The FITC-conjugated NDNF antibody has been validated in multiple experimental contexts:
Western Blotting: Detects NDNF in protein lysates, with a predicted band size of ~55 kDa .
Flow Cytometry: Enables quantification of NDNF-expressing cells in heterogeneous populations .
Immunohistochemistry: Localizes NDNF in formalin-fixed paraffin-embedded (FFPE) tissues .
Comparative studies using similar FITC-conjugated antibodies (e.g., anti-IFN-alpha and anti-NeuN ) demonstrate standardized protocols for fluorescent staining, including counterstaining with DAPI for nuclear visualization .
FITC conjugation typically involves:
Antibody Modification: Incubation with a modifier to expose reactive amine groups .
FITC Binding: Covalent attachment of FITC isothiocyanate to lysine residues .
Quenching: Addition of a quencher to stabilize the conjugate .
The Lightning-Link® FITC Conjugation Kit (ab188285) exemplifies this process, achieving >90% conjugation efficiency in 20 minutes .
Researchers used FITC conjugates in lateral flow assays to quantify folate, achieving a linear response (R² > 0.98) via T/C ratio analysis .
FITC-conjugated anti-NeuN antibodies enabled flow cytometry-based identification of neuronal nuclei in rat and human samples, with minimal non-specific binding (IgG controls <2% background) .
NENF (Neuron-derived neurotrophic factor, also known as Neudesin) is a secreted protein that functions as a neurotrophic factor in postnatal mature neurons, enhancing neuronal survival . Its biological significance extends to several key neural processes:
Promotes cell proliferation and neurogenesis in undifferentiated neural progenitor cells during embryonic development
Inhibits differentiation of astrocytes
Exerts neurotrophic activity via MAPK1/ERK2, MAPK3/ERK1, and AKT1/AKT signaling pathways
Functions as an anorexigenic neurotrophic factor contributing to energy balance
Plays a role in maintaining cellular mechanisms such as synaptic plasticity and neural circuit formation
NENF's neurotrophic activity is notably enhanced by binding to heme, and it interacts with proteins such as the tyrosine receptor kinase (Trk) family, influencing downstream effects on neuron maintenance and plasticity .
FITC (fluorescein isothiocyanate) is a fluorochrome dye widely used for antibody labeling due to its favorable optical properties and relatively simple conjugation chemistry. The key spectral characteristics include:
| Property | Specification | Notes |
|---|---|---|
| Excitation maximum | 495 nm | Efficiently excited by blue light |
| Emission maximum | 525 nm | Visible yellow-green fluorescence |
| Quantum efficiency | High | Results in bright signal |
| Molecular stability | Good | Under proper storage conditions |
| pH sensitivity | Yes | Optimal fluorescence at pH 8-9 |
FITC conjugation to proteins occurs via reaction with free amino groups to form stable conjugates . This chemistry is relatively simple and typically preserves the biological activity of the labeled protein . The molecular structure allows for efficient energy absorption and emission, though attention to pH is important as fluorescence intensity can be reduced at lower pH values.
The NENF Antibody, FITC conjugated has the following technical specifications:
| Characteristic | Description |
|---|---|
| Host species | Rabbit |
| Clonality | Polyclonal |
| Isotype | IgG |
| Target | NENF (Neudesin) |
| Reactivity | Human |
| Form | Liquid |
| Immunogen | Peptide sequence from Human Neudesin protein (149-167AA) |
| Purification method | Antigen Affinity Purified |
| Storage buffer | 0.03% Proclin 300, 50% Glycerol, 0.01M PBS, pH 7.4 |
| Storage conditions | -20°C or -80°C; avoid repeated freeze-thaw cycles |
| Research applications | Neuroscience; cell markers |
This antibody specifically recognizes the NENF protein, which has aliases including Cell growth inhibiting protein 47, Cell immortalization-related protein 2, and Secreted protein of unknown function (SPUF) .
