The ENHO antibody targets the Adropin peptide (amino acids 37–54 in humans) , while the FITC conjugate emits green fluorescence (excitation: 495 nm, emission: 525 nm) upon binding . FITC conjugation typically preserves antibody specificity but requires optimization to avoid reduced binding affinity or nonspecific staining .
FITC-conjugated ENHO antibodies are used in:
Immunofluorescence (IF): Detecting ENHO expression in paraffin-embedded tissues .
Flow Cytometry: Secondary detection in multi-label experiments (e.g., combined with anti-tubulin antibodies) .
Validation: Confirming successful FITC labeling of other proteins .
| Conjugate | Applications | Sensitivity |
|---|---|---|
| FITC | IF, IHC, flow cytometry | High (fluorescence-based) |
| Biotin | ELISA | Moderate (requires streptavidin-HRP) |
| HRP | Immunoblotting | High (chemiluminescence) |
ENHO (Energy Homeostasis Associated) is a protein involved in the regulation of glucose homeostasis and lipid metabolism. As a key regulator in metabolic pathways, ENHO (also known as adropin) has gained significant interest in research related to cardiovascular health, metabolic diseases, and energy balance regulation. The protein is encoded by the ENHO gene located on chromosome 9 in humans, with the UniProt ID Q6UWT2. Research interest in ENHO stems from its potential role in metabolic disorders including obesity, diabetes, and cardiovascular disease, making it an important target for immunological detection and quantification in various experimental systems .
Antibody conjugation is the process of chemically linking an antibody to another molecule, such as a fluorescent dye, enzyme, biotin, or nanoparticle. This process enhances the antibody's detection capabilities by enabling visualization or measurement in various assays. Specifically, FITC conjugation involves the chemical attachment of the fluorescent dye fluorescein isothiocyanate to an antibody molecule, typically through covalent bonding to primary amine groups on the antibody. This conjugation occurs most commonly at lysine residues or the N-terminus of the antibody's heavy and light chains .
The conjugation chemistry typically utilizes the isothiocyanate group of FITC, which reacts with primary amines on the antibody under slightly alkaline conditions (pH 8.0-9.5) to form stable thiourea bonds. The reaction must be carefully controlled to ensure sufficient labeling without compromising the antibody's antigen-binding capacity. Modern conjugation kits like Lightning-Link® systems have simplified this process, allowing researchers to conjugate antibodies in just three simple steps while maintaining antibody functionality .
Commercial ENHO Antibody, FITC conjugated products typically have the following specifications:
| Characteristic | Specification |
|---|---|
| Antibody Type | Polyclonal |
| Host Species | Rabbit |
| Target Species Reactivity | Human |
| Immunogen | Peptide sequence from Human Adropin protein (AA 37-54) |
| Purification Method | Protein G purification (>95% purity) |
| Conjugate | FITC (Fluorescein isothiocyanate) |
| Isotype | IgG |
| Buffer Composition | 50% Glycerol, 0.01M PBS, pH 7.4, 0.03% Proclin 300 |
| Storage Recommendation | -20°C or -80°C |
| Target Aliases | Adropin, C9orf165, Energy homeostasis associated protein |
| Applications | Immunofluorescence, Flow cytometry |
This information helps researchers select the appropriate antibody for their specific experimental needs and understand the product's properties for optimal use in laboratory settings .
ENHO Antibody, FITC conjugated serves multiple research applications, particularly in studies investigating energy metabolism, glucose homeostasis, and lipid regulation. The principal applications include:
Immunofluorescence microscopy: For visualizing the subcellular localization of ENHO protein in tissue sections or cultured cells. The direct FITC conjugation eliminates the need for secondary antibody incubation steps, reducing background and cross-reactivity issues while simplifying the experimental workflow .
Flow cytometry: For quantitative analysis of ENHO expression in cell populations. The FITC fluorophore is excited at 488 nm and emits green fluorescence at approximately 520 nm, making it compatible with standard flow cytometry lasers and filter sets. This application is particularly valuable for studying ENHO expression in different cell types or under various metabolic conditions .
