FITC conjugation involves covalent bonding of the dye to primary amines (e.g., lysine residues) on the antibody. Key steps include:
Purification: Removal of sodium azide to prevent interference with FITC reactivity .
Titration: Optimizing FITC-to-antibody ratios (typically 3–6 FITC molecules per antibody) to balance brightness and solubility .
Validation: Testing conjugates for signal intensity and background noise via staining titrations .
Quenching Risk: Excessive FITC molecules reduce fluorescence efficiency.
Stability: FITC-conjugated antibodies are light-sensitive and require storage at -20°C .
ERVFRD-1 expression in KIRC is linked to distinct immune phenotypes:
Supplier | Conjugate | Applications | Reactivity | Price (USD) | Citations |
---|---|---|---|---|---|
Antibodies-online | FITC | IF | Human | $470.46 | N/A |
Biomatik | Unconjugated | ELISA, IHC | Human | $314.00 | N/A |
Abcam (ab230235) | Unconjugated | WB | Human | N/A | 1 publication |
FITC-Conjugated Antibodies: Primarily used for IF to visualize ERVFRD-1 subcellular localization .
Unconjugated Antibodies: Suitable for Western blotting or ELISA .
Cross-Reactivity: Potential off-target binding due to structural similarities with other retroviral envelope proteins .
Conjugation Efficiency: Optimal FITC-to-antibody ratios require iterative testing .
ERVFRD-1 is an endogenous retroviral envelope glycoprotein with a canonical length of 538 amino acid residues and a mass of 59.5 kDa. It belongs to the Gamma type-C retroviral envelope protein family and is primarily localized in the cell membrane . The protein plays a major role in placental development and trophoblast fusion .
ERVFRD-1 has the characteristics of a typical retroviral envelope protein, featuring a cleavage site that separates the surface (SU) and transmembrane (TM) proteins, which form a heterodimer . It is expressed at notably high levels in the placenta, suggesting functional importance in human reproduction . The gene is part of a human endogenous retrovirus provirus on chromosome 6 that has inactivating mutations in the gag and pol genes, while the envelope glycoprotein gene appears to have been selectively preserved through evolution .
For FITC-conjugated ERVFRD-1 antibodies, the following detection methods are recommended:
Method | Optimal Dilution | Detection Sensitivity | Main Applications |
---|---|---|---|
Flow Cytometry | 1:100-1:500 | Medium-High | Cell surface expression quantification |
Immunofluorescence | 1:200-1:500 | High | Subcellular localization studies |
Confocal Microscopy | 1:200-1:400 | Very High | Co-localization with other proteins |
FACS | 1:100-1:300 | High | Cell sorting based on expression |
When using these methods, researchers should optimize protocols by titrating antibody concentrations, as ERVFRD-1 expression varies significantly between tissue types, with particularly high expression in placental tissues . Flow cytometry can effectively quantify cell surface expression, while immunofluorescence and confocal microscopy are ideal for examining subcellular localization patterns, particularly in studying membrane fusion events in trophoblast research.
Including appropriate controls is critical for reliable interpretation of results with FITC-conjugated ERVFRD-1 antibodies:
Isotype Control: Use a FITC-conjugated antibody of the same isotype but with irrelevant specificity to assess non-specific binding.
Negative Tissue Control: Include tissues known to express minimal ERVFRD-1 (non-placental tissues) to establish background signal levels .
Positive Tissue Control: Placental tissue samples should be included as positive controls due to known high expression of ERVFRD-1 .
Absorption Controls: Pre-incubate the antibody with purified ERVFRD-1 protein before application to verify specificity.
Secondary-only Control: For indirect detection methods, include samples treated only with secondary reagents to assess non-specific binding.
These controls help distinguish true signal from autofluorescence or non-specific binding, particularly important in tissues with high autofluorescence like kidney tissue when studying KIRC .
Quantification of ERVFRD-1 expression differences between normal and cancerous tissues requires rigorous methodological approaches:
Digital Image Analysis: For immunofluorescence studies, use software like ImageJ with consistent thresholding parameters across all samples.
Mean Fluorescence Intensity (MFI) Measurement: For flow cytometry, compare MFI values between normal and cancer samples with proper normalization to account for background fluorescence.
Reference Gene Normalization: When quantifying at the transcript level alongside protein detection, normalize to multiple reference genes for accurate comparison.
ROC Curve Analysis: Generate receiver operating characteristic curves to determine optimal cut-off values for distinguishing normal from cancerous tissues. In KIRC studies, ERVFRD-1 expression demonstrated significant discriminatory power with an area under the curve (AUC) of 0.952 (95% CI=0.932-0.972) .