For successful immunofluorescence staining with NENF Antibody, FITC conjugated, follow this methodological approach:
Sample Preparation:
Fix cells or tissue sections using 4% paraformaldehyde
Permeabilize with 0.1-0.5% Triton X-100 in PBS (for intracellular antigens)
Block with 5-10% normal serum (from species unrelated to primary antibody) for 1 hour
Staining Protocol:
Dilute NENF Antibody, FITC conjugated to optimal concentration (typically 1-10 μg/mL, determined by titration)
Apply to samples and incubate at 4°C overnight or at room temperature for 1-2 hours
Wash 3× with PBS for 5 minutes each
Counterstain nuclei with DAPI if desired
Mount with anti-fade mounting medium
Critical Parameters:
Avoid exposure to light during and after staining to prevent photobleaching of FITC
Include appropriate positive and negative controls
Consider using a blocking peptide control to confirm specificity
Optimize antibody concentration by titration to obtain best signal-to-noise ratio
For multi-color experiments, select secondary fluorophores with minimal spectral overlap with FITC (Ex: 495nm, Em: 525nm) .
When using NENF Antibody, FITC conjugated for flow cytometric analysis, implement these methodological considerations:
Sample Preparation:
Prepare single-cell suspensions (1×10^6 cells/100 μL)
Fix cells with 4% paraformaldehyde if required
For intracellular staining, permeabilize with 0.1% saponin in PBS
Staining Protocol:
Block Fc receptors to reduce non-specific binding
Titrate antibody to optimal concentration (typically ≤0.5 μg per million cells)
Incubate cells with antibody for 30-60 minutes at 4°C, protected from light
Wash twice with flow cytometry buffer
Analyze immediately or fix in 1-2% paraformaldehyde for later analysis
Controls and Validation:
Include unstained cells to establish autofluorescence baseline
Use isotype control (rabbit IgG-FITC) at the same concentration
For specificity confirmation, pre-block the antibody with recombinant NENF protein
Instrument Settings:
Use 488 nm laser for FITC excitation
Collect emission through a 525/20 nm bandpass filter
Adjust photomultiplier tube voltage to place negative population in first decade of log scale
Compensate for spectral overlap if using multiple fluorophores
The quality of flow cytometry data can be improved by carefully optimizing cell preparation, antibody concentration, and instrument settings for each specific application .
While the FITC-conjugated version is not typically used for Western blotting, unconjugated NENF antibodies can be employed with the following protocol:
Sample Preparation:
Extract proteins using RIPA buffer with protease inhibitors
Quantify protein concentration (BCA or Bradford assay)
Denature samples in reducing buffer at 95°C for 5 minutes
Gel Electrophoresis and Transfer:
Load 20-50 μg protein per lane on 12-15% SDS-PAGE gel
Transfer to PVDF membrane at 100V for 60-90 minutes
Immunodetection:
Block membrane with 5% non-fat milk in TBST for 1 hour at room temperature
Incubate with NENF primary antibody (1-2 μg/mL) overnight at 4°C
Wash 3× with TBST, 5 minutes each
Incubate with HRP-conjugated anti-rabbit secondary antibody (1:50,000 dilution) for 1 hour
Wash 3× with TBST
Develop using chemiluminescent substrate
Expected Results:
Human NENF protein should be detected at approximately 21 kDa
Validation data shows clear bands when using 1-2 μg/mL concentration
To confirm specificity, consider using a competitive blocking assay with the immunizing peptide or recombinant NENF protein.
NENF's documented neurotrophic properties position it as a significant factor in several neurological contexts:
Neuroprotection Mechanisms:
NENF enhances neuronal survival through activation of MAPK1/ERK2, MAPK3/ERK1, and AKT1/AKT pathways
This activation may offer protection against neurodegenerative processes
Research suggests NENF may influence survival signaling in mature neurons under stress conditions
Neurogenesis and Neural Development:
NENF promotes proliferation of neural progenitor cells during embryonic development
It inhibits astrocyte differentiation, potentially directing cell fate toward neuronal lineages
These functions suggest research applications in developmental neurobiology and regenerative medicine
Methodological Approach for Disease Models:
Establish baseline NENF expression in normal neural tissues using NENF Antibody, FITC conjugated
Compare expression patterns in disease models through quantitative immunofluorescence
Correlate NENF expression levels with markers of neuronal health and function
Assess whether NENF supplementation or knockdown modifies disease progression
Investigate downstream signaling events using phospho-specific antibodies for ERK1/2 and AKT pathways
These characteristics make NENF a compelling target for investigations into neuroprotective strategies and understanding the molecular basis of neurodegenerative disorders .