Confocal microscopy: For high-resolution imaging of ENHO protein distribution in cells and tissues, enabling detailed analysis of its co-localization with other cellular structures or proteins when combined with additional fluorophore-labeled antibodies targeting other proteins of interest .
Multiplex immunostaining: FITC's spectral characteristics allow it to be combined with other fluorophores with minimal spectral overlap, enabling simultaneous detection of multiple targets in the same sample .
These applications make FITC-conjugated ENHO antibodies valuable tools in metabolism research, cardiovascular studies, and investigations of metabolic disorders .
A methodologically sound protocol for immunofluorescence using ENHO Antibody, FITC conjugated includes the following steps:
Sample preparation:
Fix cells or tissue sections with 4% paraformaldehyde for 15-20 minutes at room temperature
Permeabilize with 0.1-0.5% Triton X-100 in PBS for 5-10 minutes
Block with 5% normal serum (from the same species as the secondary antibody would be if using an indirect method) in PBS with 0.1% Tween-20 for 1 hour at room temperature
Antibody incubation:
Dilute the ENHO Antibody, FITC conjugated to the optimal working concentration (typically 1-10 μg/mL, but this should be empirically determined)
Apply to the sample and incubate in a humidified chamber for 1-2 hours at room temperature or overnight at 4°C
Protect from light throughout the incubation to prevent photobleaching of the FITC fluorophore
Washing steps:
Wash 3-5 times with PBS containing 0.1% Tween-20, 5 minutes each
Perform all washes in the dark or under reduced lighting
Counterstaining and mounting:
Counterstain nuclei with DAPI (4′,6-diamidino-2-phenylindole) at 0.1-1 μg/mL for 5-10 minutes
Mount using an anti-fade mounting medium to minimize photobleaching
Seal the edges of the coverslip with nail polish or commercial sealant
Imaging considerations:
This protocol should be optimized for each specific experimental system and research question.
When encountering weak signal intensity with ENHO Antibody, FITC conjugated, researchers should systematically troubleshoot the following potential causes:
Antibody concentration: Insufficient antibody concentration is a common cause of weak signals. Perform a titration experiment using different antibody concentrations (0.5-20 μg/mL) to determine the optimal working concentration for your specific sample type. Unlike enzyme-conjugated antibodies which offer signal amplification, fluorophore-conjugated antibodies provide direct visualization, requiring careful optimization of concentration .
Target protein expression levels: ENHO may be expressed at low levels in certain tissues or under specific conditions. Consider using positive control samples known to express ENHO abundantly (such as liver tissue) to validate antibody performance. Alternative detection methods with higher sensitivity may be necessary for low-abundance targets .
Sample preparation issues:
Inadequate fixation or over-fixation can mask epitopes
Insufficient permeabilization can prevent antibody access to intracellular targets
Suboptimal antigen retrieval (for FFPE tissues) can prevent antibody binding
Test different fixation methods and permeabilization conditions to optimize epitope accessibility
Fluorophore deterioration:
FITC is relatively susceptible to photobleaching compared to more modern fluorophores
Check the antibody's manufacturing date and storage conditions
Consider using an alternative fluorophore with greater photostability (such as Alexa Fluor 488) if photobleaching is a significant issue
Always store the conjugate at recommended temperatures (-20°C to -80°C) and protect from light exposure during all steps
Signal enhancement strategies:
Increase the exposure time during image acquisition, balancing against background increase
Use a more sensitive detection system (e.g., confocal microscopy, PMT-based detectors)
Consider signal amplification using anti-FITC antibodies or tyramide signal amplification if compatible with your experimental design
By systematically addressing these factors, researchers can optimize signal detection for ENHO using FITC-conjugated antibodies.
