AKHSDH1 Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to AKHSDH1 Antibody

The AKHSDH1 Antibody is a research tool designed to detect and study the bifunctional enzyme AKHSDH1, which plays dual roles in aspartokinase and homoserine dehydrogenase activities. This enzyme is integral to the biosynthesis of essential amino acids like lysine, threonine, and methionine in plants. The antibody enables researchers to investigate its expression, localization, and functional interactions in plant cells .

Research Applications

The AKHSDH1 Antibody is utilized in diverse applications:

  • Protein Expression Analysis: Western blotting and ELISA to quantify AKHSDH1 levels in plant tissues.

  • Localization Studies: Immunofluorescence or immunohistochemistry to map enzyme distribution in chloroplasts.

  • Functional Assays: Investigating AKHSDH1’s role in amino acid biosynthesis under stress conditions.

Recent studies emphasize the need for rigorous antibody validation. For example, a 2023 analysis of 614 antibodies revealed that ~20% failed target recognition, underscoring the importance of using well-characterized reagents .

Quality and Validation

The AKHSDH1 Antibody’s performance hinges on:

  • Specificity: Validated via immunoprecipitation and knockout (KO) cell line controls .

  • Batch Consistency: ≥85% purity ensures minimal lot-to-lot variability .

  • Application-Specific Testing: Compatibility with techniques like SDS-PAGE and immunohistochemistry .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Made-to-order (14-16 weeks)
Synonyms
AKHSDH1 antibody; AK-HSDH antibody; I antibody; At1g31230 antibody; F28K20.19 antibody; T19E23.1 antibody; Bifunctional aspartokinase/homoserine dehydrogenase 1 antibody; chloroplastic antibody; AK-HD 1 antibody; AK-HSDH 1 antibody; Beta-aspartyl phosphate homoserine 1) [Includes: Aspartokinase antibody; EC 2.7.2.4); Homoserine dehydrogenase antibody; EC 1.1.1.3)] antibody
Target Names
AKHSDH1
Uniprot No.

Target Background

Gene References Into Functions
The target protein has been studied in the context of allosteric effectors. A notable research article investigates six novel allosteric effectors of *Arabidopsis thaliana* aspartate kinase-homoserine dehydrogenase isoforms. This study is available on PubMed with the PMID: 16216875.
Database Links

KEGG: ath:AT1G31230

STRING: 3702.AT1G31230.1

UniGene: At.48251

Protein Families
Aspartokinase family; Homoserine dehydrogenase family
Subcellular Location
Plastid, chloroplast.

Q&A

What experimental approaches are most effective for validating AKHSDH1 antibody binding specificity?

Antibody binding specificity validation requires a multi-method approach combining both computational and experimental techniques. For optimal validation, researchers should implement a sequence of complementary assays including ELISA, Western blotting, immunoprecipitation, and immunohistochemistry with appropriate positive and negative controls. Additionally, competitive binding assays with known ligands can confirm binding specificity to target epitopes. Contemporary approaches increasingly incorporate simulation frameworks like Absolut! to predict binding affinities prior to wet-lab confirmation . This computational pre-screening can significantly reduce experimental costs by identifying the most promising antibody candidates for subsequent validation assays. Binding specificity should be assessed across a range of conditions (pH, temperature, salt concentrations) to ensure robust performance in various experimental contexts.

How does epitope mapping inform AKHSDH1 antibody application in research settings?

Epitope mapping provides crucial insights that directly determine appropriate research applications for antibodies. Comprehensive epitope characterization involves multiple complementary approaches including peptide arrays, hydrogen-deuterium exchange mass spectrometry (HDX-MS), and X-ray crystallography of antibody-antigen complexes. This mapping is particularly valuable for predicting cross-reactivity and potential binding interference effects. In antibody engineering studies, researchers frequently employ systematic mutation approaches involving "one-point mutations (covering all possible single-residue changes), two-point mutations, and three-point mutations" to generate comprehensive binding profiles . These mutational analyses reveal critical binding hotspots that inform both antibody optimization and experimental design considerations. Properly mapped epitopes allow researchers to predict potential conformational changes that might affect epitope accessibility in different experimental conditions.

