ABAT Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
PBS with 0.1% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze / thaw cycles.
Lead Time
Typically, we can ship the products within 1-3 business days after receiving your order. Delivery times may vary depending on the purchase method or location. Please consult your local distributors for specific delivery timeframes.
Synonyms
(S) 3 amino 2 methylpropionate transaminase antibody; (S)-3-amino-2-methylpropionate transaminase antibody; 4 aminobutyrate aminotransferase antibody; 4 aminobutyrate aminotransferase; mitochondrial antibody; 4-aminobutyrate aminotransferase antibody; ABAT antibody; FLJ17813 antibody; FLJ30272 antibody; GABA aminotransferase antibody; GABA AT antibody; GABA T antibody; GABA transaminase antibody; GABA transferase antibody; GABA-AT antibody; GABA-T antibody; GABAT antibody; GABT_HUMAN antibody; Gamma amino N butyrate transaminase antibody; Gamma-amino-N-butyrate transaminase antibody; hCG1984265 antibody; L AIBAT antibody; L-AIBAT antibody; LAIBAT antibody; mitochondrial antibody; NPD009 antibody
Target Names
ABAT
Uniprot No.

Target Background

Function
ABAT Antibody catalyzes the conversion of gamma-aminobutyrate (GABA) and L-beta-aminoisobutyrate to succinate semialdehyde and methylmalonate semialdehyde, respectively. It can also convert delta-aminovalerate and beta-alanine.
Gene References Into Functions
  1. A study using ER+ IBC identified a metagene including ABAT and STC2 as predictive biomarkers for endocrine therapy resistance. PMID: 25771305
  2. An A-to-G transition at nucleotide 754 of the human ABAT gene identified in lymphoblast cDNA (c.754A>G) results in the substitution of an invariant arginine at amino acid 220 by lysine (p.Arg220Lys). This point mutation destabilizes the binding of pyridoxal-5'-phosphate to GABA-transaminase (essential for transamination of GABA to succinic semialdehyde), leading to GABA-transaminase deficiency. PMID: 25485164
  3. Findings suggest a potential role of ABAT gene-regulated GABA catabolism in altered processing of somatosensory stimuli, potentially serving as a vulnerability marker for affective disorders. PMID: 22225676
  4. Research points to the direct involvement of ABAT in pathways affecting lower esophageal sphincter (LES) control in gastroesophageal reflux disease. PMID: 21552517
  5. Observational study and genome-wide association study of gene-disease association. (HuGE Navigator) PMID: 20659789
  6. Excessive prenatal GABA exposure in the central nervous system (CNS) is responsible for the clinical manifestations of GABA transaminase deficiency [case report]. PMID: 20052547
  7. Significant differences in platelet uptake of GABA and activity of the catabolic enzyme GABA-T were observed between patients with generalized and localization-related epileptic syndromes. This may indicate an impairment in the function of brain GABAergic systems. PMID: 12694932
  8. Research suggests that the Cys321 residue is essential for the catalytic function of GABAT and is involved in the formation of a disulfide link between two monomers of human brain GABAT. PMID: 15528998
  9. Lysine 357 is essential for the catalytic function of brain GABA transaminase and is involved in binding PLP at the active site. PMID: 15650327
  10. Analysis of the autistic disorder susceptibility locus suggests an association on chromosome 16p between GRIN2A and ABAT. PMID: 15830322

Show More

Hide All

Database Links

HGNC: 23

OMIM: 137150

KEGG: hsa:18

STRING: 9606.ENSP00000268251

UniGene: Hs.336768

Involvement In Disease
GABA transaminase deficiency (GABATD)
Protein Families
Class-III pyridoxal-phosphate-dependent aminotransferase family
Subcellular Location
Mitochondrion matrix.
Tissue Specificity
Liver > pancreas > brain > kidney > heart > placenta.

Q&A

What is ABAT and why is it a significant research target?

ABAT (4-aminobutyrate aminotransferase) is a mitochondrial enzyme with a length of 500 amino acid residues and a molecular mass of 56.4 kDa in humans . Its significance stems from its role as a key enzyme in the metabolism of gamma-aminobutyric acid (GABA), the primary inhibitory neurotransmitter in the mammalian central nervous system. ABAT catalyzes the conversion of gamma-aminobutyrate to succinate semialdehyde and L-beta-aminoisobutyrate to methylmalonate semialdehyde . This enzymatic activity places ABAT at a critical junction in neurotransmitter metabolism, making it relevant to neurological research, particularly in studies of GABAergic signaling, neurological disorders, and mitochondrial function. The protein is widely expressed across multiple tissue types, suggesting diverse physiological roles beyond the nervous system . Research targeting ABAT frequently aims to understand its contribution to neurological disorders, metabolic diseases, and cellular energy homeostasis.

