The LRAT Antibody, FITC conjugated, is a fluorescently labeled mouse monoclonal antibody targeting Lecithin Retinol Acyltransferase (LRAT), an enzyme critical for vitamin A metabolism and visual pigment synthesis. This antibody is designed for fluorescence-based detection in techniques such as immunofluorescence (IF), flow cytometry, and enzyme-linked immunosorbent assay (ELISA). The FITC (Fluorescein Isothiocyanate) conjugation enables visualization under fluorescence microscopy, emitting green light at 519 nm .
Antibody Type: Monoclonal IgG1 λ isotype, ensuring specificity for LRAT in human, mouse, and rat samples .
FITC Conjugation: Covalently linked to the antibody via amine groups, enhancing signal brightness and stability .
Target Specificity: Binds to the active site of LRAT, which catalyzes the conversion of vitamin A into retinyl esters for visual pigment regeneration .
Ocular Health: LRAT deficiency causes retinal dystrophy due to impaired retinyl ester synthesis . FITC-conjugated antibodies enable visualization of LRAT in retinal pigmented epithelium, aiding disease modeling .
Cancer Pathways: Reduced LRAT expression correlates with invasive bladder cancer, suggesting a role in tumor suppression . FITC-based assays can monitor LRAT levels in cancer research .
Biodistribution: FITC conjugation may enhance hepatic clearance of labeled antibodies, as observed with FITC-tagged peptides . This highlights the need for careful dosing in in vivo studies.
FITC conjugation involves the covalent attachment of fluorescein isothiocyanate molecules to antibodies, creating fluorescent probes that can be detected through various optical methods. This conjugation occurs under optimized conditions to ensure maximum labeling efficiency while preserving antibody functionality. The primary advantage is that FITC emits a strong green fluorescence (peak excitation ~495 nm, emission ~520 nm) when excited by appropriate wavelengths, enabling sensitive detection of target antigens in flow cytometry, immunofluorescence microscopy, and other fluorescence-based techniques. FITC-conjugated antibodies eliminate the need for additional detection steps required with unconjugated primary antibodies, streamlining experimental workflows and reducing background signal often associated with multiple-step detection methods .
FITC-conjugated antibodies should be stored at 4°C and protected from prolonged light exposure to prevent photobleaching of the fluorochrome . Most manufacturers recommend against freezing these conjugates to avoid potential damage to both the antibody functionality and the fluorescent properties. The buffer formulation typically includes phosphate-buffered saline containing <0.1% sodium azide as a preservative . When properly stored, these conjugates generally maintain activity for at least 12 months. Working stock solutions should be prepared fresh and kept on ice, protected from light during experimental procedures to minimize fluorescence degradation .
The fundamental difference lies in their epitope recognition profiles and production methods:
Polyclonal antibodies (e.g., Rabbit anti-Rat IgG or Goat anti-Rat Ig):
Derived from multiple B-cell lineages in immunized animals
Recognize multiple epitopes on rat immunoglobulins
Offer broader reactivity, often binding to both heavy and light chains
Examples include FITC Goat Anti-Rat Ig (polyclonal) which reacts with rat IgG and IgM
Useful for applications requiring amplified signal due to multiple binding sites
Monoclonal antibodies (e.g., Mouse Anti-Rat IgG1, clone RG11/39.4):
Produced from single B-cell clones
Recognize specific epitopes with high precision
Offer superior specificity for particular immunoglobulin isotypes or subclasses
Provide consistent lot-to-lot reproducibility
Particularly valuable when discriminating between rat immunoglobulin isotypes is critical
The choice between these types depends on the experimental goals and whether broad reactivity or highly specific detection is required.
Cross-reactivity presents a significant challenge when working with multiple species in the same experimental system. Several strategic approaches can mitigate this issue:
Selection of pre-adsorbed antibodies: Some anti-rat antibodies are specifically adsorbed against immunoglobulins from other species to reduce cross-reactivity. For example, the FITC Goat Anti-Rat Ig has been adsorbed with mouse Ig to minimize mouse cross-reactivity, though this adsorption may reduce reactivity with some rat IgM antibodies .