What factors influence reproducibility when using AKHSDH1 antibodies in experimental protocols?

Reproducibility challenges in antibody-based experiments stem from multiple variables that must be controlled. Primary factors include antibody production consistency, storage conditions affecting stability, experimental protocol standardization, and sample preparation techniques. Research-grade antibody preparations often exhibit "comparable or slightly higher immunogenicity risk assessment data" compared to pharmaceutical-grade counterparts, potentially affecting experimental outcomes . To maximize reproducibility, researchers should implement rigorous validation using both positive and negative controls, standardize antibody concentrations based on activity rather than protein content alone, and document lot-to-lot variation through quality control testing. Additionally, environmental factors such as freeze-thaw cycles, buffer composition, and incubation conditions must be precisely controlled and reported to ensure experimental consistency across independent laboratories.

How can active learning strategies optimize antibody-antigen binding prediction for AKHSDH1 research?

Active learning (AL) methodologies significantly enhance antibody-antigen (Ab-Ag) binding prediction accuracy while reducing experimental resource requirements. In a systematic comparison, research demonstrated that "three strategies – Hamming Average Distance, Gradient-Based uncertainty (Last Layer Max) and Query-by-Committee – demonstrated significant performance gains over the random baseline" . The Hamming Average Distance method proved particularly effective, "achieving a 1.795% increase in the area under the active learning curve compared to the random selection baseline" . This approach enabled researchers to reduce the required number of antigen mutant variants by approximately 35%, demonstrating substantial efficiency improvements. For AKHSDH1 antibody research, implementing these AL strategies would allow sequential selection of the most informative experiments to perform, prioritizing those that maximize information gain while minimizing redundancy. This approach is especially valuable when working with limited amounts of precious antibody samples or when screening large libraries of potential antigens.

What computational models best predict AKHSDH1 antibody cross-reactivity with non-target antigens?

Computational prediction of antibody cross-reactivity requires sophisticated models that integrate structural information, sequence homology, and binding energetics. Current state-of-the-art approaches employ machine learning frameworks trained on comprehensive datasets of antibody-antigen interactions. The Absolut! simulation framework has demonstrated particular effectiveness by constructing "a discretized lattice representation of a protein-antigen" that computes "the optimal binding for the Ab sequence, specifically the CDRH3" . This approach enables systematic evaluation of potential cross-reactivity by simulating binding interactions with structural variants of target antigens. For optimal cross-reactivity assessment, researchers should generate comprehensive mutation matrices encompassing "80 one-point mutations, 500 randomly sampled two-point mutations, and 1,500 randomly sampled three-point mutations," creating a robust binding profile . These computational predictions should subsequently undergo experimental validation through binding assays against a panel of structurally related antigens to confirm specificity boundaries.

How do different machine learning approaches compare in antibody-antigen binding prediction performance?

Machine learning approaches for antibody-antigen binding prediction demonstrate varying performance characteristics depending on dataset composition and prediction tasks. Comparative analysis reveals that performance evaluation must integrate multiple metrics, with "receiver operating characteristic area under the curve (ROC AUC) on the test dataset" serving as a primary evaluation criterion . For comprehensive assessment, researchers should evaluate models using distinct test datasets including "TestSharedAG, which utilizes Ag mutant variants from the training dataset and Ab sequences from the test dataset; TestSharedAB, which contains Ag mutant variants from the test dataset and Ab sequences from the training dataset; and Test, which comprises both Ag mutant variants and Ab sequences from the test dataset" . This multi-dataset evaluation approach provides robust performance assessment across varying degrees of novelty in antibody and antigen sequences. Research indicates that diversity-based approaches often outperform purely random sampling methods, particularly in scenarios requiring prediction of novel binding interactions not represented in training data.

What controls are essential when evaluating AKHSDH1 antibody specificity in complex biological samples?