High background fluorescence is a common challenge when working with FITC-conjugated antibodies. Researchers can implement the following methodological approaches to reduce background and improve signal-to-noise ratio:
Optimize blocking conditions:
Use 5-10% normal serum from the same species as the host of the primary antibody
Add 0.1-0.3% Triton X-100 or 0.05% Tween-20 to blocking buffers to reduce non-specific binding
Consider adding 1-5% BSA to further reduce non-specific interactions
Extend blocking time to 1-2 hours at room temperature or overnight at 4°C for challenging samples
Reduce autofluorescence:
For tissues with high autofluorescence (particularly formalin-fixed tissues), treat with 0.1-1% sodium borohydride in PBS for 10 minutes before blocking
For samples with lipofuscin (common in aged tissues), incubate with 0.1-0.3% Sudan Black B in 70% ethanol for 20 minutes
Include unstained controls to assess the level of tissue autofluorescence
Improve washing steps:
Increase the number of washes (5-6 washes instead of 3)
Extend washing times to 10-15 minutes per wash
Include 0.05-0.1% Tween-20 in wash buffers
Use gentle agitation during washing to remove unbound antibody effectively
Address antibody quality issues:
Optimize imaging parameters:
Adjust the dynamic range during image acquisition to maximize signal while minimizing background
Use narrower bandpass filters to reduce spectral bleed-through
Employ computational background subtraction during image processing when appropriate
By implementing these methods systematically, researchers can significantly improve the signal-to-noise ratio when using ENHO Antibody, FITC conjugated in their experiments.
Multiplex immunofluorescence allows for simultaneous detection of multiple targets in a single sample, providing valuable co-expression and co-localization data. To effectively incorporate ENHO Antibody, FITC conjugated into multiplex panels, researchers should follow these methodological guidelines:
Spectral compatibility planning:
FITC has excitation/emission maxima at approximately 490/525 nm (green fluorescence)
Select additional fluorophores with minimal spectral overlap, such as:
Panel design considerations:
Reserve FITC for medium-to-high abundance targets like ENHO when expression levels are strong
For lower abundance targets, consider brighter fluorophores like PE or APC
Account for potential cross-reactivity between antibodies by selecting primaries raised in different host species when using indirect detection methods alongside your direct FITC conjugate
Sequential staining approach:
When combining directly conjugated antibodies with indirect detection (primary + secondary):
First apply all unconjugated primary antibodies
Then apply fluorophore-conjugated secondary antibodies
Block any remaining active sites on secondary antibodies
Finally apply directly conjugated antibodies like ENHO-FITC
This prevents secondary antibodies from recognizing the FITC-conjugated primary
Controls for multiplex panels:
Single-color controls: Stain separate samples with each individual antibody to confirm proper localization and assess bleed-through
Fluorescence-minus-one (FMO) controls: Include panels with each fluorophore omitted to identify spectral overlap issues
Isotype controls: Include appropriate isotype controls for each antibody to assess non-specific binding
Image acquisition and analysis considerations:
Use sequential scanning (for confocal microscopy) to minimize spectral crosstalk
Employ spectral unmixing algorithms for fluorophores with partial overlap
Consider using ENHO Antibody conjugated to alternative fluorophores (e.g., Cy3 or Alexa Fluor 647) if spectral conflicts emerge with other targets in your panel
By carefully designing multiplex panels and following these methodological guidelines, researchers can effectively incorporate ENHO Antibody, FITC conjugated into complex immunofluorescence studies examining multiple markers simultaneously.