What are the common experimental applications for ABAT antibodies?

ABAT antibodies are employed across multiple experimental platforms in molecular and cellular biology research. The most frequently utilized applications include:

  • Western Blot (WB): The predominant application for ABAT antibodies, allowing researchers to detect and quantify ABAT protein expression in tissue or cell lysates .

  • Immunohistochemistry (IHC): Both paraffin-embedded (IHC-p) and frozen section (IHC-fr) techniques are commonly used to visualize ABAT distribution in tissue specimens .

  • Immunofluorescence (IF) and Immunocytochemistry (ICC): These techniques enable subcellular localization studies of ABAT, confirming its mitochondrial positioning and potential interactions with other proteins .

  • Flow Cytometry (FCM): Some ABAT antibodies are validated for flow cytometry, facilitating quantitative analysis of ABAT expression in cell populations .

  • Immunoprecipitation (IP): Several antibodies are suitable for pulling down ABAT protein complexes to study protein-protein interactions .

These applications provide complementary approaches to investigate ABAT expression, localization, and function in various experimental systems, offering researchers flexibility in experimental design based on their specific research questions.

What species reactivity should be considered when selecting an ABAT antibody?

Species reactivity is a critical consideration when selecting an ABAT antibody for cross-species studies or when working with animal models. ABAT demonstrates high evolutionary conservation, with orthologs identified in multiple species including mouse, rat, bovine, frog, zebrafish, chimpanzee, and chicken . The majority of commercially available ABAT antibodies demonstrate reactivity to human, mouse, and rat ABAT proteins . This broad cross-reactivity reflects the conserved nature of ABAT across mammalian species.

When designing experiments involving less common model organisms, researchers should carefully verify antibody reactivity. Some antibodies offer extended reactivity profiles including rabbit, bovine, dog, goat, guinea pig, horse, and zebrafish . For comparative studies across evolutionary distant species, sequence alignment analysis between the antibody's immunogen sequence and the target species' ABAT sequence is recommended to predict potential cross-reactivity before experimental validation.

The following table summarizes typical reactivity patterns observed in commercially available ABAT antibodies:

Species GroupCommon ReactivityLess Common Reactivity
MammalsHuman, Mouse, RatRabbit, Bovine, Dog, Goat, Guinea Pig, Horse
BirdsChickenVaries by antibody
FishZebrafishVaries by antibody
AmphibiansFrogVaries by antibody

Selecting antibodies with validated reactivity to your species of interest ensures reliable experimental outcomes and valid cross-species comparisons.

How can epitope selection impact ABAT antibody performance in different experimental contexts?

Epitope selection significantly influences ABAT antibody performance across experimental platforms. ABAT antibodies are generated against different regions of the protein, including N-terminal, middle region, and C-terminal epitopes . The effectiveness of these antibodies varies based on the structural accessibility of these epitopes in different experimental contexts.

For Western blot applications, antibodies targeting linear epitopes often perform effectively since proteins are denatured during sample preparation . Commercial antibodies targeting the middle region of ABAT (amino acids approximately 100-400) frequently demonstrate robust signal detection in Western blot applications . In contrast, applications involving native protein conformations, such as immunoprecipitation or certain immunohistochemistry protocols, benefit from antibodies recognizing accessible surface epitopes in the folded protein.

A significant consideration for ABAT research is the protein's mitochondrial localization. Antibodies targeting epitopes that become inaccessible upon mitochondrial import may show differential performance between applications examining whole-cell lysates versus intact mitochondria. Additionally, post-translational modifications can mask epitopes; ABAT undergoes phosphorylation and other modifications that potentially alter antibody binding efficiency.

Researchers should also consider potential cross-reactivity with ABAT isoforms or related aminotransferase family members. Epitopes in highly conserved catalytic domains may cross-react with other Class-III pyridoxal-phosphate-dependent aminotransferases, whereas antibodies targeting unique regions of ABAT offer higher specificity.

For comprehensive ABAT research, employing multiple antibodies targeting different epitopes provides validation through convergent results and mitigates the limitations inherent to any single antibody.

What are the critical considerations for optimization of immunohistochemical detection of ABAT in neural tissues?

Immunohistochemical detection of ABAT in neural tissues requires methodological optimization due to the protein's mitochondrial localization and the complex architecture of neural tissue. Several critical parameters influence successful detection:

  • Fixation Protocol: ABAT's mitochondrial localization necessitates careful fixation optimization. Standard 4% paraformaldehyde fixation may be insufficient for adequate mitochondrial preservation and epitope accessibility. A combination approach using glutaraldehyde (0.1-0.5%) with paraformaldehyde can better preserve ultrastructure while maintaining antigenicity .