Cross-reactivity testing: Perform thorough validation using appropriate controls. For instance, the FITC Goat Anti-Rat Ig shows little reactivity with rat non-B splenocytes, mouse splenocytes, or human peripheral blood leukocytes, but exhibits weak cross-reactivity with some hamster immunoglobulins .
Alternative detection strategies: When cross-reactivity cannot be eliminated, consider isotype-specific detection. For instance, when rat IgM detection is compromised by mouse Ig adsorption, FITC-conjugated anti-rat IgM mAb G53-238 or FITC-conjugated anti-rat Ig κ light chain mAb MRK-1 may provide better specificity .
Sequential blocking protocols: Implementing blocking steps with unconjugated antibodies from potentially cross-reactive species before adding the FITC-conjugated anti-rat antibody can significantly reduce non-specific binding .
Immunoelectrophoresis validation: Confirm specificity through immunoelectrophoresis, as demonstrated with the Rabbit anti-Rat IgM antibody, which showed a single precipitin arc against anti-Fluorescein, anti-Rabbit Serum, Rat IgM, and Rat Serum without reaction against other rat heavy or light chain proteins .
Successful flow cytometry experiments with FITC-conjugated anti-rat antibodies require careful attention to several parameters:
Signal intensity optimization: Titrate the antibody concentration to determine optimal signal-to-noise ratio. Most FITC-conjugated anti-rat antibodies are effective at ≤1 μg per million cells, but this should be empirically determined for each application .
Spectral overlap considerations: FITC emission overlaps with other commonly used fluorochromes like PE. When designing multi-color panels, appropriate compensation controls must be included to correct for spectral overlap. Single-stained controls with each fluorochrome used in the experiment are essential .
Cell preparation protocols: Proper single-cell suspensions are crucial. For intracellular staining, follow validated fixation/permeabilization protocols such as those outlined for the BD PharmingenTM FITC Mouse Anti-Rat IgG1, which include specific buffer requirements and incubation times .
FcR blocking: To prevent non-specific binding via Fc receptors, implement blocking steps with reagents like purified 2.4G2 antibody (0.2 μg per test) prior to staining .
Surface vs. intracellular detection: When detecting intracellular immunoglobulins, block surface Ig first (e.g., with purified RG11/39.4 mAb) before proceeding with fixation, permeabilization, and intracellular staining .
Sample viability: Include viability dyes compatible with FITC to exclude dead cells, which can bind antibodies non-specifically and generate false-positive results.
F(ab')2 fragments offer distinct advantages in certain research applications compared to whole IgG molecules:
Reduced non-specific binding: F(ab')2 fragments lack the Fc portion responsible for binding to Fc receptors on cells. This significantly reduces background staining in tissues rich in Fc receptor-expressing cells such as macrophages, neutrophils, and B cells .
Tissue penetration efficiency: The smaller size of F(ab')2 fragments (~110 kDa vs. ~150 kDa for whole IgG) enables better penetration into tissues and dense cell aggregates, particularly beneficial in immunohistochemistry and confocal microscopy applications .
Complement activation avoidance: Unlike whole IgG, F(ab')2 fragments do not activate complement, preventing potential artifacts in complement-sensitive assays or when working with live cells .
Species-specific considerations: F(ab')2 fragments of anti-rat antibodies are particularly useful when staining rat tissues, as they avoid binding to endogenous rat Fc receptors while still recognizing the target antigens efficiently .
Signal intensity trade-offs: F(ab')2 fragments typically produce slightly lower signal intensity than whole IgG due to reduced valency (4 binding sites in whole IgG vs. 2 in F(ab')2), which may necessitate higher concentrations for equivalent detection .
The choice between F(ab')2 and whole IgG should be based on the specific requirements of the experimental system, particularly when working with Fc receptor-rich samples or when background reduction is critical.
When utilizing FITC-conjugated anti-rat antibodies for immunohistochemistry, researchers should follow these methodological guidelines:
For Frozen Sections:
Section preparation: Cut 5-8 μm cryostat sections and allow to air dry for 1-2 hours before fixing.