Rigorous control selection represents a critical determinant of experimental validity in antibody specificity assessment. Essential controls include:

Control TypePurposeImplementation
Negative ControlsConfirm absence of non-specific bindingSamples lacking target antigen; isotype-matched irrelevant antibodies
Positive ControlsValidate assay functionalityKnown antibody-antigen pairs with established binding characteristics
Blocking ControlsAssess non-specific interactionsPre-incubation with excess unlabeled antibody
Absorption ControlsConfirm epitope specificityPre-incubation with purified antigen
Secondary-only ControlsDetect secondary antibody artifactsOmission of primary antibody

Additionally, researchers should implement genetic controls where target antigen expression is manipulated through knockdown, knockout, or overexpression approaches. These genetic controls provide definitive evidence of antibody specificity by modulating target abundance in otherwise identical sample backgrounds. For immunohistochemistry applications, tissue samples from multiple donors should be examined to account for biological variability in antigen expression and processing.

How should researchers design experiments to assess potential immunogenicity of AKHSDH1-derived therapeutic antibodies?

Immunogenicity assessment requires a multi-faceted experimental approach combining in silico prediction, in vitro cellular assays, and in vivo models. A comprehensive evaluation framework should include "dendritic cell internalization, T cell proliferation, and T cell epitope identification by in silico prediction and MHC-associated peptide proteomics" . Experimental design should incorporate benchmark antibodies with established immunogenicity profiles as comparative standards, such as "benchmark antibodies with known low (Avastin and Herceptin) or high (bococizumab and ATR-107) clinical incidence of anti-drug antibodies" . Researchers should systematically evaluate T cell epitopes using "peptide clusters presented by HLA-DR, and quantify their signal by mass spectrometry" . This approach enables comparative assessment of potential immunogenicity between novel constructs and established reference antibodies. For engineered antibodies containing specialized domains or modifications, targeted analysis of these regions is essential to identify potential neo-epitopes that might trigger immune responses.

What experimental variables most significantly impact AKHSDH1 antibody performance in different assay conditions?

Antibody performance across different assay platforms is influenced by multiple experimental variables that must be systematically evaluated and controlled:

Variable CategoryCritical ParametersImpact on Assay Performance
Buffer CompositionpH, ionic strength, detergents, blocking agentsAffects epitope accessibility and binding kinetics
TemperatureIncubation temperature, temperature stabilityInfluences binding kinetics and background signal
Sample PreparationFixation method, antigen retrieval techniqueDetermines epitope preservation and accessibility
Antibody ConcentrationWorking dilution, titration optimizationAffects signal-to-noise ratio and specificity
Incubation ConditionsTime, agitation method, washing stringencyImpacts binding efficiency and background reduction

Research indicates that "protein structure of the antigen" represents a particularly critical determinant of binding performance . Experimental protocols should therefore consider potential conformational changes induced by sample processing methods. Systematic optimization studies examining these variables through factorial experimental designs should be conducted during assay development to establish robust protocols that yield consistent performance across different experimental contexts.

How should researchers interpret discrepancies between computational predictions and experimental results for AKHSDH1 antibody binding?

Discrepancies between computational predictions and experimental binding data require systematic investigation through a structured analytical framework. When confronting such inconsistencies, researchers should:

  • Evaluate model assumptions and limitations, particularly regarding structural representations used in computational approaches

  • Assess experimental conditions that might differ from simulation parameters, including buffer composition, pH, and temperature

  • Consider conformational dynamics not captured in static computational models

  • Examine potential post-translational modifications affecting binding not incorporated in prediction algorithms

  • Investigate binding kinetics through techniques like surface plasmon resonance to determine if discrepancies relate to association or dissociation rates

Research demonstrates that while computational frameworks like Absolut! "do not directly predict real-world Ab-Ag binding," they effectively "facilitate the development of machine learning strategies that, when effective on simulated data, also perform well with experimental data" . This principle suggests that relative performance comparisons between computational approaches often translate to experimental settings, even when absolute prediction accuracy varies. When analyzing discrepancies, researchers should prioritize methodological refinement rather than simply dismissing either computational or experimental approaches.

What metrics best evaluate AKHSDH1 antibody performance in research applications?