  • Antigen Retrieval Methods: Heat-induced epitope retrieval using citrate buffer (pH 6.0) or Tris-EDTA buffer (pH 9.0) significantly enhances ABAT immunoreactivity in paraffin-embedded neural tissues. The optimal retrieval method depends on the specific epitope targeted by the antibody .

  • Section Thickness and Permeabilization: For adequate antibody penetration, section thickness between 5-10 μm is recommended. Enhanced permeabilization using Triton X-100 (0.2-0.5%) or saponin (0.01-0.05%) facilitates antibody access to mitochondrial compartments .

  • Signal Amplification Systems: The relatively low abundance of ABAT in certain neural regions may require signal amplification. Tyramide signal amplification or polymer-based detection systems enhance sensitivity without increasing background .

  • Co-localization Studies: Paired immunostaining with mitochondrial markers (such as COX IV or TOMM20) and cell-type specific markers (such as NeuN for neurons or GFAP for astrocytes) provides contextual information about ABAT expression patterns .

When optimizing protocols, sequential testing of individual parameters while maintaining others constant allows systematic identification of optimal conditions. Negative controls lacking primary antibody and positive controls using tissues with known high ABAT expression (kidney, liver) should be included in all experiments. Cross-validation using multiple ABAT antibodies targeting different epitopes provides additional verification of specificity.

How do post-translational modifications affect ABAT antibody binding and experimental outcomes?

Post-translational modifications (PTMs) of ABAT significantly impact antibody recognition and experimental interpretations. ABAT undergoes several types of PTMs that can mask epitopes, alter protein conformation, or create novel epitopes, thereby affecting antibody binding efficiency.

Phosphorylation represents a major regulatory PTM for ABAT, with multiple serine, threonine, and tyrosine residues serving as potential phosphorylation sites. These modifications can induce conformational changes that either expose or conceal antibody binding sites . Additionally, ABAT undergoes acetylation, particularly at lysine residues, which can neutralize positive charges and potentially disrupt antibody-epitope interactions.

Researchers investigating ABAT PTMs should consider the following approaches:

  • Epitope-specific antibodies: Select antibodies whose epitopes do not contain known PTM sites when absolute quantification is required.

  • PTM-specific antibodies: When available, phospho-specific or acetyl-specific ABAT antibodies can detect specific modified forms.

  • Multiple detection methods: Complement immunological detection with mass spectrometry to identify and characterize PTMs.

  • Enzymatic treatments: Phosphatase treatment of samples prior to antibody application can remove phosphorylation-dependent epitope masking.

The table below summarizes major PTMs of ABAT and their potential impact on antibody recognition:

PTM TypeKnown/Predicted SitesPotential Impact on Antibody Binding
PhosphorylationSer137, Thr325, Tyr141Conformational changes, epitope masking
AcetylationLys374, Lys410Charge neutralization, epitope alteration
OxidationCys residuesStructural changes affecting tertiary structure
Proteolytic processingN-terminal mitochondrial targeting sequenceAltered N-terminal epitope accessibility

Understanding these PTM-dependent effects is essential for accurate interpretation of experimental results, particularly in studies examining ABAT regulation under different physiological or pathological conditions.

What controls should be implemented when validating ABAT antibodies for specific applications?

Comprehensive validation of ABAT antibodies requires systematic implementation of positive and negative controls to ensure specificity, sensitivity, and reproducibility. A robust validation framework should include:

  • Positive Controls:

    • Cell/tissue lysates with known high ABAT expression: Liver, kidney, and brain tissues demonstrate consistent ABAT expression and serve as reliable positive controls .

    • Recombinant ABAT protein: Purified recombinant protein provides a defined positive control with known concentration for sensitivity assessment.

    • Overexpression systems: Cells transfected with ABAT expression plasmids establish a controlled high-expression system.

  • Negative Controls:

    • Antibody omission: Samples processed without primary antibody identify non-specific secondary antibody binding.

    • Isotype controls: Non-relevant antibodies of the same isotype and concentration identify non-specific binding due to Fc receptor interactions.

    • ABAT-depleted samples: Cells treated with validated ABAT siRNA/shRNA or ABAT-knockout tissues created via CRISPR/Cas9 provide definitive negative controls.

  • Specificity Controls:

    • Peptide competition assays: Pre-incubation of antibody with immunizing peptide should eliminate specific signal.

    • Cross-reactivity assessment: Testing antibody against related aminotransferase family members evaluates potential off-target binding.

  • Technical Controls:

    • Multiple antibody comparison: Using different antibodies targeting distinct ABAT epitopes provides convergent validation.