Quantifying ENHO expression using FITC-conjugated antibodies requires systematic approaches to ensure accuracy and reproducibility. Researchers can employ the following quantitative methods:
Flow cytometry-based quantification:
Prepare single-cell suspensions from tissues or cultured cells
Stain with ENHO Antibody, FITC conjugated using optimized concentrations
Include calibration beads with known fluorescence intensities to convert arbitrary fluorescence units to molecules of equivalent soluble fluorochrome (MESF)
Analyze mean fluorescence intensity (MFI) or median fluorescence intensity of positive populations
Present data as fold-change relative to controls or as absolute MESF values
Incorporate appropriate controls including isotype-FITC control, unstained cells, and positive/negative control samples
Quantitative immunofluorescence microscopy:
Capture images under identical acquisition settings for all experimental conditions
Include internal reference standards in each experiment (such as fluorescent microspheres)
Apply thresholding to segment positive areas from background
Quantify parameters such as:
Mean fluorescence intensity per cell
Integrated density (the product of area and mean gray value)
Percent positive area in tissue sections
Normalize to cell number using nuclear counterstains or to tissue area for comparative analyses
Digital image analysis workflows:
Use software platforms like ImageJ/FIJI, CellProfiler, or QuPath for automated quantification
Develop analysis pipelines that include:
Nuclear segmentation (using DAPI)
Cell boundary identification
Background subtraction
Intensity measurement within defined cellular compartments
Apply batch processing for consistent analysis across multiple images
Export quantitative data for statistical analysis in programs like R or GraphPad Prism
Statistical considerations for quantitative analyses:
Account for non-normal distributions in fluorescence intensity data
Apply appropriate transformations (log, square root) before parametric statistical tests
Consider cell-by-cell analysis rather than population averages when heterogeneity is expected
Report both technical and biological replicates with appropriate measures of central tendency and dispersion
By implementing these quantitative approaches, researchers can obtain reliable measurements of ENHO expression levels using FITC-conjugated antibodies across various experimental systems and conditions.
When selecting fluorophore conjugates for ENHO detection, researchers should consider the relative advantages and limitations of FITC compared to alternative fluorophores. The following comparative analysis provides guidance for informed decision-making:
| Fluorophore | Excitation/Emission (nm) | Relative Brightness | Photostability | pH Sensitivity | Key Advantages | Limitations |
|---|---|---|---|---|---|---|
| FITC | 490/525 | Moderate | Low-Moderate | High (quenches below pH 7) | - Widely compatible with standard filter sets - Well-established protocols - Relatively inexpensive | - Susceptible to photobleaching - pH-sensitive - Moderate brightness |
| Alexa Fluor 488 | 495/519 | High | High | Low | - Superior photostability - pH-insensitive (pH 4-10) - Brighter than FITC | - Higher cost - Requires specific filter sets for optimal performance |
| R-PE | 488, 546, 565/575-585 | Very High (1,960,000 M⁻¹cm⁻¹) | Moderate | Moderate | - Extremely bright - Good for low abundance targets - Multiple excitation peaks | - Larger size may affect penetration - More sensitive to storage conditions - More expensive |
| iFluor® 488 | 491/516 | High | High | Low | - Superior brightness - High photostability - Good for advanced imaging | - Less commonly available - Higher cost - Less literature precedent |
| Cy3 | 550/570 | High | Moderate-High | Low | - Red-shifted (less autofluorescence) - Higher signal-to-noise in tissues - Good photostability | - Different filter set than FITC - Cannot be used with FITC in same channel |
| Quantum Dots | Variable | Very High | Very High | Low | - Exceptional photostability - Very bright - Narrow emission peaks | - Larger size - Potential for aggregation - Higher cost - Specialized equipment needed |
For ENHO detection specifically:
For routine applications (basic immunofluorescence, flow cytometry):
FITC conjugates provide adequate performance with standard equipment
Consider when budget constraints exist or when comparing to historical FITC-based data
For challenging samples (low ENHO expression, high autofluorescence tissues):
Alexa Fluor 488 offers superior performance with similar spectral properties to FITC
R-PE provides significantly higher brightness for detecting low abundance ENHO
For advanced imaging applications (confocal, super-resolution):
For multiplexing considerations:
When designing multiplex panels, the spectral properties of each fluorophore must be considered holistically
FITC occupies the green channel, leaving red and far-red channels available for other targets
For complex multiplexing, spectral unmixing with narrower bandwidth fluorophores may be advantageous
This comparative analysis enables researchers to select the optimal fluorophore conjugate for their specific ENHO detection needs based on experimental requirements, equipment availability, and research goals.