Designing effective multiplexed immunofluorescence experiments requires careful attention to several technical parameters:
Spectral Considerations:
Select companion fluorophores with minimal spectral overlap (e.g., DAPI, Cy5, Texas Red)
Consider the spectral limitations of your microscope's filter sets
Antibody Compatibility:
Ensure all primary antibodies are from different host species or use directly conjugated antibodies
When using multiple rabbit antibodies, sequential staining with complete elution between rounds may be necessary
NENF Antibody, FITC conjugated (rabbit host) is compatible with mouse, goat, or rat-derived antibodies in single-step protocols
Protocol Optimization:
Perform single-color controls to establish proper exposure settings
Include fluorescence-minus-one (FMO) controls
Validate staining patterns individually before combining antibodies
Consider the order of antibody application (typically start with weakest signal)
Optimize fixation conditions that preserve all target epitopes
Data Analysis:
Use software capable of spectral unmixing if bleed-through occurs
Quantify colocalization using appropriate statistical methods
Consider automated image analysis for objective quantification of staining patterns
For detecting NENF in combination with other neural markers, this approach enables comprehensive phenotypic characterization while minimizing false colocalization due to spectral overlap .
To ensure experimental reproducibility, researchers should validate each batch of NENF Antibody, FITC conjugated using these parameters:
Fluorophore Characterization:
Fluorescein/protein (F/P) ratio: Optimal range is typically 3-6 molecules of FITC per antibody
Higher F/P ratios (>6) can cause self-quenching and increased non-specific binding
Antibody Specificity:
Western blot analysis with recombinant NENF protein and tissue lysates
Competitive binding assays with unlabeled antibody
Testing with NENF-knockout or knockdown samples as negative controls
Cross-reactivity assessment with related proteins
Functional Validation:
Flow cytometry analysis comparing staining index across batches
Immunofluorescence with standardized positive control samples
Quantitative comparison of signal-to-noise ratios between batches
Confirmation of expected subcellular localization pattern
Storage Stability:
Evaluate fluorescence intensity after storage at recommended conditions (-20°C to -80°C)
Assess performance after multiple freeze-thaw cycles (generally limited to 5 cycles)
Thorough validation using these parameters ensures consistent performance across experiments and minimizes variability due to reagent differences.
High background fluorescence can significantly impact data quality when using NENF Antibody, FITC conjugated. Here are common causes and methodological solutions:
| Problem | Possible Causes | Solutions |
|---|---|---|
| Non-specific binding | Insufficient blocking, excessive antibody concentration | Optimize blocking (5-10% serum, 1-3% BSA); Titrate antibody concentration; Add 0.1-0.5% Tween-20 to washing buffer |
| Autofluorescence | Fixation artifacts, endogenous fluorescent molecules | Reduce fixation time; Use Sudan Black B (0.1-0.3%) to quench autofluorescence; Use spectral unmixing during imaging |
| Cross-reactivity | Antibody binding to related epitopes | Perform pre-adsorption with blocking peptide; Use more stringent washing; Increase salt concentration in wash buffer |
| Over-fixation | Excessive aldehyde groups causing high background | Add glycine (100mM) after fixation to quench free aldehydes; Optimize fixation time; Consider alternative fixatives |
| Photobleaching | FITC signal fading during imaging | Use anti-fade mounting media; Minimize exposure time; Capture FITC images first in multicolor experiments |
Additionally, when using NENF Antibody, FITC conjugated, consider:
FITC has pH-dependent fluorescence; maintain buffer pH at 7.4-8.0 for optimal signal
Storage at -20°C or -80°C and protection from light preserves signal integrity
For flow cytometry, optimize compensation settings to account for FITC spillover into other channels
Methodical optimization of these parameters will significantly improve signal-to-noise ratio and data quality.