    • Multi-platform validation: Confirming results across different applications (WB, IHC, IF) strengthens confidence in specificity.

    • Batch consistency testing: Evaluating lot-to-lot variability ensures reproducibility across experiments.

Systematic implementation of these controls should be documented with appropriate images and quantification in supplementary materials of publications. Researchers should report all validation steps, including conditions where antibodies failed to perform as expected, to advance community knowledge about reagent limitations.

How can researchers optimize Western blot protocols for maximum specificity and sensitivity when detecting ABAT?

Optimizing Western blot protocols for ABAT detection requires attention to several critical parameters to achieve maximum specificity and sensitivity:

  • Sample Preparation:

    • Include protease inhibitors to prevent ABAT degradation during extraction.

    • Add phosphatase inhibitors if phosphorylated forms of ABAT are of interest.

    • For mitochondrial proteins like ABAT, specialized extraction buffers containing digitonin (0.2-0.5%) or moderate concentrations of Triton X-100 (0.5-1%) improve solubilization .

    • Heat samples at 95°C for 5 minutes in reducing sample buffer containing DTT or β-mercaptoethanol to ensure complete denaturation.

  • Gel Electrophoresis:

    • Use 10-12% acrylamide gels for optimal resolution around ABAT's 56.4 kDa molecular weight .

    • Consider gradient gels (4-15%) when examining potential ABAT isoforms or processing variants.

    • Load appropriate protein amounts: 20-30 μg total protein from tissue lysates or 10-15 μg from mitochondrial-enriched fractions.

  • Transfer Conditions:

    • For ABAT's molecular weight, semi-dry transfer at 15V for 30-45 minutes or wet transfer at 30V overnight at 4°C provides efficient transfer.

    • Use PVDF membranes for higher protein binding capacity and signal-to-noise ratio compared to nitrocellulose.

  • Blocking and Antibody Incubation:

    • 5% non-fat dry milk in TBST generally provides effective blocking for ABAT detection.

    • For phospho-specific detection, 5% BSA in TBST is preferred to avoid phosphatases in milk.

    • Primary antibody dilutions typically range from 1:500 to 1:2000 depending on the specific antibody .

    • Overnight incubation at 4°C often yields better signal-to-noise ratio than short incubations at room temperature.

  • Detection Optimization:

    • Enhanced chemiluminescence (ECL) detection systems with intermediate sensitivity are generally sufficient for ABAT detection in most samples.

    • For low-abundance samples, consider fluorescent secondary antibodies and detection, which often provide better quantitative linearity.

The table below summarizes troubleshooting approaches for common Western blot issues with ABAT detection:

IssuePotential CauseSolution
No signalInsufficient protein extractionUse stronger lysis buffers with appropriate detergents
Multiple bandsIsoforms, degradation, or PTMsInclude protease inhibitors; validate with additional antibodies
High backgroundNon-specific bindingIncrease blocking time; optimize antibody dilution; include 0.05% Tween-20 in wash buffers
Incorrect MW bandNon-specific binding or degradationVerify with positive controls; include protease inhibitors

Implementing these optimization steps systematically improves both the specificity and sensitivity of ABAT detection in Western blot applications.

What methodological approaches enable accurate quantification of ABAT expression in comparative studies?

Accurate quantification of ABAT expression in comparative studies requires rigorous methodological approaches to control for technical and biological variability. The following strategies enable reliable quantitative analysis:

  • Standardized Sample Processing:

    • Collect samples under consistent conditions, considering circadian variations in ABAT expression.

    • Process all comparative samples simultaneously using identical protocols.

    • For tissue samples, consistent anatomical sampling is critical, as ABAT expression varies across brain regions and tissue types .

  • Multi-platform Validation:

    • Combine protein-level quantification (Western blot, ELISA) with transcript-level analysis (qPCR, RNA-seq) to distinguish between transcriptional and post-transcriptional regulation.

    • When available, enzymatic activity assays provide functional validation of ABAT expression differences.

  • Western Blot Quantification Optimization:

    • Use gradient loading series to establish linear dynamic range for ABAT detection.

    • Implement digital image acquisition with exposure optimization to avoid signal saturation.

    • Normalize ABAT signal to appropriate loading controls: total protein stains (REVERT, Ponceau S) are preferred over housekeeping proteins, which may vary under experimental conditions .

    • For mitochondrial proteins like ABAT, normalization to mitochondrial markers (VDAC, COX IV) provides context for changes relative to mitochondrial content.

  • Immunohistochemical Quantification:

    • Employ stereological approaches for tissue-level quantification to account for sampling bias.

    • Use automated image analysis with consistent thresholding parameters across all comparative samples.