Research has shown consistently lower ERVFRD-1 expression in KIRC compared to normal kidney tissues (p < 0.001) across multiple cohorts, making proper quantification essential for biomarker development .
For optimal detection of ERVFRD-1 using FITC-conjugated antibodies, fixation and permeabilization protocols should be carefully selected:
Fixation Method | Recommended Duration | Advantages | Considerations |
---|---|---|---|
4% Paraformaldehyde | 10-15 minutes | Preserves membrane proteins | May reduce epitope accessibility |
Methanol/Acetone (1:1) | 10 minutes at -20°C | Enhanced permeabilization | May disrupt some epitopes |
2% Formaldehyde + 0.1% Glutaraldehyde | 5-10 minutes | Strong fixation for long-term storage | May increase autofluorescence |
For permeabilization:
0.1-0.5% Triton X-100 (5-10 minutes) for intracellular epitopes
0.1% Saponin for gentler permeabilization with less disruption of membrane structures
Since ERVFRD-1 is a membrane-localized protein, avoid excessive permeabilization which might disrupt membrane integrity and alter staining patterns . For placental tissues, which express high levels of ERVFRD-1, shorter fixation times may be sufficient, while tissues with lower expression (such as KIRC samples) may require optimized antigen retrieval methods .
ERVFRD-1 has shown significant involvement in tumor immunoregulation, particularly in KIRC. To investigate these relationships:
Multiplex Immunofluorescence: Combine FITC-conjugated ERVFRD-1 antibodies with antibodies against immune cell markers (using different fluorophores) to assess co-localization. Research has shown close relationships between ERVFRD-1 expression and infiltration levels of mast cells and Treg cells (P < 0.001) .
Single-cell Analysis Workflow:
Disaggregate tumor tissue into single-cell suspensions
Stain with FITC-conjugated ERVFRD-1 antibody alongside immune cell markers
Perform flow cytometry or mass cytometry analysis
Use dimensionality reduction techniques (tSNE, UMAP) to visualize relationships
Spatial Analysis: Employ techniques like multiplexed immunofluorescence or imaging mass cytometry to maintain spatial context. This approach has revealed that ERVFRD-1 expression patterns correlate with distinct immune microenvironments in KIRC .
Analysis using ssGSEA (single-sample Gene Set Enrichment Analysis) techniques has revealed correlations between ERVFRD-1 expression and immune cell infiltration scores, suggesting functional relationships in the tumor microenvironment that can be further investigated at the protein level using FITC-conjugated antibodies .
Researchers investigating ERVFRD-1 across cancer types may encounter contradictory findings. To resolve these discrepancies:
Tissue-Specific Context Analysis: ERVFRD-1 shows different expression patterns across cancer types. For example, pan-cancer analysis revealed generally low expression in multiple cancers including BLCA, BRCA, CESC, and KIRC, yet its prognostic significance varies . Use FITC-conjugated antibodies with identical protocols across multiple cancer types for direct comparison.
Isoform-Specific Detection: Design experiments to distinguish between potential ERVFRD-1 isoforms:
Use antibodies targeting different epitopes
Combine with RT-PCR to identify transcript variants
Compare results with computational predictions of isoform expression
Methylation-Expression Correlation: Analyze methylation patterns alongside protein expression using:
Multi-omics Integration Framework:
Data Type | Analysis Method | Integration Approach |
---|---|---|
Protein Expression (FITC signal) | Quantitative IF/Flow cytometry | Correlation with other data types |
Transcriptomic | RNA-seq differential expression | Compare protein vs. mRNA trends |
Epigenetic | Methylation arrays, ChIP-seq | Identify regulatory mechanisms |
Clinical | Survival analysis, multivariate Cox | Connect molecular findings to outcomes |
This integrated approach has helped clarify that in KIRC, lower ERVFRD-1 expression correlates with poorer outcomes, despite its role in other cancers potentially being different .
ERVFRD-1 (syncytin-2) plays a crucial role in trophoblast fusion. To study these mechanisms:
Live Cell Imaging Protocol:
Use cell-permeable nuclear dyes in combination with FITC-conjugated ERVFRD-1 antibody fragments
Employ spinning disk confocal microscopy with environmental control
Capture time-lapse images at 5-minute intervals for 24-48 hours
Analyze membrane dynamics during fusion events
Fusion Assay Quantification:
Seed differentially labeled cells (one population with FITC-ERVFRD-1 antibody)
Measure fusion index: (number of nuclei in syncytia/total number of nuclei) × 100
Track ERVFRD-1 localization before, during, and after fusion events
Mutation-Function Analysis:
Generate cells expressing wild-type or mutant ERVFRD-1
Label with FITC-conjugated antibodies targeting preserved epitopes
Quantify fusion efficiency relative to ERVFRD-1 surface expression levels
Research has demonstrated that ERVFRD-1 exhibits fusogenic properties, with in vitro experiments showing that cells transfected with syncytin-2 displayed altered tumorigenic potential when engrafted into mice, suggesting complex roles beyond simple membrane fusion .
