frameworks:

  • Establish multi-laboratory validation studies: Participate in collaborative projects that:

    • Test the same compounds across different laboratories

    • Compare different methodological approaches

    • Establish inter-laboratory reproducibility metrics

    • Develop consensus standards for the field

  • Create shared resources: Contribute to and utilize:

    • Open-access databases of human AChE variants and their properties

    • Repositories of validated human AChE models

    • Biobanks of human samples for AChE research

    • Open-source analysis tools specific to cholinesterase research

Training and Education:

  • Develop specialized training programs: Create or participate in educational initiatives that:

    • Train new researchers in standardized methodologies

    • Promote understanding of common pitfalls and limitations

    • Teach critical evaluation of AChE research literature

    • Share expertise across subspecialties

  • Promote rigorous study design: Incorporate and advocate for:

    • Proper application of PICOT and FINER frameworks

    • Adequate statistical power considerations

    • Appropriate controls and blinding procedures

    • Replication studies and validation cohorts

Innovation Leadership:

  • Develop improved methods: Invest in creating:

    • More selective and sensitive assay technologies

    • Physiologically relevant cell and tissue models

    • Advanced imaging techniques for AChE localization and activity

    • Novel approaches to study AChE in its native environment

  • Bridge basic and clinical research: Work to connect:

    • Basic mechanistic studies with clinical observations

    • Preclinical models with human outcomes

    • Computational predictions with in vivo findings

    • Laboratory discoveries with therapeutic applications

By actively engaging in these improvement efforts, researchers can collectively elevate the standards for human AChE research, leading to more reliable, reproducible, and clinically relevant findings that advance both scientific understanding and therapeutic development.

What future technologies might transform how we study human AChE interactions?

Future technologies are poised to revolutionize the study of human AChE interactions by enabling unprecedented precision, physiological relevance, and systems-level understanding. These emerging approaches will likely transform the field in the coming years:

  • Cryo-electron microscopy (cryo-EM) advances: Next-generation cryo-EM technology will allow:

    • Visualization of human AChE in its native membrane environment

    • Structural determination of AChE complexes with interacting proteins

    • Observation of conformational changes during catalysis at near-atomic resolution

    • Analysis of dynamic inhibitor binding without the need for crystallization

  • AI-driven drug design and screening:

    • Deep learning algorithms will predict inhibitor binding with greater accuracy than current docking approaches

    • Generative adversarial networks will design novel inhibitor scaffolds with optimized selectivity profiles

    • AI systems will integrate structural, functional, and clinical data to identify patterns invisible to human researchers

    • Machine learning will accelerate virtual screening of billions of compounds against specific AChE variants

  • Single-molecule imaging and spectroscopy:

    • Super-resolution microscopy will visualize individual AChE molecules in living cells

    • Single-molecule FRET will monitor real-time conformational changes during substrate binding and catalysis

    • Optical tweezers combined with fluorescence will measure forces and kinetics at the single-molecule level

    • Correlative light and electron microscopy will connect function to ultrastructure

  • Advanced organoid and microphysiological systems:

    • Brain organoids with functional cholinergic circuits will model human AChE in its native cellular context

    • Organ-on-chip platforms will recreate the neuromuscular junction for studying AChE in peripheral cholinergic transmission

    • Multi-organ platforms will model systemic effects of AChE inhibitors, including metabolism and off-target effects

    • Patient-derived systems will enable personalized testing of AChE inhibitors

  • In vivo molecular imaging:

    • PET tracers with improved specificity for AChE will enable non-invasive monitoring of enzyme activity

    • Optogenetic sensors for acetylcholine will allow visualization of cholinergic signaling in real-time

    • Multimodal imaging will connect AChE activity to functional outcomes

    • Molecular imaging agents will detect specific AChE conformational states

  • CRISPR/Cas technologies for precision modification:

    • Base editing will create precise point mutations in the ACHE gene to study structure-function relationships

    • Prime editing will enable modeling of human genetic variants in cellular and animal models

    • CRISPR activation/interference systems will allow temporal control of AChE expression

    • In vivo CRISPR screens will identify novel regulators of AChE function

  • Nanotechnology-based approaches:

    • Nanobiosensors will detect AChE activity with unprecedented sensitivity

    • Nanoparticle-based drug delivery will target specific pools of AChE in different tissues

    • DNA origami scaffolds will position AChE in defined spatial arrangements to study clustering effects

    • Nanopore sensing will analyze single-molecule interactions between AChE and inhibitors

  • Systems biology integration:

    • Multi-omics approaches will connect AChE function to broader physiological networks

    • Digital twin modeling will create personalized simulations of cholinergic systems

    • Network pharmacology will predict system-wide effects of AChE modulation

    • Quantitative systems pharmacology models will optimize dosing regimens for AChE inhibitors

These transformative technologies will address many current limitations in human AChE research by providing more physiologically relevant contexts, greater temporal and spatial resolution, and the ability to connect molecular interactions to system-level outcomes. Researchers who adopt and integrate these approaches will be positioned to make significant advances in understanding AChE biology and developing more effective therapeutic strategies.

Product Science Overview

Structure and Function

Recombinant human acetylcholinesterase (rhAChE) is a fast-acting enzyme with a molecular weight of approximately 67,796 Da per subunit, forming a homodimer with a total molecular weight of 135,792 Da . Each subunit consists of 556 residues, resulting in a total of 1,112 residues for the dimer . The enzyme’s primary structure includes a high number of aromatic residues, along with positively and negatively charged residues, which are integral to its function .

The secondary structure of rhAChE contains 12 β-sheets, 14 α-helices, and 74 random coils . The tertiary structure features an α/β hydrolase fold, a common motif in many hydrolytic enzymes, providing a stable scaffold for the active site . This fold consists of a mostly parallel, eight-stranded β-sheet surrounded by α-helices .

Biological Role

AChE’s primary role is to terminate cholinergic neurotransmission by hydrolyzing acetylcholine, thus preventing continuous stimulation of neurons . This function is vital for maintaining the balance and proper functioning of the nervous system. Additionally, AChE is involved in neuronal apoptosis, further highlighting its importance in neural health .

Clinical Significance

Recombinant human acetylcholinesterase has been extensively studied for its potential therapeutic applications, particularly in treating neurological disorders such as Alzheimer’s disease . AChE inhibitors are used to slow down the breakdown of acetylcholine in the brain, thereby enhancing cholinergic transmission and potentially alleviating symptoms of dementia .

Research and Applications

The recombinant form of AChE is produced using Chinese Hamster Ovary (CHO) cells, ensuring high purity and activity . This recombinant enzyme is used in various research applications, including studying the enzyme’s kinetics, structure-function relationships, and potential drug interactions .

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.