    • For fluorescence-based quantification, include calibration standards to convert intensity values to absolute units.

  • Statistical Considerations:

    • Calculate sample sizes based on power analysis using preliminary data on ABAT expression variability.

    • Account for potential covariates (age, sex, medication status) in statistical models.

    • Report effect sizes alongside p-values to indicate biological significance.

The table below summarizes recommended quantification methods based on research question type:

Research QuestionRecommended Primary MethodValidation MethodSpecial Considerations
Tissue expression comparisonWestern blot with total protein normalizationRNAscope or qPCRAccount for mitochondrial content differences
Cellular localization changesHigh-resolution confocal microscopy with co-localization analysisSubcellular fractionationQuantify co-localization coefficients
Response to interventionsELISA or automated Western blot systemsEnzymatic activity assayInclude time-course analysis
Disease-associated changesMultiplexed immunofluorescenceProteomicsMatch cases/controls for confounding variables

By implementing these methodological approaches, researchers can achieve accurate, reproducible quantification of ABAT expression changes across experimental conditions or disease states.

How should researchers interpret unexpected banding patterns in Western blots for ABAT?

Unexpected banding patterns in ABAT Western blots require systematic interpretation to distinguish between biological significance and technical artifacts. The canonical human ABAT protein appears at approximately 56.4 kDa, but several scenarios may produce alternative patterns :

  • Multiple Bands: The appearance of multiple bands may represent:

    • Post-translational modifications: Phosphorylated ABAT may appear as higher molecular weight bands (typically 2-3 kDa shift per phosphorylation site).

    • Alternative splicing: ABAT has known splice variants that may produce proteins of different molecular weights.

    • Processing intermediates: As a mitochondrial protein, ABAT undergoes cleavage of its N-terminal targeting sequence, potentially producing bands of different sizes during processing .

    • Degradation products: Incomplete protease inhibition can result in lower molecular weight fragments.

  • Higher Molecular Weight Bands: Bands significantly above 56.4 kDa may indicate:

    • Oligomeric complexes: Incomplete denaturation can preserve ABAT dimers or tetramers.

    • Covalent modifications: Ubiquitination or SUMOylation can add substantial molecular weight.

    • Cross-reactivity: Antibodies may recognize structurally similar proteins from the Class-III pyridoxal-phosphate-dependent aminotransferase family .

  • Lower Molecular Weight Bands: Bands below 56.4 kDa may represent:

    • Proteolytic cleavage: Physiological or artifactual proteolysis during sample preparation.

    • Alternative translation initiation: Internal start sites may generate N-terminally truncated proteins.

    • Cross-reactivity with related family members of smaller size.

To systematically interpret these patterns, researchers should:

  • Compare banding patterns across multiple antibodies targeting different ABAT epitopes.

  • Perform peptide competition assays to determine which bands are specifically recognized by the antibody.

  • Include positive controls (recombinant ABAT, tissues with known high expression).

  • Verify with knockout/knockdown samples to identify which bands disappear with ABAT depletion.

The table below provides a troubleshooting guide for common unexpected banding patterns:

PatternPossible CausesVerification Approach
Multiple bands around 56.4 kDaPhosphorylation, acetylationPhosphatase/deacetylase treatment of samples
High MW band (~110-120 kDa)Dimerization, cross-linkingIncreased reducing agent concentration; stronger denaturation
Multiple lower MW bandsDegradationFresh sample preparation with additional protease inhibitors
Unexpected band pattern across samplesSample overheating, lane contaminationRepeat with fresh samples and careful loading

Through systematic investigation, researchers can determine whether unexpected banding patterns represent biologically meaningful ABAT variants or technical artifacts requiring optimization.

What approaches help resolve discrepancies between different antibodies when studying ABAT expression?

Resolving discrepancies between different ABAT antibodies requires a systematic investigative approach that addresses potential sources of variation and establishes convergent validity. When antibodies yield contradictory results, consider the following resolution strategies:

  • Epitope Mapping Analysis:

    • Identify the exact epitopes recognized by each antibody through manufacturer information or epitope mapping.

    • Epitopes in regions subject to alternative splicing or post-translational modifications may explain differential detection .

    • Antibodies targeting conformational epitopes versus linear epitopes may perform differently across applications.

  • Multi-platform Validation:

    • Test all antibodies across multiple techniques (WB, IHC, IF) to identify application-specific discrepancies.

    • Complementary non-antibody techniques (mass spectrometry, RNA analysis) can resolve protein expression questions independent of antibody limitations.

    • Enzymatic activity assays provide functional validation of ABAT presence that circumvents antibody specificity issues .