Fixation: Use freshly prepared 4% paraformaldehyde (10 minutes) or acetone (5-10 minutes at -20°C) depending on epitope sensitivity.
Blocking: Apply 5-10% normal serum from the same species as the secondary antibody for 30-60 minutes to reduce non-specific binding.
Primary antibody incubation: If using a two-step method, apply the primary rat antibody at optimized concentration and incubate at 4°C overnight or at room temperature for 1-2 hours.
FITC-conjugated anti-rat antibody application: Apply at 2-10 μg/mL (titration recommended) and incubate in a dark, humidified chamber for 30-60 minutes at room temperature.
Nuclear counterstaining: Use DAPI or Hoechst, ensuring compatibility with FITC fluorescence.
Mounting: Mount with anti-fade mounting medium specifically formulated for fluorescence preservation .
For Paraffin Sections:
Antigen retrieval: This critical step requires optimization based on the target antigen. Common methods include:
Heat-induced epitope retrieval using citrate buffer (pH 6.0) or EDTA buffer (pH 9.0)
Enzymatic retrieval using proteinase K or trypsin
Autofluorescence reduction: Treat with 0.1% sodium borohydride or 0.3% Sudan Black B in 70% ethanol to reduce background autofluorescence common in paraffin-embedded tissues.
Proceed with blocking and staining: Follow steps similar to frozen section protocol but with potentially higher antibody concentrations due to antigen masking during paraffin embedding .
These protocols have been successfully applied in studies of lung eosinophilia, airway hyperresponsiveness in asthma models, and inflammatory responses in cigarette smoke-exposed mice .
Western blot applications using FITC-conjugated anti-rat antibodies require specific adaptations to traditional protocols:
Sample preparation and gel electrophoresis: Standard SDS-PAGE procedures apply, with protein loading typically 10-30 μg per lane depending on target abundance.
Transfer optimization: Use PVDF membranes rather than nitrocellulose when possible, as they provide better protein retention and lower autofluorescence. Transfer at 30V overnight at 4°C to ensure complete transfer of high-molecular-weight proteins.
Blocking considerations: Use 5% BSA in TBS-T rather than milk proteins, as milk can contribute to background fluorescence. Block for 1-2 hours at room temperature or overnight at 4°C.
Primary antibody application: When using a two-step detection method, apply the primary rat antibody at optimized dilution (typically 1:1000 to 1:5000) in blocking buffer and incubate overnight at 4°C.
FITC-conjugated antibody incubation: Dilute according to manufacturer recommendations (typically 1:500 to 1:2000) in blocking buffer and incubate for 1-2 hours at room temperature in the dark. The Rabbit Anti-Rat IgG(H+L)-FITC has been validated for western blot applications at these concentrations .
Washing steps: Perform at least 3-5 washes for 5-10 minutes each with TBS-T, keeping membranes protected from light during washing steps.
Detection methods:
Quantification considerations: Include appropriate loading controls and calibration standards for accurate quantification. For multiplexing with other fluorophores, ensure spectral compatibility and use appropriate filters to avoid bleed-through .
Intracellular staining for flow cytometry presents unique challenges that require specific optimization steps:
Surface marker vs. intracellular marker discrimination: When studying both surface and intracellular markers, complete surface staining before fixation and permeabilization. If the same antibody target exists on both the surface and intracellularly, block surface antigens first as described in the BD Pharmingen protocol .
Fixation and permeabilization optimization:
Use specialized buffers like BD Cytofix/Cytoperm™ for 30 minutes at room temperature
Ensure complete permeabilization by monitoring kinetics with time-course experiments
Recognize that some epitopes may be sensitive to specific fixatives; paraformaldehyde-based fixatives preserve FITC fluorescence better than methanol or acetone-based options
Control selection:
Protocol validation: The detailed protocol from BD Pharmingen provides a validated method:
Block Fcγ receptors with 0.2 μg of purified 2.4G2 antibody
Block surface Ig with purified unconjugated antibody when necessary
Use BD Cytofix/Cytoperm™ buffer for 30 minutes
Wash with specialized Perm/Wash™ buffer before and after antibody staining
Signal amplification strategies: For low-abundance intracellular targets, consider:
Data analysis considerations: When analyzing intracellular staining:
High background signal is a common challenge when using FITC-conjugated antibodies. Systematic troubleshooting approaches include:
Antibody titration: Determine the optimal antibody concentration through systematic titration experiments. Excessive antibody concentrations often contribute to high background. As noted in product documentation: "Since applications vary, each investigator should titrate the reagent to obtain optimal results" .
