LRAT Antibody, Biotin conjugated (Catalog #NBP2-50444B) is a biotin-labeled monoclonal antibody derived from mouse IgG₁. It targets human LRAT, a key enzyme that catalyzes the esterification of retinol (vitamin A) to retinyl esters for storage in hepatic stellate cells and retinal pigment epithelium .
Biotin conjugation enhances detection sensitivity through streptavidin-biotin binding systems, enabling signal amplification in assays . For LRAT studies, this antibody facilitates:
Quantitative Analysis: ELISA-based measurement of LRAT levels in biological samples .
Localization Studies: IHC and IF for spatial LRAT expression in tissues .
Western Blotting: Detection of LRAT protein bands in lysates .
Primary Antibody Incubation: LRAT-specific antibody binds to target epitopes.
Biotin-Streptavidin Bridging: Streptavidin-enzyme/fluorophore conjugates bind to the biotinylated antibody.
Signal Amplification: Enzymatic (e.g., HRP) or fluorescent reporters amplify detection .
Biotinylated antibodies, including LRAT-targeted variants, are integral to avidin-biotin systems for therapeutic delivery. For example:
Fusion proteins like LDL receptor–avidin constructs enable targeted delivery of biotinylated ligands to specific tissues .
Avidin-biotin nanoparticles enhance drug penetration across biological barriers (e.g., blood-brain barrier) for cancer therapy .
High-Sensitivity Assays: Biotinylated antibodies improve detection limits in ELISA and WB by 8–20x compared to unconjugated counterparts .
Multiplexing: Compatible with streptavidin-functionalized quantum dots (Qdots) for simultaneous detection of multiple biomarkers .
Dilution Range: 2–10 µg/mL for most applications; validate empirically .
Cross-Reactivity Mitigation: Use blocking buffers containing 2% normal serum from the tissue’s host species .
LRAT (Lecithin retinol acyltransferase) is an enzyme that transfers the acyl group from the sn-1 position of phosphatidylcholine to all-trans retinol, producing all-trans retinyl esters which function as storage forms of vitamin A. LRAT plays a critical role in vision by providing the all-trans retinyl ester substrates for the isomerohydrolase that processes these esters into 11-cis-retinol in the retinal pigment epithelium. Through a membrane-associated alcohol dehydrogenase, 11-cis-retinol is oxidized and converted into 11-cis-retinaldehyde, which serves as the chromophore for rhodopsin and cone photopigments . LRAT defects are linked to severe early-onset retinal dystrophy, underscoring its importance in visual function .
The biotin-conjugated LRAT antibody is a polyclonal IgG antibody typically raised in rabbit hosts against recombinant Human Lecithin retinol acyltransferase protein (amino acids 24-182) . The antibody is purified using Protein G affinity purification with purity >95%. The biotin conjugation allows for signal amplification in detection systems, as biotin can be recognized by avidin or streptavidin molecules, each capable of binding four biotin groups, thus enhancing detection sensitivity . The antibody is typically supplied in a buffer containing preservatives (0.03% Proclin 300), 50% glycerol, and 0.01M PBS at pH 7.4 .
While the specifications may vary between manufacturers, LRAT antibodies typically demonstrate reactivity with human samples . Some LRAT antibodies (non-biotin conjugated) have been validated to react with human, mouse, and rat samples . When selecting an LRAT antibody for your research, it is crucial to verify the specific species reactivity of the product you intend to use, especially if working with non-human models.
The biotin-conjugated LRAT antibody has been validated for ELISA applications . The biotin conjugation makes this antibody particularly useful in avidin-biotin or streptavidin-biotin detection systems, where signal amplification can significantly enhance sensitivity. While the biotin-conjugated version is primarily validated for ELISA, unconjugated LRAT antibodies have been used successfully in Western blot applications , suggesting potential cross-application utility with appropriate protocol modifications.
Although the biotin-conjugated LRAT antibody in the search results is primarily validated for ELISA , biotin-conjugated antibodies generally can be utilized in immunohistochemistry through avidin-biotin or streptavidin-biotin detection systems. When adapting the LRAT antibody for IHC, researchers should consider:
Antigen retrieval methods: Optimize based on tissue fixation (FFPE vs. frozen sections)
Blocking endogenous biotin: Pre-treatment with avidin-biotin blocking reagents to prevent non-specific binding
Detection system: Selection between ABC (Avidin-Biotin Complex) or streptavidin systems
Visualization method: DAB, AP, or fluorescent-labeled streptavidin
A recommended starting dilution would be 1:200, with optimization required for specific tissue types and fixation methods .
Biotin-conjugated LRAT antibody enhances detection sensitivity in ELISA assays through the avidin/streptavidin-biotin amplification system. Both avidin and streptavidin are tetrameric proteins capable of binding 4 biotin groups per molecule, creating a significant amplification effect . This amplification mechanism increases the concentration of reporters at the antigenic site, substantially enhancing signal intensity.
For optimized ELISA protocols using biotin-conjugated LRAT antibody:
Coat wells with capture antibody against LRAT
Add sample containing LRAT protein
Add biotin-conjugated LRAT antibody (1:500-1:1000 dilution)
Add streptavidin-HRP or streptavidin-AP conjugate
Add appropriate substrate for signal development
Measure signal intensity using spectrophotometry
This method can achieve significantly higher sensitivity compared to direct detection methods, particularly advantageous when detecting low-abundance LRAT protein in biological samples.
For optimal preservation of biotin-conjugated LRAT antibody activity, store the antibody at -20°C or -80°C upon receipt . Avoid repeated freeze-thaw cycles, as these can lead to protein denaturation and loss of binding activity. For regular use, consider aliquoting the antibody into smaller volumes before freezing to minimize freeze-thaw cycles. The presence of 50% glycerol in the storage buffer helps prevent freeze-thaw damage . For short-term storage (less than one month), the antibody can be kept at 2-8°C . Always centrifuge the antibody vial briefly before opening to ensure all liquid is at the bottom of the tube.
Dilution optimization is crucial for achieving optimal signal-to-noise ratio with biotin-conjugated LRAT antibody. For ELISA applications, typically start with a dilution range of 1:500-1:1000 . For other applications like immunohistochemistry or Western blotting (if attempting to adapt the biotin-conjugated antibody), begin with a dilution of 1:200 and adjust based on signal intensity and background levels .
To determine optimal dilution:
Prepare a dilution series (e.g., 1:100, 1:200, 1:500, 1:1000, 1:2000)
Test each dilution with positive and negative controls
Evaluate based on:
Signal strength with positive control
Signal-to-noise ratio
Background staining with negative control
Select the dilution that provides strong specific signal with minimal background
Remember that optimal dilution may vary between sample types and detection methods.
To ensure experimental validity when working with biotin-conjugated LRAT antibody, incorporate the following controls:
Positive control: Samples known to express LRAT (e.g., retinal pigment epithelium, testis tissue , or Y79 retinoblastoma cells )
Negative control: Samples known not to express LRAT or tissues from LRAT knockout models
Antibody controls:
Isotype control (rabbit IgG with biotin conjugation)
No primary antibody control (to assess non-specific binding of detection reagents)
Biotin-specific controls:
Endogenous biotin blocking control (especially important in biotin-rich tissues)
Streptavidin-only control (without biotin-conjugated antibody)
Peptide competition control: Pre-incubation of antibody with immunizing peptide to verify specificity
These controls help distinguish specific LRAT detection from potential artifacts and validate experimental findings.
Distinguishing between LRAT monomers (26 kDa) and dimers (50-54 kDa) requires careful consideration of sample preparation and analytical techniques:
Sample preparation conditions:
Use of reducing agents (DTT or β-mercaptoethanol) in sample buffers disrupts disulfide bonds, favoring detection of the 26 kDa monomeric form
Non-reducing conditions preserve disulfide-linked dimers, allowing detection of the 50-54 kDa form
Native conditions maintain protein-protein interactions that stabilize dimers
Analytical techniques:
SDS-PAGE with Western blotting using different sample buffer conditions
Native PAGE to preserve native protein-protein interactions
Size exclusion chromatography to separate monomers and dimers based on molecular size
Cross-linking experiments to stabilize transient interactions before analysis
Data interpretation:
This approach provides insights into the functional state of LRAT protein in experimental systems and may reveal important regulatory mechanisms.
When investigating LRAT in retinal disease models, several important considerations should guide experimental design:
Disease relevance:
Model selection:
Technical approaches:
Immunohistochemical localization to assess LRAT distribution in retinal layers
Western blot to quantify LRAT expression levels (monomer vs. dimer ratios)
Functional assays to measure retinyl ester formation activity
Gene expression analysis to investigate regulatory mechanisms
Biomarker correlation:
Correlate LRAT expression/activity with visual function metrics
Assess relationship between LRAT levels and other components of the visual cycle
Determine progression markers for retinal degeneration in relation to LRAT status
This comprehensive approach facilitates mechanistic understanding of LRAT's role in retinal pathology and may identify therapeutic targets.
Biotin conjugation can influence antibody binding kinetics and epitope accessibility in several ways that researchers should consider:
Understanding these factors is essential for accurate interpretation of experimental results and may necessitate protocol adjustments when transitioning from unconjugated to biotin-conjugated LRAT antibodies.
Non-specific binding is a common challenge when working with biotin-conjugated antibodies. To address this issue with biotin-conjugated LRAT antibody:
Endogenous biotin blocking:
Pretreat samples with avidin followed by biotin blocking solutions
This prevents detection reagents from binding to endogenous biotin in tissues
Buffer optimization:
Secondary reagent considerations:
Use streptavidin conjugates with minimal batch-to-batch variation
Pre-absorb streptavidin conjugates against tissues of interest
Titrate streptavidin reagents to minimize background
Sample-specific considerations:
Protocol modifications:
Include additional washing steps with increased detergent concentration
Reduce incubation temperature (4°C overnight instead of room temperature)
Decrease incubation time of detection reagents
These strategies can significantly improve signal-to-noise ratio and experimental reliability.