  • Knockout/Knockdown Validation:

    • CRISPR/Cas9 knockout or siRNA knockdown of ABAT provides definitive samples to test antibody specificity.

    • Antibodies showing persistent signal in knockout/knockdown samples likely exhibit off-target binding.

    • Partial signal reduction in knockdown samples helps quantify the proportion of specific versus non-specific binding.

  • Cross-reactivity Assessment:

    • Test antibodies against recombinant ABAT and related family members to identify potential cross-reactivity.

    • Pre-adsorption with related proteins can improve specificity of problematic antibodies.

  • Technical Optimization:

    • Systematically test different fixation/extraction conditions for each antibody.

    • Optimize antibody concentration through titration experiments to determine optimal signal-to-noise ratio.

    • Adjust incubation conditions (time, temperature, buffer composition) for each antibody individually.

When persistent discrepancies remain after systematic investigation, researchers should:

By implementing these resolution strategies, researchers can establish confidence in ABAT expression patterns despite initial antibody discrepancies, ultimately strengthening the validity of their findings.

How can researchers effectively compare ABAT expression across diverse experimental models and disease states?

Effective comparison of ABAT expression across diverse experimental models and disease states requires rigorous standardization and contextual interpretation. To ensure valid cross-model comparisons, researchers should implement the following methodological approaches:

  • Standardized Detection Methodology:

    • Employ identical antibodies, dilutions, and detection protocols across all experimental groups.

    • Process and analyze all samples simultaneously to minimize batch effects.

    • Include calibration standards (recombinant ABAT at known concentrations) to enable absolute quantification.

    • Leverage automated Western blot systems or ELISA when available to reduce technical variability .

  • Contextual Normalization Strategies:

    • For human samples with variable mitochondrial content, normalize ABAT to mitochondrial markers (VDAC, COX IV) rather than general housekeeping proteins.

    • In neurodegenerative conditions or models, account for neuronal loss by normalizing to neuron-specific markers.

    • Consider cell-type specific normalization in heterogeneous tissues, as ABAT expression varies between neurons and glia .

  • Multi-level Analysis Approach:

    • Integrate protein quantification with transcript analysis to distinguish transcriptional from post-translational regulation.

    • Include enzymatic activity measurements to correlate expression levels with functional outcomes.

    • Employ spatial analysis methods (immunohistochemistry, in situ hybridization) to capture region-specific changes that might be diluted in whole-tissue analyses.

  • Statistical Considerations for Cross-model Comparisons:

    • Employ mixed-effects models to account for both within-group and between-group variability.

    • Use appropriate transformations when comparing across species with different baseline ABAT expression levels.

    • Report standardized effect sizes (Cohen's d, Hedges' g) alongside p-values to facilitate cross-study comparisons.

  • Contextual Interpretation Framework:

    • Interpret ABAT changes in the context of related metabolic enzymes within the GABA shunt pathway.

    • Consider compensatory mechanisms that may emerge in chronic models versus acute manipulations.

    • Account for known species differences in GABA metabolism when comparing across animal models .

The table below summarizes important considerations for cross-model ABAT analysis:

Experimental ComparisonKey ConsiderationsRecommended Approaches
Across speciesEvolutionary differences in ABAT structure and regulationFocus on relative changes; use species-specific antibodies when possible
Across disease modelsDifferent pathogenic mechanisms may affect ABAT differentlyIntegrate with pathway analysis; consider disease progression timeline
Developmental studiesABAT expression changes during developmentAge-matched controls; developmental trajectory analysis rather than single timepoints
Drug/intervention studiesDirect effects on ABAT versus indirect metabolic effectsInclude washout periods; dose-response relationships; mechanistic validation

Through implementation of these standardized approaches, researchers can generate valid comparisons of ABAT expression across diverse experimental paradigms, enhancing translational relevance of findings from model systems to human disease states.

What emerging methodologies are enhancing ABAT antibody applications in neuroscience research?

Emerging methodologies are significantly expanding the utility and precision of ABAT antibody applications in neuroscience research. These advanced approaches are enabling deeper insights into ABAT's role in neural function and pathology:

  • Super-resolution Microscopy: Techniques like STORM, PALM, and Expansion Microscopy overcome the diffraction limit of conventional microscopy, allowing visualization of ABAT's precise subcellular localization within mitochondrial compartments . These approaches reveal previously undetectable details of ABAT distribution at synapses and in relation to GABA transporters and receptors.

  • Proximity Labeling Proteomics: Approaches such as BioID or APEX2 fused to ABAT enable identification of proximal interacting proteins in living cells. When combined with ABAT antibodies for validation, these methods map the dynamic ABAT interactome under various physiological and pathological conditions.