To ensure reproducibility and valid comparisons across different experimental conditions, implement these methodological approaches:
Standardization Protocols:
Create a standard curve using recombinant NENF protein at known concentrations
Include a common positive control sample in each experiment
Normalize fluorescence intensity to internal reference markers
Maintain consistent imaging or flow cytometry parameters across experiments
Quantitative Assessment:
For imaging: Measure mean fluorescence intensity (MFI) within defined regions of interest
For flow cytometry: Track median fluorescence intensity and staining index
Calculate signal-to-noise ratio by comparing specific signal to background
Use equivalent antibody concentrations (molar basis) when comparing different detection systems
Cross-Platform Validation:
Validate findings using orthogonal detection methods (e.g., if using flow cytometry, confirm with immunofluorescence microscopy)
Employ standardized reference materials and calibration beads
Document all experimental parameters comprehensively
Statistical Analysis:
Apply appropriate statistical tests to determine significance of observed differences
Account for batch effects using statistical modeling
Consider using robust metrics less sensitive to outliers
By implementing these standardization approaches, researchers can meaningfully compare results across different experimental platforms, time points, or laboratory settings .
NENF Antibody, FITC conjugated provides valuable tools for investigating neural circuit formation and plasticity:
Developmental Profiling:
Track spatiotemporal expression of NENF during neural development
Correlate NENF expression with critical periods of circuit formation
Examine co-expression with cell-type specific markers to identify NENF-expressing populations
Methodological Approach:
Prepare brain sections from animals at different developmental stages
Perform immunofluorescence using NENF Antibody, FITC conjugated (1-10 μg/mL)
Co-stain with markers for neuronal subtypes, glial cells, or synaptic components
Analyze patterns of expression using confocal microscopy and image analysis software
Functional Studies:
Combine NENF immunolabeling with functional assays (calcium imaging, electrophysiology)
Correlate NENF expression with markers of synaptic plasticity
Investigate the effects of NENF knockdown or overexpression on circuit development
Translational Applications:
Compare NENF expression patterns between normal and pathological conditions
Assess potential roles in neurodevelopmental disorders
Explore NENF as a biomarker for specific neural populations or developmental stages
Given NENF's known roles in promoting neuronal survival and regulating neurogenesis, these approaches can yield insights into the molecular mechanisms underlying circuit formation and maintenance .
FITC-conjugated antibodies like NENF Antibody, FITC conjugated are finding new applications in advanced imaging methodologies:
Super-Resolution Microscopy:
FITC provides compatible fluorescence properties for techniques such as Structured Illumination Microscopy (SIM) and Stimulated Emission Depletion (STED)
Resolution improvement from ~250nm (confocal) to ~50-100nm enables visualization of subcellular NENF localization
Protocol modifications for super-resolution:
Use higher antibody dilutions (1:500-1:1000) to reduce background
Employ thinner tissue sections (≤10μm) or monolayer cultures
Use specialized mounting media optimized for super-resolution techniques
Expansion Microscopy:
Physical expansion of specimens allows visualization of protein distribution with enhanced resolution
FITC-conjugated antibodies maintain fluorescence after expansion protocols
Implementation guidelines:
Perform standard immunolabeling with NENF Antibody, FITC conjugated
Cross-link antibodies to the hydrogel matrix
Digest cellular components and expand the specimen
Image with standard confocal microscopy to achieve effective super-resolution
Correlative Light and Electron Microscopy (CLEM):
FITC immunolabeling can be performed prior to EM sample processing
Enables correlation between fluorescence patterns and ultrastructural context
Specialized protocols preserve both fluorescence signal and ultrastructural integrity
Multiplexed Imaging with Spectral Unmixing:
Advanced spectral detectors can separate FITC signal from spectrally similar fluorophores
Enables higher-order multiplexing (5+ colors) in single specimens
Requires calibration with single-fluorophore controls for accurate unmixing
These emerging applications expand the research utility of NENF Antibody, FITC conjugated beyond conventional fluorescence microscopy .
NENF's neurotrophic properties make it relevant to various neurological conditions. Research approaches utilizing NENF Antibody, FITC conjugated include:
Expression Analysis in Disease Models:
Compare NENF expression patterns between normal and diseased tissues
Quantify changes in expression levels during disease progression
Correlate NENF expression with clinical parameters or disease severity
Methodological Protocol:
Collect matched specimens (normal vs. pathological)
Process for immunofluorescence using standardized protocols
Label with NENF Antibody, FITC conjugated at optimized concentration
Co-stain with disease-specific markers
Quantify fluorescence intensity and co-localization patterns
Apply statistical analysis to identify significant differences
Therapeutic Target Validation:
Assess whether modulation of NENF levels affects disease progression
Determine if NENF treatment rescues disease phenotypes in cellular or animal models
Visualize NENF localization before and after therapeutic interventions
Biomarker Development:
Evaluate NENF as a potential diagnostic or prognostic biomarker
Develop flow cytometry protocols for NENF detection in patient-derived samples
Establish correlations between NENF expression and disease outcomes
Given NENF's involvement in neuronal survival pathways and its relationship with the MAPK/ERK and AKT signaling pathways, this research direction may yield insights into neuroprotective mechanisms and potential therapeutic targets for conditions including neurodegenerative diseases and neural injury .
