Growth Hormone Releasing Hormone (GHRH) antibodies conjugated with Fluorescein Isothiocyanate (FITC) are specialized immunological reagents designed for the detection and visualization of GHRH in various research applications. GHRH, also known as Somatoliberin, Growth hormone-releasing factor (GRF), or Somatocrinin, functions as a critical hypothalamic peptide that stimulates growth hormone secretion from the anterior pituitary gland . The conjugation of these antibodies with FITC, a bright fluorescent compound, enables researchers to visualize GHRH expression and localization using fluorescence-based detection methods.
GHRH antibodies are primarily available as polyclonal preparations, derived from host animals immunized with recombinant GHRH proteins or synthetic peptides corresponding to specific regions of the GHRH molecule. The resulting antibodies recognize and bind to GHRH with high specificity, while the FITC conjugation provides a fluorescent signal detectable by various imaging and analytical techniques .
Fluorescein Isothiocyanate (FITC) represents one of the most widely utilized fluorescent probes for antibody conjugation in biological research. The process of conjugating GHRH antibodies with FITC involves chemical linking of the fluorophore to primary amino groups on the antibody molecule, primarily on lysine residues and the N-terminal amino group .
FITC offers several advantages that make it particularly suitable for antibody conjugation and fluorescence-based applications:
High water solubility, facilitating conjugation reactions in aqueous environments
Bright fluorescence with large extinction coefficients and high quantum yields
Minimal non-specific binding to most biological tissues
Optimal excitation at 492 nm and emission at approximately 520 nm (green fluorescence)
Typically, three to five fluorescein molecules can be conjugated to each IgG antibody before self-quenching effects and alterations in binding affinity become problematic. The fluorophore-to-protein ratio in commercial FITC-GHRH antibodies is carefully optimized to maintain antibody functionality while providing sufficient fluorescence intensity for detection .
GHRH plays a pivotal role in the regulation of growth hormone (GH) secretion and subsequent physiological processes. Released by the hypothalamus, GHRH acts on the adenohypophyse (anterior pituitary) to stimulate the secretion of growth hormone, which subsequently mediates various growth and metabolic processes throughout the body .
Recent research utilizing FITC-conjugated GHRH antibodies has revealed previously unknown functions of GHRH signaling beyond its classical role in growth regulation. Studies have demonstrated that GHRH receptor (GHRH-R) signaling significantly impacts immune cell function, particularly in T helper 17 (Th17) cell differentiation and related inflammatory processes .
Investigation of GHRH-R signaling in Th17 cells has shown that:
GHRH-R expression is induced throughout Th17 cell differentiation in vitro
GHRH-R activates the JAK-STAT3 pathway, increasing STAT3 phosphorylation
This signaling enhances both non-pathogenic and pathogenic Th17 cell differentiation
GHRH-R signaling promotes gene expression signatures associated with pathogenic Th17 cells
These findings suggest that GHRH signaling extends beyond growth regulation to influence immune function and inflammatory processes. FITC-conjugated GHRH antibodies have been instrumental in visualizing GHRH expression patterns and tracking its interactions with cellular components in these research contexts .
FITC-conjugated GHRH antibodies find utility across a spectrum of laboratory techniques and research applications. Their fluorescent properties enable sensitive detection methods for investigating GHRH expression, localization, and interaction with other cellular components.
The following table summarizes common laboratory applications of FITC-conjugated GHRH antibodies:
| Technique | Application | Working Dilution | Notes |
|---|---|---|---|
| Immunofluorescence (IF) | Visualization of GHRH expression in cells/tissues | 1:25-100 | Enables subcellular localization studies |
| Immunohistochemistry (IHC) | Detection of GHRH in tissue sections | 1:25-100 | Used for both frozen and paraffin sections |
| Immunocytochemistry (ICC) | Examination of GHRH in cultured cells | 1:25-100 | Allows for co-localization studies |
| Western Blotting (WB) | Analysis of GHRH protein expression | 1:250-2500 | Quantitative analysis of protein levels |
| Flow Cytometry | Quantification of GHRH-expressing cells | 1:20-100 | Enables cell sorting based on GHRH expression |
| ELISA | Quantitative detection of GHRH | Varies by kit | High-throughput analysis |
Optimal working dilutions must be determined experimentally for each specific application and sample type .