  • Multiplexed Immunofluorescence: Technologies like Cyclic Immunofluorescence (CycIF), CO-Detection by indEXing (CODEX), or imaging mass cytometry enable simultaneous detection of ABAT alongside dozens of other proteins in single tissue sections . This multiplexed approach contextualizes ABAT expression within specific cell types and signaling pathways.

  • Single-cell Proteomics: Emerging platforms combining flow cytometry with mass spectrometry enable quantification of ABAT protein levels in individual cells, revealing cell-to-cell variability masked in bulk tissue analyses.

  • CRISPR-based Tagging: CRISPR knock-in of fluorescent tags or epitope tags to endogenous ABAT enables live-cell imaging and antibody-based purification without overexpression artifacts. When combined with ABAT antibodies for validation, these approaches offer unprecedented specificity.

  • Extracellular Vesicle Analysis: Specialized protocols for isolating neuronal extracellular vesicles, combined with sensitive ABAT antibody detection methods, are revealing potential roles for ABAT in intercellular communication and as biomarkers in biological fluids .

These methodological advances are collectively transforming ABAT research from static expression analyses to dynamic, systems-level understanding of its roles in neural metabolism and signaling. Researchers integrating these emerging technologies with validated ABAT antibodies are positioned to make significant discoveries about GABA metabolism in both normal function and neurological disorders.

How should researchers integrate ABAT antibody findings with other experimental approaches for comprehensive pathway analysis?

Integrating ABAT antibody-based findings with complementary experimental approaches enables comprehensive pathway analysis and contextual interpretation of results. A multi-modal integration strategy includes:

  • Functional Enzymatic Analysis:

    • Complement antibody-detected ABAT expression with direct measurement of enzymatic activity using radiolabeled substrates or spectrophotometric assays.

    • Correlate protein levels with functional capacity to identify post-translational regulatory mechanisms.

    • Assess substrate availability and product formation rates to place ABAT activity within the broader GABA shunt pathway context .

  • Metabolomic Integration:

    • Pair ABAT protein quantification with targeted metabolomics of GABA, glutamate, succinate semialdehyde, and related metabolites.

    • Use stable isotope tracing to track carbon flux through ABAT-mediated reactions in cellular or animal models.

    • Correlate ABAT expression changes with metabolite alterations to establish functional consequences of expression differences .

  • Transcriptomic Correlation:

    • Analyze co-expression networks from RNA-seq data to identify genes consistently regulated alongside ABAT.

    • Compare transcript and protein levels to distinguish transcriptional from post-transcriptional regulation.

    • Examine transcription factor binding at the ABAT promoter to elucidate upstream regulatory mechanisms.

  • Genetic Manipulation Approaches:

    • Use conditional knockout/knockdown models to determine the consequences of ABAT depletion on pathway function.

    • Employ rescue experiments with wild-type versus mutant ABAT to establish structure-function relationships.

    • Correlate natural genetic variations in ABAT (from GWAS or sequencing studies) with expression and function.

  • Computational Modeling:

    • Integrate experimental data into computational models of GABA metabolism.

    • Use flux balance analysis to predict the system-wide impact of observed ABAT expression changes.

    • Simulate perturbations to guide experimental design for testing model predictions.

  • Translation to Human Studies:

    • Correlate findings from model systems with human tissue samples when available.

    • Examine ABAT in accessible human samples (fibroblasts, induced neurons) from patients with relevant neurological disorders.

    • Consider potential biomarker applications by assessing ABAT in extracellular vesicles from biological fluids .

This integrative approach transforms isolated ABAT antibody findings into mechanistic insights by establishing relationships between ABAT expression, activity, and broader metabolic consequences. The resulting comprehensive pathway analysis provides context for interpreting ABAT alterations in developmental processes, stress responses, and disease states.

What reference data should researchers consult when designing ABAT antibody experiments?

Researchers designing ABAT antibody experiments should consult several categories of reference data to optimize experimental design and interpretation. The following table compiles essential reference information:

Table 1: ABAT Protein Reference Data

ParameterHuman ABATMouse ABATRat ABAT
Amino Acid Length500 aa 500 aa500 aa
Molecular Weight56.4 kDa 56.3 kDa56.4 kDa
UniProt IDP80404P61922P50554
Gene ID (NCBI)1826886079251
Isoforms2 confirmed1 confirmed1 confirmed
Key PTM sitesPhospho: S137, T325, Y141Phospho: S137, T325Phospho: S137, T325
Subcellular LocalizationMitochondria MitochondriaMitochondria

Table 2: Tissue Expression Hierarchy (Relative Expression Levels)