While commercial NENF Antibody, FITC conjugated products utilize standard conjugation methods, researchers pursuing specialized applications may benefit from site-specific conjugation approaches:
Enzymatic Conjugation Methods:
Deglycosylate antibody using PNGase F to expose Gln295 in the Fc region
Introduce an azide-functional handle using microbial transglutaminase (MTGase)
Perform copper-free click chemistry with DBCO-modified FITC
This produces a controlled 1:1 or 2:1 FITC:antibody ratio at specific sites
Advantages of Site-Specific Conjugation:
Preserves antigen-binding capacity by keeping Fab regions unmodified
Creates homogeneous conjugate population with consistent F/P ratio
Reduces potential for antibody aggregation
Enables precise control over conjugation stoichiometry
Analytical Validation:
Confirm conjugation success using SDS-PAGE with fluorescence imaging
Verify F/P ratio through UV-visible spectroscopy analysis
Assess binding kinetics compared to unconjugated antibody
Test functional activity in appropriate assay systems
Applications Benefiting from Site-Specific Conjugation:
Super-resolution microscopy requiring precise localization
Quantitative studies where consistent signal-to-antibody ratio is critical
Development of advanced antibody-based nanoparticle conjugates
Studies involving multimodal imaging where multiple labels are attached
This enzymatic approach represents an advanced alternative to traditional chemical conjugation methods that typically result in heterogeneous products with random attachment points .
Leveraging NENF Antibody, FITC conjugated in high-throughput contexts requires optimization for automated systems:
Sample Preparation for High-Throughput Screening:
Standardize cell plating density in multi-well formats (96/384-well plates)
Implement automated fixation and staining protocols using liquid handling systems
Optimize antibody concentration to balance signal intensity and cost efficiency
Develop streamlined washing protocols compatible with automated systems
Automation-Compatible Staining Protocol:
Image Acquisition Parameters:
Establish consistent exposure settings based on control samples
Program focus algorithms optimized for FITC signal detection
Implement tile-based imaging for larger sample coverage
Consider confocal over widefield to improve signal-to-noise in thick specimens
Data Analysis Pipeline:
Develop automated image segmentation algorithms to identify cellular compartments
Extract quantitative features (intensity, texture, morphology) from NENF-positive structures
Implement machine learning approaches for pattern recognition
Create standardized data visualization and statistical analysis workflows
These methodological adaptations enable screening of compound libraries, genetic perturbations, or patient-derived samples for effects on NENF expression or localization, supporting drug discovery and personalized medicine applications .
Three-dimensional visualization of NENF distribution in intact tissues requires compatible tissue clearing and immunolabeling approaches:
Clearing Method Compatibility:
Solvent-based methods (e.g., BABB, 3DISCO): May cause FITC quenching; not recommended
Aqueous methods (e.g., CLARITY, CUBIC): Generally preserve FITC fluorescence; preferred choice
Hyperhydration methods (e.g., Scale): Moderate FITC preservation; may require higher antibody concentration
Optimized Immunolabeling Protocol for Cleared Tissues:
Clear tissue using CLARITY or CUBIC protocol
Wash extensively to remove clearing reagents that may interfere with antibody binding
Incubate with NENF Antibody, FITC conjugated at 5-10× higher concentration than for thin sections
Extend incubation time to 3-7 days at 37°C with gentle agitation
Wash extensively (3-5 days) to remove unbound antibody
Return sample to clearing solution for final imaging
Signal Enhancement Strategies:
Consider tyramide signal amplification to boost FITC signal in thick specimens
Use two-photon excitation to improve penetration depth and reduce phototoxicity
Implement light-sheet microscopy for rapid volumetric imaging with reduced photobleaching
Analysis Considerations:
Apply 3D deconvolution algorithms to improve signal clarity
Implement tissue registration to anatomical atlases
Use 3D rendering software to visualize NENF distribution patterns throughout tissue volumes
These approaches enable unprecedented insights into the three-dimensional organization of NENF expression patterns across intact neural tissues, providing context impossible to achieve with traditional section-based methods .