Fc receptor blocking: Implement effective blocking of Fc receptors, particularly in samples rich in Fc receptor-expressing cells:
Optimization of washing procedures:
Reduce autofluorescence:
Buffer composition adjustments:
Sample-specific considerations:
Comprehensive validation of FITC-conjugated anti-rat antibodies should include:
Specificity verification:
Immunoelectrophoresis confirmation showing reaction patterns with target immunoglobulins only
Testing against a panel of relevant tissues/cells with known expression patterns
Competitive binding assays with unconjugated antibodies of the same clone
Western blot analysis to confirm molecular weight specificity
Performance metrics assessment:
Cross-reactivity evaluation:
Application-specific validation:
Documentation requirements:
The relationship between fluorescence intensity and target abundance is complex and requires careful consideration:
Correlation analysis fundamentals:
FITC fluorescence intensity generally shows a positive correlation with target abundance, but this relationship is rarely perfectly linear
At very high target concentrations, fluorescence may plateau due to steric hindrance or fluorescence quenching effects
At extremely low concentrations, instrument detection limits become the constraining factor
Quantification standardization:
Use calibration beads with known quantities of fluorochrome to convert arbitrary fluorescence units to Molecules of Equivalent Soluble Fluorochrome (MESF)
Implement standardized protocols that include reference standards in each experiment
Account for day-to-day instrument variation through consistent quality control procedures
Technical factors affecting correlation accuracy:
Antibody saturation: Ensure antibody concentration is sufficient to reach binding equilibrium
Fluorophore:protein ratio: Higher F/P ratios may lead to quenching or altered binding
Fixation effects: Different fixation methods can affect epitope availability and fluorescence intensity
Laser power and detector voltage settings must be standardized across experiments
Biological variables impacting quantification:
Relative vs. absolute quantification approaches:
For most applications, relative quantification between experimental groups is sufficient
When absolute quantification is required, consider using calibration standards with known quantities of target molecule
Flow cytometry absolute counting beads can provide quantitative cell counts when combined with fluorescence measurements
The relationship between fluorescence intensity and target abundance should be empirically determined for each experimental system, recognizing that while generally correlative, it requires careful standardization for truly quantitative applications.
Multiplex immunofluorescence using FITC-conjugated anti-rat antibodies requires strategic planning and optimization:
Spectral compatibility planning:
Sequential staining strategies:
When using multiple antibodies from the same host species (e.g., multiple rat primary antibodies):
Tyramide signal amplification (TSA) integration:
For low-abundance targets, combine FITC-conjugated anti-rat antibodies with HRP-conjugated secondaries for other targets
FITC-tyramide can be deposited through HRP-catalyzed reactions for significant signal amplification
This approach allows multiple antibodies from the same species to be used sequentially with heat-mediated antibody stripping between cycles
Cross-talk minimization techniques:
Image acquisition optimization:
For microscopy applications, acquire channels sequentially rather than simultaneously
Begin with longer wavelength fluorophores and end with FITC to minimize photobleaching effects
Standardize exposure settings for quantitative comparisons
Apply consistent background subtraction methods across all channels
These multiplex approaches have been successfully applied in immunohistochemistry studies of lung tissue inflammation and in flow cytometric analysis of complex cell populations from various tissues .