When studying LRAT using antibody-based techniques, researchers should be aware of several potential pitfalls in data interpretation:
Awareness of these potential pitfalls enables more rigorous experimental design and more accurate interpretation of LRAT research data.
Validating the specificity of biotin-conjugated LRAT antibody is crucial for ensuring reliable research findings. Implement these validation strategies:
Peptide competition assay:
Genetic models:
Test antibody in LRAT-knockout or LRAT-overexpressing systems
Signal should correlate with genetic LRAT expression status
Use siRNA knockdown to create gradient expression levels for validation
Orthogonal detection methods:
Compare antibody-based detection with mRNA expression (RT-PCR, RNA-seq)
Correlate protein detection with enzymatic activity assays
Use multiple antibodies targeting different LRAT epitopes
Cross-reactivity assessment:
Test against recombinant proteins from the H-rev107 family
Evaluate specificity across multiple species if cross-species reactivity is claimed
Perform immunoprecipitation followed by mass spectrometry to identify all bound proteins
Technical validation:
Compare staining/detection patterns between biotin-conjugated and unconjugated antibody versions
Evaluate signal in tissues/cells known to express or lack LRAT
Test different detection systems to confirm consistent results
Comprehensive validation ensures that experimental findings accurately reflect LRAT biology rather than technical artifacts.
Biotin-conjugated LRAT antibody offers significant potential for multiplexed imaging applications through these innovative approaches:
Orthogonal detection systems:
Combine with directly fluorophore-labeled antibodies against other targets
Use streptavidin conjugated to spectrally distinct fluorophores or quantum dots
Implement sequential detection with antibody stripping between rounds
Multi-omics integration:
Couple with RNA detection methods (RNAscope, FISH) for protein-transcript correlation
Combine with proximity ligation assays to detect LRAT protein interactions
Integrate with metabolic labeling to track retinyl ester production
Advanced microscopy applications:
Super-resolution microscopy to resolve LRAT subcellular localization
Live-cell imaging using cell-permeable streptavidin conjugates
FRET-based approaches to detect LRAT-protein interactions
Tissue and disease profiling:
Multiplex with markers of retinal cell types to map LRAT distribution
Compare normal vs. diseased tissue with parallel detection of disease markers
Developmental profiling with stage-specific markers
These approaches would provide unprecedented insights into LRAT's spatial relationships with other proteins and its role in retinal physiology and pathology.
Several emerging technologies hold promise for enhancing LRAT antibody detection systems:
Advanced amplification methods:
Tyramide signal amplification (TSA) with biotin-conjugated LRAT antibody
DNA-barcoded antibodies for ultrasensitive digital counting
Proximity extension assays for highly specific detection
Nanotechnology integration:
Plasmonic nanoparticles for enhanced optical detection
Magnetic nanoparticles for extraction and enrichment of LRAT-expressing cells
Nanobody-based detection systems with improved tissue penetration
Microfluidic applications:
Single-cell LRAT protein quantification platforms
Automated immunostaining systems for improved reproducibility
Organ-on-chip models incorporating LRAT detection
Computational advances:
AI-assisted image analysis for quantitative LRAT expression mapping
Machine learning algorithms to correlate LRAT patterns with disease phenotypes
Integrative multi-omics data analysis frameworks
In vivo applications:
Development of non-invasive imaging for LRAT activity in animal models
Translational biomarkers for retinal diseases based on LRAT status
Therapeutic monitoring systems for interventions targeting the visual cycle
These technological advances will expand the utility of LRAT antibodies beyond current applications and enable new discoveries about LRAT biology and pathology.
LRAT detection using biotin-conjugated antibodies and other advanced methods can significantly contribute to understanding retinal disease mechanisms and therapeutic development:
Disease mechanism elucidation:
Mapping LRAT expression changes in progressive retinal degeneration
Identifying cell type-specific LRAT dysfunction in disease models
Understanding post-translational modifications of LRAT in pathological states
Correlating LRAT distribution with functional visual deficits
Biomarker development:
Establishing LRAT expression patterns as diagnostic or prognostic indicators
Monitoring treatment response through LRAT function restoration
Identifying patient subgroups based on LRAT-related pathology patterns
Developing companion diagnostics for visual cycle-targeting therapies
Therapeutic target validation:
Screening compounds that modulate LRAT expression or activity
Validating gene therapy approaches for LRAT-associated retinal dystrophies
Testing small molecule chaperones to rescue misfolded LRAT variants
Developing enzyme replacement strategies for LRAT deficiency
Regenerative medicine applications:
Assessing LRAT functionality in stem cell-derived retinal pigment epithelium
Quality control for cell replacement therapies
Monitoring integration and function of transplanted cells
Validating tissue-engineered retinal constructs
These applications demonstrate how advanced LRAT detection methods, including biotin-conjugated antibodies, can accelerate both basic understanding of retinal biology and the development of targeted therapeutics for vision-threatening diseases.