Future developments in antibody technology will likely enhance NENF detection capabilities:
Engineered Antibody Formats:
Single-chain variable fragments (scFvs) against NENF: Smaller size enables better tissue penetration
Nanobodies derived from camelid antibodies: ~15 kDa size improves access to sterically hindered epitopes
Bispecific antibodies: Simultaneous detection of NENF and interaction partners
Advanced Fluorescent Technologies:
Self-labeling protein tags (SNAP, CLIP, Halo) for versatile fluorophore attachment
Photoactivatable or photoswitchable fluorophores for super-resolution applications
Near-infrared fluorescent proteins for deeper tissue imaging with reduced autofluorescence
Smart Antibody Systems:
Fluorescence resonance energy transfer (FRET)-based reporters for detecting NENF-protein interactions
Split-fluorescent protein systems for visualization of NENF proximity to other proteins
pH-sensitive fluorophore conjugates to track NENF internalization and trafficking
Methodological Implementation:
Generate recombinant antibody fragments against specific NENF domains
Validate binding affinity and epitope specificity
Optimize conjugation strategies for novel fluorophores
Compare performance metrics against conventional antibodies
These advancements will enable more precise spatial localization, dynamic monitoring of NENF distribution, and multiplexed detection of NENF alongside other biomolecules of interest, expanding research capabilities beyond what current FITC-conjugated antibodies provide .
NENF's established roles in neuronal survival and neural progenitor cell proliferation position it as a key target in neuroregeneration research:
Neural Stem Cell Investigations:
Track NENF expression during neural stem cell differentiation
Investigate correlation between NENF levels and regenerative capacity
Assess whether NENF supplementation enhances neurogenesis in vitro and in vivo
Methodological Approach:
Isolate neural stem/progenitor cells from appropriate sources
Induce differentiation under various conditions
Use NENF Antibody, FITC conjugated for live or fixed-cell imaging
Correlate NENF expression with stem cell markers and differentiation outcomes
Manipulate NENF levels through genetic or pharmacological approaches
Injury Response Studies:
Monitor NENF expression patterns following neural injury
Identify cell populations upregulating NENF during recovery phases
Test whether NENF modulation affects regenerative outcomes
Translational Applications:
Screen compounds for effects on NENF expression in neural precursors
Develop therapeutic strategies targeting NENF-related pathways
Use NENF immunolabeling as a biomarker for regenerative capacity
This research direction leverages NENF Antibody, FITC conjugated as both an analytical tool and a means to validate NENF as a potential therapeutic target in neuroregeneration contexts .
Advanced quantitative analysis of NENF immunofluorescence requires sophisticated computational approaches:
Image Preprocessing Pipeline:
Apply flat-field correction to compensate for illumination heterogeneity
Implement background subtraction algorithms specific to tissue autofluorescence patterns
Apply deconvolution to improve signal-to-noise ratio
Register multi-channel images to correct for chromatic aberration
Segmentation Strategies:
Implement machine learning-based segmentation to identify NENF-positive structures
Use watershed algorithms for separating closely packed cells
Apply intensity thresholding based on control samples
Incorporate nuclear counterstains as anchors for cell identification
Quantitative Feature Extraction:
Measure intensity parameters: mean, integrated density, maximum intensity
Analyze morphological features: area, perimeter, shape descriptors
Quantify texture features: entropy, contrast, homogeneity
Assess spatial relationships: nearest neighbor distance, clustering coefficients
Statistical Analysis Framework:
Apply appropriate normalization to account for technical variations
Use hierarchical linear models to account for nested experimental designs
Implement multiple testing correction for high-dimensional data
Visualize results using dimensionality reduction techniques
These computational approaches transform qualitative NENF staining patterns into quantitative metrics suitable for robust statistical comparison across experimental conditions, enabling detection of subtle changes in NENF expression or localization .