FITC-conjugated GHRH antibodies have contributed to several significant research findings:
Development of novel ssDNA X-aptamers targeting GHRH peptides, with binding affinities (Kd) in the nanomolar range (1.21-4.75 nM)
Visualization of GHRH-R expression during Th17 cell differentiation, revealing its role in autoimmune inflammatory processes
Investigation of GHRH signaling in neurocognitive functions and potential neuroprotective effects
Co-immunofluorescence staining demonstrating dose-dependent binding of X-aptamers to GHRHR in pancreatic cancer cells (MIA PaCa-2)
These studies highlight the versatility of FITC-GHRH antibodies in diverse research contexts, from basic science investigations to potential therapeutic applications.
Researchers employing FITC-conjugated GHRH antibodies should consider several technical factors to ensure experimental success and reliable results.
Verification of antibody specificity and performance is crucial:
Include appropriate positive and negative controls in experimental design
Validate specificity using samples with known GHRH expression patterns
Consider the species reactivity limitations when selecting experimental models
Be aware that antibody performance may vary across applications and sample types
Ongoing research utilizing FITC-conjugated GHRH antibodies continues to expand our understanding of GHRH biology and potential therapeutic applications.
Recent studies have revealed an unexpected role for GHRH in modulating immune responses, particularly in the context of autoimmune diseases. GHRH-R signaling has been shown to function as a critical factor in regulating Th17 cell differentiation and Th17 cell-mediated autoimmune ocular and neural inflammation .
Experimental evidence demonstrates that:
GHRH-R deficient mice show significantly lower frequency of IL-17A+CD4+ T cells co-expressing pathogenic Th17 cytokines
Inhibiting GHRH-R signaling reduces Th17 cell differentiation
GHRH antagonists can attenuate Th17 cell-mediated inflammation in vivo
These findings suggest potential therapeutic applications for GHRH antagonists in treating autoimmune conditions characterized by dysregulated Th17 responses.
GHRH signaling also appears to influence neurocognitive function. Research has demonstrated that administration of GHRH agonist JI-34 attenuates intermittent hypoxia-induced neurocognitive deficits and anxiety, suggesting neuroprotective effects of GHRH signaling .
UniGene: Oar.14370
GHRH (Growth Hormone Releasing Hormone), also known as Somatoliberin, GRF, or Somatocrinin, is a hypothalamic peptide hormone that acts on the adenohypophyse to stimulate the secretion of growth hormone . It plays a critical role in the growth hormone axis and has significant implications in developmental biology, endocrinology, and metabolic research. Understanding GHRH function is essential for studies on growth disorders, aging, and certain metabolic conditions where the growth hormone pathway is implicated.
FITC (Fluorescein Isothiocyanate) conjugation refers to the chemical attachment of the fluorescent FITC molecule to the antibody structure. This conjugation enables direct visualization of the antibody-antigen binding through fluorescence microscopy or flow cytometry without requiring secondary antibody detection steps . The FITC molecule absorbs blue light (maximum at approximately 490 nm) and emits green light (maximum at approximately 520 nm), providing a specific signal when bound to the target GHRH protein in experimental samples.
The GHRH antibodies described in the search results are primarily polyclonal antibodies , which recognize multiple epitopes on the GHRH protein. This characteristic provides higher sensitivity for detecting low-abundance targets and greater tolerance for minor protein modifications compared to monoclonal antibodies. Polyclonal antibodies are generated by immunizing host animals (typically rabbits) with recombinant GHRH protein or peptide immunogens . For example, one product uses "Recombinant Sheep Somatoliberin protein (1-44AA)" as the immunogen to generate rabbit polyclonal antibodies . When selecting between polyclonal and monoclonal GHRH antibodies, researchers should consider the balance between sensitivity (favoring polyclonals) and specificity (potentially better with monoclonals).
FITC-conjugated GHRH antibodies have been validated for multiple experimental applications. Primary applications include:
Immunocytochemistry (ICC)/Immunofluorescence (IF): For cellular localization of GHRH with recommended dilutions of 5-20μg/mL (1:25-100)
Immunohistochemistry (IHC): For tissue localization at dilutions of 5-20μg/mL (1:25-100)
Western Blotting (WB): For protein detection at dilutions of 0.2-2μg/mL (1:250-2500)
These applications allow researchers to investigate GHRH expression, localization, and quantification across different experimental systems.
When using GHRH-FITC antibodies in a new experimental system, a systematic dilution optimization approach is recommended. Begin with the manufacturer's suggested range (e.g., 5-20μg/mL for ICC/IHC or 0.2-2μg/mL for WB) , and perform a dilution series experiment. For immunofluorescence applications, start with three dilutions spanning the recommended range (e.g., 5μg/mL, 10μg/mL, and 20μg/mL), evaluating signal strength versus background fluorescence. For Western blotting, test dilutions at 0.2μg/mL, 1μg/mL, and 2μg/mL to identify optimal conditions . Include appropriate positive and negative controls to distinguish specific from non-specific binding. The optimal working dilution may need to be determined empirically by each end user based on their specific experimental conditions .
Autofluorescence can significantly impact the signal-to-noise ratio when using FITC-conjugated antibodies. To minimize this issue:
Implement an autofluorescence quenching step using:
0.1% Sudan Black B in 70% ethanol (10-20 minutes) followed by thorough washing
Commercial autofluorescence quenching reagents specific for FITC wavelengths
Photobleaching by pre-exposure to the excitation wavelength
Optimize fixation protocols:
Minimize fixation time with paraformaldehyde
Consider alternative fixatives that produce less autofluorescence
Include appropriate controls:
Non-immune IgG-FITC controls at matching concentrations
Tissue treated with all reagents except the primary antibody
Use spectral imaging or confocal microscopy with narrow bandpass filters to distinguish FITC signal from autofluorescence
These approaches can significantly improve the signal-to-noise ratio when visualizing GHRH in tissues with intrinsic autofluorescence.
Species cross-reactivity is a critical consideration when selecting GHRH antibodies. The products in the search results show varying species reactivity profiles. For example, some antibodies react specifically with sheep GHRH , while others react with mouse GHRH or have broader reactivity including human, mouse, and rat samples .
To determine cross-reactivity:
Examine sequence homology between your species of interest and the immunogen species
Review validation data from manufacturers for your specific species
Perform preliminary validation experiments:
Western blot against recombinant GHRH from different species
Peptide competition assays to confirm specificity
Side-by-side comparisons using tissues from different species with known GHRH expression patterns
For novel species applications, perform validation controls by comparing with tissues/cells known to express or not express GHRH, and consider peptide blocking experiments to confirm specificity.
GHRH antibodies are typically raised against either full-length recombinant proteins or specific peptide sequences. Some products specifically use recombinant GHRH proteins as immunogens , while others may target specific epitope regions. For example, one product uses "Recombinant Sheep Somatoliberin protein (1-44AA)" as the immunogen , suggesting that antibodies were raised against the full bioactive GHRH peptide.
The epitope recognition affects experimental design in several ways:
Protein conformation requirements - antibodies against internal epitopes may not work in applications where the protein maintains native conformation
Accessibility considerations - Some epitopes may be masked in certain applications or tissue preparations
Potential for cross-reactivity with related peptides - particularly important for GHRH which shares structural features with other hypothalamic peptides
When designing experiments, researchers should consider whether denaturation (as in Western blotting) or fixation (as in IHC) might affect epitope accessibility based on the antibody's specific recognition region.
GHRH belongs to a family of structurally related hypothalamic neuropeptides. To assess and minimize potential cross-reactivity:
Perform comprehensive validation experiments:
Western blot analysis comparing GHRH with related peptides (e.g., VIP, PACAP, GLP)
Peptide competition assays using both GHRH and related peptides
Immunostaining in tissues known to express related peptides but not GHRH
Use bioinformatics approaches:
Analyze sequence similarity between GHRH and related peptides
Identify unique epitope regions that could minimize cross-reactivity
Experimental controls:
Include samples from GHRH knockout models if available
Use corroborating methods (e.g., in situ hybridization) to confirm antibody specificity
FITC-conjugated GHRH antibodies can be valuable tools for studying GHRH receptor-ligand dynamics through advanced live-cell imaging approaches:
Pulse-chase experiments:
Label extracellular GHRH with FITC-conjugated antibodies
Track internalization and trafficking through confocal time-lapse imaging
Quantify endosomal trafficking and receptor recycling dynamics
Single-molecule tracking:
Use high-sensitivity cameras and appropriate optical setups to track individual FITC-labeled GHRH molecules
Analyze binding kinetics, diffusion rates, and interaction times with receptors
FRET (Förster Resonance Energy Transfer) experiments:
Combine FITC-conjugated GHRH antibodies with appropriately labeled GHRH receptors
Monitor interaction dynamics through changes in FRET efficiency
Quantify binding events in real-time
These approaches require careful optimization of antibody concentrations to avoid interfering with natural binding dynamics, and may require sophisticated image analysis tools to extract quantitative data.
A comprehensive experimental design to study GHRH-receptor interactions would integrate multiple methodologies:
Receptor expression and localization studies:
Functional signaling analysis:
Receptor binding kinetics:
Species specificity comparisons:
This multi-faceted approach provides comprehensive understanding of GHRH-receptor biology across structural, functional, and species-specific dimensions.
When faced with contradictory results between FITC-antibody detection and other methods, systematic troubleshooting approaches include:
Technical validation:
Evaluate potential photobleaching effects specific to FITC
Assess pH sensitivity of FITC fluorescence (optimal at pH 8.0-9.0)
Compare results with alternative detection methods (e.g., non-conjugated primary + secondary antibody)
Biological considerations:
Examine epitope accessibility differences between methods
Consider post-translational modifications that might affect antibody binding
Evaluate potential degradation of GHRH in different sample preparations
Experimental controls:
Use recombinant GHRH protein as positive control
Implement competitive binding with unlabeled GHRH
Consider tissue-specific matrix effects that might interfere with binding
Methodological analysis:
Design side-by-side comparisons with standardized samples
Implement peptide blocking controls with the immunizing peptide
Consult reference standards or established model systems
Orthogonal approaches:
Corroborate findings with mRNA expression data
Use multiple antibodies targeting different epitopes
Implement genetic models (knockdown/knockout) to validate specificity
Scientific rigor requires addressing these contradictions through systematic investigation rather than discarding conflicting results.
To maximize the performance and shelf-life of FITC-conjugated GHRH antibodies:
Storage temperature:
Light protection:
FITC is photosensitive - store in amber vials or wrapped in aluminum foil
Minimize exposure to light during experimental procedures
Consider working under reduced ambient lighting when performing experiments
Buffer conditions:
Aliquoting recommendations:
Upon receipt, prepare small single-use aliquots
Use sterile techniques to prevent microbial contamination
Record date of aliquoting and track time out of frozen storage
These handling practices help maintain both antibody binding efficiency and FITC fluorescence intensity over time.
A comprehensive validation approach for GHRH-FITC antibodies should include:
Positive controls:
Tissues or cells known to express GHRH (e.g., hypothalamic tissue)
Recombinant GHRH protein at known concentrations
Previously validated samples from published studies
Negative controls:
Tissues or cells known not to express GHRH
Isotype control: non-specific IgG-FITC from the same species at equivalent concentration
Secondary antibody only controls (for comparison with non-conjugated systems)
Specificity controls:
Peptide competition/blocking with the immunizing peptide
GHRH knockout or knockdown samples if available
Gradient of recombinant GHRH protein to establish detection limits
Technical controls:
Titration series to determine optimal antibody concentration
Multiple fixation/permeabilization methods comparison
Internal standard samples across experiments for consistent quantification
Cross-method validation:
Parallel analysis using alternative detection methods
Correlation with mRNA expression data
Comparison with published findings using different antibodies
Systematic implementation of these controls ensures reliable and reproducible results when introducing these antibodies to new experimental systems.
For quantitative comparison of GHRH expression using FITC-conjugated antibodies:
Flow cytometry approaches:
Measure mean fluorescence intensity (MFI) of cell populations
Use calibration beads with defined FITC molecules for absolute quantification
Implement consistent gating strategies across experiments
Fluorescence microscopy quantification:
Use consistent exposure settings and microscope parameters
Implement automated image analysis with consistent thresholding
Quantify integrated fluorescence intensity normalized to area or cell number
Include reference standards in each imaging session
Western blot quantification (if using FITC for direct detection):
Use recombinant GHRH protein to generate standard curves
Implement consistent scanning parameters
Normalize to loading controls and reference standards
Analyze using appropriate software that accommodates the fluorescence dynamic range
Experimental design considerations:
Process all comparative samples simultaneously
Include internal controls across experimental batches
Randomize sample processing to avoid systematic bias
Blind analysts to experimental conditions during quantification
Statistical approaches:
Implement appropriate statistical tests for the experimental design
Report both normalized values and measures of variance
Consider power analysis to determine appropriate sample sizes
Account for technical variation through appropriate replication
These methodological considerations ensure reliable quantitative comparisons of GHRH expression across experimental conditions.