While FITC-conjugated antibodies are valuable tools for ENHO detection, alternative methodologies may be more appropriate in certain research contexts. Researchers should consider the following situations and alternative approaches:
For quantitative protein expression analysis:
Enzyme-linked immunosorbent assay (ELISA): When precise quantification of ENHO levels in solution is required, ELISA using HRP or AP conjugates offers superior quantification compared to fluorescence-based methods
Western blotting with enzyme conjugates: For determining protein size, post-translational modifications, or when sample autofluorescence is problematic
Methodology: Use unconjugated anti-ENHO primary antibody followed by enzyme-conjugated secondary antibody (HRP or AP) for enhanced sensitivity through signal amplification
For challenging tissue samples:
Immunohistochemistry with chromogenic detection: When working with tissues having high autofluorescence (e.g., liver, kidney) or with archived FFPE samples
Metal-conjugated antibodies for mass cytometry (CyTOF): For high-dimensional analysis without fluorescence spectral overlap limitations
Methodology: Use biotinylated anti-ENHO antibody followed by streptavidin-HRP and DAB substrate for chromogenic detection in difficult tissues
For high sensitivity requirements:
Tyramide signal amplification (TSA): When detecting very low levels of ENHO expression
Biotin-streptavidin amplification systems: For enhanced signal through multiple binding sites
Methodology: Use unconjugated primary anti-ENHO antibody, followed by biotinylated secondary antibody, then streptavidin-HRP and tyramide-fluorophore for exponential signal amplification
For live cell applications:
For spatial context in tissue architecture:
Multiplex immunohistochemistry with sequential chromogenic detection: When spatial relationships between ENHO and multiple markers are critical
RNA in situ hybridization: When antibody specificity is a concern or to correlate protein with mRNA expression
Methodology: Use RNAscope or similar technology to detect ENHO mRNA in conjunction with protein detection
The decision to use alternative detection methods should be guided by the specific research question, sample characteristics, required sensitivity, and available instrumentation. Each method offers distinct advantages in particular experimental contexts, and researchers may benefit from employing multiple complementary approaches to validate and extend their findings on ENHO biology.
Validating antibody specificity is crucial for ensuring reliable and reproducible research outcomes. For ENHO Antibody, FITC conjugated, researchers should implement the following methodological validation strategies:
Positive and negative control samples:
Genetic validation approaches:
Compare staining in wild-type versus ENHO knockout models (if available)
Use ENHO-overexpressing systems as positive controls
Employ siRNA or shRNA knockdown of ENHO to demonstrate reduced staining intensity
These genetic approaches represent the gold standard for antibody validation
Peptide competition assays:
Pre-incubate the ENHO Antibody, FITC conjugated with excess immunizing peptide (the peptide sequence from Human Adropin protein AA 37-54 used to generate the antibody)
Compare staining with and without peptide pre-absorption
Specific staining should be significantly reduced or eliminated after peptide competition
Orthogonal method validation:
Confirm ENHO expression using independent methods such as:
Western blotting with different anti-ENHO antibodies
Mass spectrometry-based protein identification
RNA expression analysis (RT-qPCR or RNA-seq)
Correlation between methods strengthens confidence in antibody specificity
Multi-antibody concordance:
Publication and reporting standards:
Document all validation steps performed
Report the specific catalog number, lot number, and dilution used
Include representative images of positive and negative controls
Disclose any limitations in specificity or cross-reactivity observed
Follow reporting guidelines such as those proposed by the International Working Group for Antibody Validation (IWGAV)
By implementing these validation strategies, researchers can establish confidence in the specificity of their ENHO Antibody, FITC conjugated and produce more reliable and reproducible research findings.
Co-localization analysis using ENHO Antibody, FITC conjugated requires careful methodological considerations to ensure accurate data interpretation. Researchers should address the following critical aspects: