LRAT Antibody, FITC conjugated

Shipped with Ice Packs
In Stock

Description

Introduction

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 .

Structure and Mechanism

  • 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 .

Applications

ApplicationDescriptionRelevance
Immunofluorescence (IF)Detects LRAT localization in retinal pigmented epithelium and liver cells .Visualizes enzyme distribution in ocular and hepatic tissues.
Flow CytometryQuantifies LRAT expression in cell populations .Useful for studying LRAT regulation in cancer or retinal diseases.
ELISAMeasures LRAT protein levels in lysates or sera .Facilitates biomarker discovery in ocular health studies.

Research Findings

  • 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.

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship products within 1-3 business days of receiving your order. Delivery times may vary depending on the purchasing method or location. Please consult your local distributors for specific delivery time information.
Synonyms
LRAT; Lecithin retinol acyltransferase; Phosphatidylcholine--retinol O-acyltransferase
Target Names
LRAT
Uniprot No.

Target Background

Function
Lecithin retinol acyltransferase (LRAT) catalyzes the transfer of an acyl group from the sn-1 position of phosphatidylcholine to all-trans retinol, resulting in the production of all-trans retinyl esters. Retinyl esters serve as storage forms of vitamin A. LRAT plays a crucial role in vision. It provides the all-trans retinyl ester substrates for the isomerohydrolase, which processes the esters into 11-cis-retinol in the retinal pigment epithelium. Due to a membrane-associated alcohol dehydrogenase, 11-cis-retinol is oxidized and converted into 11-cis-retinaldehyde, which is the chromophore for rhodopsin and the cone photopigments. LRAT is essential for the survival of cone photoreceptors and the maintenance of correct rod photoreceptor cell morphology.
Gene References Into Functions
  1. Genetic analysis of our proband revealed a novel homozygous mutation on codon 119 of the lecithin-cholesterol acyltransferase gene, leading to the substitution of glycine with aspartate. PMID: 28942093
  2. While the instability of LRAT(E14L) did not completely eliminate the production of the visual chromophore in a cell-based assay, its expression resulted in a rapid increase in cellular levels of retinoic acid upon retinoid supplementation. PMID: 28758396
  3. LRAT hypermethylation was associated with decreased mRNA levels in colorectal cancer clinical specimens. PMID: 25260806
  4. Lecithin retinol acyltransferase influences all-trans retinoic acid levels and plays a role in retinoid sensitivity in malignant melanoma cells. PMID: 25236354
  5. These findings reveal structural adaptations that facilitate selective catalysis and elucidate the mechanism responsible for diverse substrate specificity within the LRAT-like enzyme family. PMID: 25383759
  6. High LRAT expression in melanoma may be critical in removing retinol as a substrate for RA production, thereby triggering signaling pathways leading to dedifferentiation, proliferation, and anti-apoptosis. PMID: 24433184
  7. Functional hepatic stellate cells coexpressing both LRAT and CRBP-1, which retain the ability to store vitamin A, contribute to the development of portal and parenchymal fibrogenesis in patients with viral hepatitis. PMID: 23890161
  8. Lecithin-retinol acyltransferase is a thermostable and highly active enzyme with a likely mode of interfacial activation. PMID: 24613493
  9. A genetic defect in LRAT has been identified as a novel cause of retinitis punctata albescens. PMID: 22559933
  10. Data suggest that acyl-modified forms of HRAS-like tumor suppressors HRASLS2 and HRASLS3 mimic the lipolytic activity of lecithin retinol acyltransferase (LRAT). PMID: 22605381
  11. LRAT mutations cause a severe, early childhood onset, progressive retinal dystrophy. PMID: 22570351
  12. This study demonstrated that malignant melanoma cells can esterify all-trans retinol and subsequently isomerize all-trans retinyl esters (RE) into 11-cis retinol, whereas their benign counterparts, melanocytes, are unable to catalyze these reactions. PMID: 21465477
  13. Data indicate that overexpression of human LRAT specifically in mice oral basal epithelial cells makes these cells more sensitive to carcinogen-induced tumorigenesis. PMID: 19471114
  14. LRAT expression is higher in renal tumors with an indolent biological behavior. PMID: 14581364
  15. Conserved residues Cys-161 and His-60 form the essential catalytic dyad of LRAT, representing a novel thiol protease motif that functions in an acyltransferase reaction. PMID: 14596594
  16. LRAT plays a role in preventing the progression of invasive bladder cancer. PMID: 15161698
  17. Results provide evidence that multiple LRAT mRNA transcripts, which are expressed in a tissue-specific manner, may result from differential splicing of the 5'UTR region and the use of multiple polyadenylation signals in the 3'UTR. PMID: 15474300
  18. LRAT has a role in retinoid absorption and storage. PMID: 16115871
  19. These experiments are consistent with an expanded role for LRAT function as a protein palmitoyl transferase. PMID: 16939223
  20. The phenotype of patients with mutations in LRAT is similar to that of patients with mutations in RPE65, suggesting the need to systematically screen both genes in cases of typical phenotype. PMID: 17011878
  21. LRAT mutations are likely a rare cause of Leber congenital amaurosis among patients from North America. PMID: 17438524
  22. Lecithin: retinol acyltransferase protein is distributed in both hepatic stellate cells and endothelial cells of normal rodent and human liver. PMID: 18544127
  23. Transcriptional regulation is aberrant in human prostate cancer and involves GATA transcription factors in normal prostate epithelial cells. PMID: 18652909
  24. The proximal region, together with basal transcription factors, may be sufficient to drive Lrat expression. PMID: 19665987

Show More

Hide All

Database Links

HGNC: 6685

OMIM: 604863

KEGG: hsa:9227

STRING: 9606.ENSP00000337224

UniGene: Hs.658427

Involvement In Disease
Leber congenital amaurosis 14 (LCA14)
Protein Families
H-rev107 family
Subcellular Location
Endoplasmic reticulum membrane; Single-pass membrane protein. Rough endoplasmic reticulum. Endosome, multivesicular body. Cytoplasm, perinuclear region.
Tissue Specificity
Hepatic stellate cells and endothelial cells (at protein level). Found at high levels in testis and liver, followed by retinal pigment epithelium, small intestine, prostate, pancreas and colon. Low expression observed in brain. In fetal tissues, expressed

Q&A

What is the principle behind FITC conjugation to antibodies and how does it benefit immunological research?

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 .

How should FITC-conjugated anti-rat antibodies be stored to maintain optimal performance?

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 .

What are the differences between polyclonal and monoclonal FITC-conjugated anti-rat antibodies?

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.

How can cross-reactivity issues be addressed when using FITC-conjugated anti-rat antibodies in multi-species studies?

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 .

What considerations are important when designing flow cytometry experiments using FITC-conjugated anti-rat antibodies?

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.

How does the performance of F(ab')2 fragments compare to whole IgG in FITC-conjugated anti-rat antibody applications?

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.

What are the optimal protocols for using FITC-conjugated anti-rat antibodies in immunohistochemistry?

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 .

What are the best practices for using FITC-conjugated anti-rat antibodies in western blot analysis?

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:

    • Direct fluorescence imaging using systems equipped with appropriate excitation (488 nm) and emission (520 nm) filters

    • Avoid exposure to light during all post-incubation steps to prevent photobleaching

    • Store membranes in the dark if repeated imaging is needed .

  • 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 .

How should researchers optimize FITC-conjugated anti-rat antibodies for flow cytometric analysis of intracellular antigens?

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:

    • Include isotype-matched FITC-conjugated antibody controls with irrelevant specificity

    • For intracellular staining, isotype controls must undergo identical fixation/permeabilization procedures

    • Include single-stained controls for each fluorochrome when performing multicolor analysis

  • 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

    • Apply FITC-conjugated antibody at optimized concentration

  • Signal amplification strategies: For low-abundance intracellular targets, consider:

    • Using polyclonal rather than monoclonal antibodies for broader epitope recognition

    • Implementing indirect detection with unconjugated primary antibodies followed by FITC-conjugated secondary antibodies at higher concentrations

    • Extending incubation times to enhance signal development

  • Data analysis considerations: When analyzing intracellular staining:

    • Use appropriate gating strategies that account for altered light scatter properties after fixation/permeabilization

    • Include unstained, permeabilized controls to establish autofluorescence baselines

    • Consider fluorescence-minus-one (FMO) controls for accurate gate placement in multicolor experiments

How can researchers resolve high background issues when using FITC-conjugated anti-rat antibodies?

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:

    • Use 0.2 μg of purified 2.4G2 antibody (Mouse BD Fc Block™) per sample

    • Incubate for 5 minutes on ice before adding the FITC-conjugated antibody

    • Consider using F(ab')2 fragments instead of whole IgG to eliminate Fc-mediated binding

  • Optimization of washing procedures:

    • Increase wash volume and number of washes (minimum 3 washes)

    • Ensure thorough suspension of cells/tissue during washing

    • Include 0.1% Tween-20 or 0.1% Triton X-100 in wash buffers to reduce non-specific binding

  • Reduce autofluorescence:

    • For formalin-fixed tissues, treat with 0.1-1% sodium borohydride

    • For cells with high intrinsic autofluorescence, consider Sudan Black B treatment

    • Adjust instrument settings to discriminate specific signal from autofluorescence using appropriate controls

  • Buffer composition adjustments:

    • Use buffers containing 1-2% protein (BSA or serum)

    • Add 0.1% sodium azide to prevent bacterial growth (with appropriate safety precautions)

    • Ensure pH is optimized (typically 7.2-7.4) for antibody binding

  • Sample-specific considerations:

    • For fixed samples, ensure complete blocking of aldehyde groups

    • For tissue sections, extend blocking times and increase blocking protein concentration

    • For cells with high endogenous biotin, use avidin/biotin blocking kits when relevant

What criteria should be used to validate FITC-conjugated anti-rat antibodies before incorporating them into critical experiments?

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:

    • Signal-to-noise ratio determination across a concentration gradient

    • Staining index calculation (mean positive signal minus mean negative signal, divided by 2× standard deviation of negative population)

    • Batch-to-batch consistency verification

    • Stability testing under experimental conditions

  • Cross-reactivity evaluation:

    • Testing against related species immunoglobulins

    • Analysis of potential cross-reactivity with other immunoglobulin classes

    • For example, the Rabbit anti-Rat IgM antibody showed a single precipitin arc against specific targets with no reaction against other rat heavy or light chain proteins

  • Application-specific validation:

    • Flow cytometry: Compare results with alternative clones or detection methods

    • Immunofluorescence: Confirm localization patterns match known biology

    • Western blot: Verify molecular weight and banding pattern accuracy

    • Include positive and negative control samples in each validation experiment

  • Documentation requirements:

    • Record lot number, concentration, validation date, and results

    • Maintain images or data files from validation experiments

    • Document optimal working conditions (concentration, incubation time, temperature)

    • Include certificate of analysis information in experimental records

How do fluorescence intensity measurements with FITC-conjugated antibodies correlate with actual target abundance?

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:

    • Epitope accessibility varies between different sample preparations

    • Post-translational modifications may affect antibody binding efficiency

    • Protein complexes may mask certain epitopes, reducing apparent signal

    • Cellular autofluorescence can differentially impact measurements in different cell types

  • 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.

How can FITC-conjugated anti-rat antibodies be effectively used in multiplex immunofluorescence protocols?

Multiplex immunofluorescence using FITC-conjugated anti-rat antibodies requires strategic planning and optimization:

  • Spectral compatibility planning:

    • FITC (excitation ~495 nm, emission ~520 nm) pairs well with fluorophores having minimal spectral overlap

    • Compatible combinations include FITC with Texas Red, APC, and far-red dyes

    • When using FITC with PE, ensure proper compensation settings to address the significant spectral overlap

  • Sequential staining strategies:

    • When using multiple antibodies from the same host species (e.g., multiple rat primary antibodies):

      • Apply the first primary antibody, followed by FITC-conjugated anti-rat antibody

      • Block remaining anti-rat binding sites with excess rat serum

      • Apply the second rat antibody directly conjugated to a spectrally distinct fluorophore

  • 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:

    • Implement controls for each individual antibody channel

    • Include fluorescence-minus-one (FMO) controls for accurate gating

    • Use linear unmixing algorithms when available to separate overlapping fluorescence signals

    • Consider photobleaching characteristics when designing acquisition sequences

  • 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 .

What considerations are important when using FITC-conjugated anti-rat antibodies in in vivo imaging applications?

In vivo imaging with FITC-conjugated antibodies presents distinct challenges not encountered in fixed or ex vivo applications:

  • Biodistribution and pharmacokinetic considerations:

    • FITC-conjugated full IgG antibodies have relatively long circulatory half-lives (days)

    • Consider using F(ab')2 or Fab fragments for faster clearance and improved tissue penetration

    • Account for potential immunogenicity of the carrier protein in repeat-dosing experiments

  • 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:

    • Include appropriate controls with isotype-matched FITC-conjugated antibodies of irrelevant specificity

    • For quantitative applications, co-inject reference standards

    • When possible, validate in vivo findings with ex vivo tissue analysis

    • Consider photobleaching effects in longitudinal imaging protocols

  • Ethical and regulatory considerations:

    • Ensure proper approval for in vivo antibody administration

    • Carefully monitor for potential adverse reactions to the antibody or conjugate

    • Minimize animal numbers through optimal experimental design

    • Consider alternative in vitro or ex vivo approaches when feasible

While challenging, in vivo applications of FITC-conjugated antibodies can provide valuable insights into dynamic biological processes when properly optimized.

How do researchers determine the optimal F/P (fluorescein/protein) ratio for FITC-conjugated antibodies in different applications?

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:

    • Batch-to-batch consistency in F/P ratio is essential for reproducible results

    • Single-molecule characterization methods can reveal conjugation heterogeneity

    • Regular validation of conjugate performance in relevant applications

    • Correlation of F/P ratio with functional performance metrics

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.

How do different host species affect the performance of FITC-conjugated anti-rat antibodies?

The host species used to generate anti-rat antibodies significantly impacts their performance characteristics:

Host SpeciesAdvantagesLimitationsOptimal Applications
Rabbit- High affinity and avidity
- Excellent for polyclonal production
- Good yield of antibody per animal
- Compatible with mouse tissue studies
- Potential cross-reactivity with mouse Ig
- More expensive than goat or chicken antibodies
- Western blotting
- Immunohistochemistry
- Applications requiring high sensitivity
Goat- Large serum volume per animal
- Low background on mouse tissues
- Good for multiple immunoglobulin formats
- Often less expensive
- Lower titer than rabbit antibodies
- May have broader specificity
- Flow cytometry
- Multiplex applications
- Large-scale studies
Mouse- Ideal for detecting rat IgG subtypes
- Excellent specificity through monoclonal production
- Good for discriminating rat Ig isotypes
- Limited to monoclonal production
- May require anti-mouse Ig blocking in mouse tissues
- Isotype-specific detection
- Rat B-cell studies
- Sequential staining protocols

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 .

What are the key differences between various commercially available FITC-conjugated anti-rat antibodies?

A comparative analysis of commercially available FITC-conjugated anti-rat antibodies reveals important differences that inform selection for specific applications:

ProductHost/TypeSpecificityKey FeaturesOptimal Applications
BD PharmingenTM FITC Goat Anti-Rat IgGoat PolyclonalRat IgG and IgM- Adsorbed against mouse Ig
- Little reactivity with rat non-B splenocytes
- Weak cross-reactivity with some hamster Ig
- Primary detection of rat antibody-producing cells
- Secondary detection after rat primary antibodies
Rabbit anti-Rat IgM Antibody (FITC)Rabbit PolyclonalRat IgM (μ chain)- Prepared by immunoaffinity chromatography
- Single precipitin arc in immunoelectrophoresis
- No reaction with other rat heavy/light chain proteins
- Flow cytometry
- Western blotting
- FLISA
- Fluorescence microscopy
BD PharmingenTM FITC Mouse Anti-Rat IgG1Mouse Monoclonal (RG11/39.4)Rat IgG1- Isotype-specific detection
- Effective for both surface and intracellular staining
- Validated for flow cytometric analysis
- Isotype-specific detection
- Intracellular immunoglobulin staining
- Primary or secondary detection in immunofluorescent staining
Rabbit Anti-Rat IgG(H+L)-FITCRabbit PolyclonalHeavy and light chains of rat IgG; light chains of rat IgM- Affinity purified
- No cross-adsorption
- May react with immunoglobulins from other species
- ELISA
- Immunohistochemistry (frozen/paraffin)
- Western blot

This comparison highlights the importance of selecting antibodies based on specific experimental requirements, particularly regarding isotype specificity, cross-reactivity profiles, and validated applications .

How are FITC-conjugated anti-rat antibodies being utilized in cutting-edge research methodologies?

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:

    • Adaptation of F(ab) fragments for reduced interference with cellular functions

    • Combination with genetically encoded reporters for multiplexed functional studies

    • Implementation in microfluidic organ-on-chip platforms for dynamic immunological studies

  • Tissue clearing technologies:

    • Validation of FITC stability in various clearing protocols (CLARITY, CUBIC, iDISCO)

    • Optimization of antibody penetration in whole-organ immunolabeling

    • Development of signal amplification strategies for deep-tissue imaging

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.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.