In vivo imaging with FITC-conjugated antibodies presents distinct challenges not encountered in fixed or ex vivo applications:
Biodistribution and pharmacokinetic considerations:
Optical property limitations:
FITC's excitation/emission profile (495/520 nm) falls within a spectral range with significant tissue autofluorescence
Tissue penetration is limited to a few millimeters due to absorption and scattering
Alternative near-infrared fluorophores may be preferable for deep tissue imaging
Surface structures or surgically exposed tissues are most suitable for FITC visualization
Signal-to-background optimization strategies:
Implement dietary modifications to reduce autofluorescence (e.g., alfalfa-free diets)
Consider spectral unmixing algorithms to distinguish specific signal from autofluorescence
Use pre-injection images for background subtraction
Optimize injection timing to allow clearance of unbound antibody (typically 24-48 hours)
Experimental design adaptations:
Ethical and regulatory considerations:
While challenging, in vivo applications of FITC-conjugated antibodies can provide valuable insights into dynamic biological processes when properly optimized.
The fluorescein/protein (F/P) ratio is a critical parameter affecting antibody performance across applications:
F/P ratio significance:
Defines the average number of FITC molecules conjugated to each antibody molecule
Typical commercial preparations range from 3-8 FITC molecules per antibody
Higher F/P ratios increase brightness but may compromise antibody affinity or solubility
Lower F/P ratios maintain native antibody properties but provide reduced signal intensity
Application-specific optimization:
Flow cytometry: Higher F/P ratios (5-8) are generally preferred for maximal sensitivity
Immunohistochemistry: Moderate F/P ratios (3-5) balance brightness with specific binding
Super-resolution microscopy: Lower F/P ratios (2-4) minimize fluorophore proximity effects
FLISA (Fluorescence-Linked Immunosorbent Assay): Higher F/P ratios maximize detection sensitivity
Empirical determination methods:
Spectrophotometric calculation using absorbance ratios at 280 nm (protein) and 495 nm (FITC)
Performance comparison of antibodies with different F/P ratios in the specific application
Competitive binding assays to assess impact of conjugation on affinity
Signal-to-noise ratio optimization across F/P ratio gradients
Technical trade-offs:
Self-quenching occurs when FITC molecules are too densely packed (typically F/P > 8)
Higher F/P ratios may increase hydrophobicity, leading to aggregation or non-specific binding
Over-conjugation can mask critical binding sites, reducing effective affinity
Storage stability generally decreases with increasing F/P ratio
Quality control considerations:
For commercially available FITC-conjugated anti-rat antibodies, the F/P ratio is typically optimized by manufacturers for general applications, but researchers may need to select specific preparations for particular experimental requirements or prepare custom conjugates for specialized applications.
The host species used to generate anti-rat antibodies significantly impacts their performance characteristics:
The selection of host species should be guided by the specific experimental requirements, with consideration of potential cross-reactivity issues, particularly in multi-species systems .
A comparative analysis of commercially available FITC-conjugated anti-rat antibodies reveals important differences that inform selection for specific applications:
This comparison highlights the importance of selecting antibodies based on specific experimental requirements, particularly regarding isotype specificity, cross-reactivity profiles, and validated applications .
FITC-conjugated anti-rat antibodies are finding novel applications in several advanced research methodologies:
Single-cell analysis technologies:
Integration with mass cytometry (CyTOF) workflows where FITC-conjugated antibodies serve as reporters for metal-tagged secondary antibodies
Combination with single-cell RNA sequencing to correlate protein expression with transcriptomic profiles
Application in microfluidic droplet-based assays for high-throughput screening of antibody-secreting cells
Super-resolution microscopy implementations:
Optimization for techniques like Structured Illumination Microscopy (SIM) and Stimulated Emission Depletion (STED)
Development of photoswitchable FITC derivatives for Single-Molecule Localization Microscopy
Integration with expansion microscopy protocols to achieve nanoscale resolution with standard fluorescence microscopes
Live cell imaging advances:
Tissue clearing technologies:
These emerging applications demonstrate the continuing utility of FITC-conjugated antibodies in cutting-edge research, particularly when integrated with complementary advanced technologies.
The integration of FITC-conjugated anti-rat antibodies with these advanced methodologies is enabling new insights in fields ranging from neuroscience to immunology, particularly in complex tissue microenvironments and dynamic cellular systems.