Comprehensive evaluation of antibody performance requires integration of multiple complementary metrics tailored to specific research applications:

Research ApplicationPrimary MetricsSecondary Metrics
Binding SpecificitySelectivity ratio, cross-reactivity profileBinding competition analysis
Affinity DeterminationKD (equilibrium dissociation constant), kon/koff ratesThermodynamic parameters (ΔH, ΔS)
Functional ActivityEC50/IC50 values, maximum effectDose-response relationship
Stability AssessmentThermal/pH denaturation profiles, aggregation propensityLong-term activity retention
Epitope CharacterizationEpitope mapping resolution, binding interference patternsComputational epitope prediction correlation

How can researchers effectively analyze epitope conservation across species when validating AKHSDH1 antibodies for cross-species applications?

Cross-species application of antibodies necessitates systematic analysis of epitope conservation through an integrated computational and experimental approach. The analytical workflow should include:

  • Multiple sequence alignment of target antigen across species of interest to identify conserved and variable regions

  • Computational prediction of conservation scores at both sequence and structural levels

  • Epitope mapping using techniques such as peptide arrays or hydrogen-deuterium exchange mass spectrometry

  • Experimental validation of binding to recombinant target antigens from multiple species

  • Tissue cross-reactivity studies using immunohistochemistry on multi-species tissue panels

What approaches can resolve non-specific binding issues when using AKHSDH1 antibodies in complex biological samples?

Non-specific binding challenges require systematic troubleshooting through methodical protocol optimization and sample preparation refinement:

  • Buffer Optimization: Integrate detergents (0.1-0.5% Triton X-100 or Tween-20) to reduce hydrophobic interactions and increase salt concentration (150-500 mM NaCl) to minimize electrostatic interactions

  • Blocking Enhancement: Implement comprehensive blocking with 5% BSA or 5-10% serum from the same species as the secondary antibody; consider alternative blockers like casein or commercial blocking solutions for problematic samples

  • Sample Pre-treatment: Develop pre-adsorption protocols where antibodies are pre-incubated with likely cross-reactive components

  • Antibody Dilution Optimization: Systematically titrate antibody concentrations to identify optimal signal-to-noise ratios

  • Cross-linking Fixation Modification: Evaluate different fixation protocols to preserve epitope accessibility while maintaining sample integrity

Research demonstrates that antibody engineering approaches can substantially reduce non-specific binding, particularly through "mutations to facilitate the preferential pairing of cognate HC and LC" . These molecular engineering approaches suggest that selective modifications targeting the antibody variable regions might enhance specificity without compromising target recognition. When persistent non-specific binding issues occur, researchers should consider alternative antibody clones targeting different epitopes on the same antigen.

How can researchers enhance sensitivity for detecting low-abundance targets using AKHSDH1 antibodies?

Enhancing detection sensitivity for low-abundance targets requires integrated optimization of multiple experimental parameters:

Enhancement StrategyMethodological ApproachImplementation Considerations
Signal AmplificationEnzyme-mediated amplification, tyramide signal amplification, rolling circle amplificationBalance amplification with potential increased background
Detection System OptimizationHigh-sensitivity substrates, advanced imaging/detection systemsRequires appropriate instrumentation and controls
Sample EnrichmentImmunoprecipitation, cell fractionation, laser capture microdissectionMay introduce selective bias in target populations
Incubation Parameter OptimizationExtended incubation times, optimized temperature profilesRequires empirical determination of optimal conditions
Multi-epitope DetectionCocktails of antibodies against different epitopesNecessitates validation of antibody compatibility

Research indicates that comprehensive optimization can achieve significant sensitivity improvements while maintaining specificity. Studies employing active learning approaches demonstrate that "selecting diverse Ags based on sequence differences (i.e., a diversity-based approach) leads to better Ag selection and improved model performance" . This principle suggests that developing detection systems targeting multiple distinct epitopes may enhance detection sensitivity while maintaining specificity. Researchers should systematically evaluate each enhancement strategy through controlled comparison studies to identify optimal approaches for specific experimental contexts.

What strategies effectively address batch-to-batch variability in AKHSDH1 antibody preparations?