RankHumanMouseRat
HighLiver, Kidney, Brain Liver, Kidney, BrainLiver, Kidney, Brain
MediumHeart, Skeletal MuscleHeart, Skeletal MuscleHeart, Skeletal Muscle
LowLung, SpleenLung, ThymusLung, Thymus
VariableBrain regions (highest in substantia nigra, hypothalamus) Brain regions (highest in substantia nigra)Brain regions (highest in substantia nigra)

Table 3: Optimal Sample Preparation Methods for Different Applications

ApplicationRecommended FixativeBuffer SystemSpecial Considerations
Western BlotN/ARIPA or NP-40 with protease inhibitorsInclude phosphatase inhibitors for PTM analysis
IHC-Paraffin4% PFA, 24-48hCitrate (pH 6.0) or Tris-EDTA (pH 9.0) retrievalOptimize antigen retrieval time (15-30 min)
IHC-Frozen4% PFA, 15-30 minPBS with 0.1-0.3% Triton X-100Cryoprotect with 30% sucrose
ICC/IF4% PFA, 10-15 minPBS with 0.1% Triton X-100Co-stain with mitochondrial markers
Flow Cytometry2% PFA, 15 minSaponin permeabilization (0.1%)Requires careful validation

Table 4: Common Cross-Reactivity Concerns

Related ProteinSequence Similarity to ABATCommon Cross-ReactivityDistinguishing Features
GCAT (glycine C-acetyltransferase)Moderate (~30% identity)OccasionalDifferent MW (45 kDa)
AGXT (alanine-glyoxylate aminotransferase)Low-moderate (~25% identity)RareDifferent subcellular localization (peroxisomal)
OAT (ornithine aminotransferase)Low-moderate (~25% identity)RareDifferent MW (49 kDa)

By consulting these reference tables before designing experiments, researchers can select appropriate antibodies, optimize protocols for specific applications, and anticipate potential pitfalls in ABAT detection. This information provides a foundation for experimental design that maximizes specificity and sensitivity in ABAT research.

What quantitative benchmarks can validate ABAT antibody specificity across experimental systems?

Establishing quantitative benchmarks for ABAT antibody validation ensures reliable and reproducible results across experimental systems. The following criteria provide a standardized framework for comprehensive antibody validation:

Table 5: Quantitative Validation Benchmarks for ABAT Antibodies

Validation ParameterMinimum Acceptable BenchmarkPreferred BenchmarkMethods of Assessment
Specificity
Signal reduction in knockdown>70% reduction>90% reductionWestern blot densitometry compared to control samples
Signal elimination in knockoutComplete absence of specific bandComplete absence of specific bandWestern blot comparison to wild-type samples
Peptide competition>80% signal reduction>95% signal reductionPre-incubation with immunizing peptide at 5-10× antibody concentration
Sensitivity
Limit of detection (recombinant)<10 ng pure protein<1 ng pure proteinSerial dilution of recombinant ABAT
Endogenous detectionVisible in 10-20 μg total protein from high-expressing tissuesVisible in 5-10 μg total proteinWestern blot with protein titration
Reproducibility
Intra-assay coefficient of variation<15%<10%Triplicate measurements within same experiment
Inter-assay coefficient of variation<20%<15%Measurements across ≥3 independent experiments
Lot-to-lot consistency>85% signal correspondence>90% signal correspondenceDirect comparison of antibody lots on identical samples
Application Performance
Western blot band claritySingle predominant band at 56.4 kDa Single predominant band with <5% non-specific bandsVisual assessment and densitometry
IHC background-to-specific signal ratio>3:1>5:1Quantitative image analysis
IP efficiency>30% target protein recovery>50% target protein recoveryCompare input to immunoprecipitated fraction

Table 6: Tissue-Specific ABAT Signal Intensity Benchmarks for Validation

Tissue TypeExpected Relative Signal IntensityCommon Interfering FactorsRecommended Loading Amount
LiverVery High (reference) High protein background, proteases10-15 μg total protein
KidneyHigh (80-90% of liver) High protein background15-20 μg total protein
Brain (whole)Medium-High (60-80% of liver) Regional variability20-30 μg total protein
Substantia NigraHigh (75-85% of liver)Limited tissue availability15-20 μg total protein
CerebellumMedium (40-60% of liver)--25-35 μg total protein
Skeletal MuscleLow-Medium (20-40% of liver)High actin/myosin background30-40 μg total protein
LungLow (10-20% of liver)--40-50 μg total protein

Implementation of these quantitative benchmarks provides objective criteria to evaluate antibody performance. Researchers should document validation results according to these metrics in publications, enhancing transparency and reproducibility in ABAT research. When antibodies fail to meet minimum benchmarks for specific applications, limitations should be clearly acknowledged, and alternative approaches should be considered.

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.