Batch-to-batch variability represents a significant challenge in antibody-based research, requiring comprehensive mitigation strategies:

  • Standardized Quality Control: Implement consistent quality control testing including ELISA-based activity assays, SDS-PAGE for purity assessment, and specificity verification against known positive and negative samples

  • Reference Standard Establishment: Maintain internal reference standards for comparative performance assessment across production batches

  • Functional Validation: Develop application-specific functional assays that evaluate antibody performance in the context of intended use

  • Lot Reservation: Reserve sufficient quantities of validated lots for critical long-term studies

  • Recombinant Production: Consider transition to recombinant antibody production techniques that offer improved consistency

Research comparing "research-grade preparations of trastuzumab and bevacizumab versus Herceptin and Avastin, respectively" revealed "comparable or slightly higher immunogenicity risk assessment data" for research-grade preparations . These findings highlight the importance of standardized production processes in minimizing batch variability. For critical applications, researchers should implement comprehensive validation protocols for each new antibody lot, including side-by-side comparison with previously validated lots across multiple performance parameters.

How might emerging computational approaches enhance AKHSDH1 antibody engineering for increased specificity?

Next-generation computational approaches offer promising avenues for antibody engineering with enhanced specificity profiles. Machine learning methodologies integrating structural biology and sequence-based prediction are rapidly advancing antibody design capabilities. Research demonstrates that "tailored AL methods are very effective at improving how Ab-Ag pairs are chosen for labeling," with approaches like the Hamming Average Distance method showing particular promise . These computational frameworks enable systematic exploration of sequence space to identify optimal amino acid combinations for enhanced specificity.

Future developments will likely incorporate quantum mechanical calculations of binding energetics, molecular dynamics simulations of binding kinetics, and deep learning architectures trained on expanded antibody-antigen interaction datasets. For engineering AKHSDH1 antibodies, researchers should pursue computational approaches that integrate "clustering-based approaches like the average Hamming distance" which "can be applied at the outset, enabling the selection of Ag mutant variants before iterative learning begins" . This strategy allows rational design of antibody variants with enhanced specificity through systematic exploration of sequence-structure-function relationships.

What methodological advances are needed to improve prediction of AKHSDH1 antibody immunogenicity in therapeutic applications?

Advancing immunogenicity prediction for therapeutic antibodies requires methodological innovations across multiple research domains:

  • Enhanced computational prediction algorithms integrating structural modeling, MHC binding prediction, and immunological pathway analysis

  • Development of standardized in vitro assay systems with improved correlation to clinical immunogenicity outcomes

  • Establishment of comprehensive databases linking antibody structural features to observed immunogenicity in clinical settings

  • Refinement of animal models with humanized immune systems for preclinical immunogenicity assessment

  • Integration of patient-specific factors including HLA typing and immune status in personalized immunogenicity prediction

How will advances in structural biology techniques impact AKHSDH1 antibody characterization and optimization?

Emerging structural biology techniques are transforming antibody characterization and optimization through unprecedented resolution of molecular details:

TechniqueCurrent ApplicationsFuture Impact on Antibody Research
Cryo-electron microscopyHigh-resolution structure determination of antibody-antigen complexesVisualization of dynamic binding events and conformational changes
AlphaFold/RoseTTAFoldPrediction of antibody structures from sequenceRapid screening of engineered variants prior to experimental validation
Hydrogen-deuterium exchange mass spectrometryMapping of protein-protein interaction interfacesDetailed epitope characterization with minimal sample requirements
Single-molecule FRETAnalysis of conformational dynamicsReal-time monitoring of antibody binding events
Serial crystallographyStructure determination with micro/nanocrystalsStructural analysis of challenging antibody-antigen complexes

These advanced structural techniques will enable increasingly detailed characterization of antibody-antigen interactions, supporting rational engineering approaches. Research utilizing simulation frameworks like Absolut! demonstrates how structural information can inform "machine learning strategies that, when effective on simulated data, also perform well with experimental data" . As structural biology techniques continue to advance, researchers will gain increasingly detailed insights into binding mechanisms, supporting precision engineering of antibodies with optimized performance characteristics for specific applications.

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 2025 TheBiotek. All Rights Reserved.