To evaluate ERVFRD-1 as an immunotherapy target in KIRC:
Epitope Accessibility Assessment:
Use FITC-conjugated ERVFRD-1 antibodies of different epitope specificities
Quantify binding to live, non-permeabilized cells from patient-derived xenografts
Determine which epitopes are accessible in the native tumor environment
Immune Response Characterization:
Co-culture KIRC cells with immune effector cells
Add FITC-conjugated ERVFRD-1 antibodies to monitor target engagement
Assess cytokine production and cytotoxicity
Predictive Biomarker Analysis:
Correlate ERVFRD-1 expression (measured by FITC signal intensity) with:
Tumor Mutation Burden (TMB)
PD-L1 expression
Immune cell infiltration scores
Response to immune checkpoint inhibitors (retrospective analysis)
In vivo Targeting Validation:
Develop ERVFRD-1-targeted constructs (CAR-T, BiTEs, ADCs)
Use FITC-conjugated antibodies to confirm target engagement
Monitor efficacy in preclinical models
Research suggests ERVFRD-1 involvement in tumor immunoregulation, showing close relationships with immune cell infiltration, particularly mast cells and Treg cells . The TIDE algorithm can be employed to predict potential immune checkpoint blockade responses based on ERVFRD-1 expression patterns .
To correlate ERVFRD-1 methylation with protein expression:
Integrated Methylation-Expression Analysis Pipeline:
Bisulfite sequencing of ERVFRD-1 promoter and gene body
Immunofluorescence with FITC-conjugated ERVFRD-1 antibodies on serial sections
Correlation analysis between methylation beta values and fluorescence intensity
Cell Line Demethylation Experiments:
Treat KIRC cell lines with demethylating agents (5-aza-2'-deoxycytidine)
Monitor changes in ERVFRD-1 expression using FITC-conjugated antibodies
Quantify expression changes via flow cytometry
CpG Site-Specific Analysis:
Methylation Site | Correlation with Expression | Clinical Significance |
---|---|---|
TSS1500 region | Strong negative correlation | Associated with survival |
Gene body | Variable correlation | Tissue-specific patterns |
3' UTR | Weak correlation | Minimal impact on expression |
Single-Cell Correlation Analysis:
Perform single-cell bisulfite sequencing
Match with single-cell protein quantification using FITC-conjugated antibodies
Develop computational methods to integrate these data types
The MethSurv database can be used to evaluate DNA methylation status of ERVFRD-1 and its prognostic value, as demonstrated in KIRC research . This approach can uncover the underlying epigenetic mechanisms controlling ERVFRD-1 expression and potentially explain the observed lower expression in KIRC tumors compared to normal kidney tissues.
For advanced spatial profiling of ERVFRD-1 in the tumor microenvironment:
Cyclic Immunofluorescence (CycIF) Protocol:
Apply FITC-conjugated ERVFRD-1 antibody in the first cycle
Image and record coordinates
Chemically inactivate fluorescence
Repeat with antibodies against immune markers, basement membrane components, etc.
Computational alignment and analysis of 30+ markers on the same tissue section
CODEX System Integration:
Conjugate ERVFRD-1 antibodies with DNA barcodes
Perform multiplexed detection with other markers
Use computational analysis to identify cellular neighborhoods and spatial relationships
Spatial Transcriptomics Correlation:
Perform immunofluorescence with FITC-conjugated ERVFRD-1 antibodies
Execute spatial transcriptomics on adjacent sections
Register images and correlate protein expression with gene expression domains
These approaches enable researchers to examine ERVFRD-1's relationship with the tumor microenvironment, particularly relevant given findings showing ERVFRD-1's involvement in tumor immunoregulation and its correlation with infiltration levels of specific immune cell populations in KIRC .
To develop and validate ERVFRD-1 as a prognostic marker:
Cohort Design Requirements:
Training cohort: minimum 200 KIRC cases with complete follow-up
Validation cohort: independent set of 100+ cases
Controls: matched normal kidney tissue
Stratification by clinical parameters (T stage, M stage, etc.)
Multivariate Biomarker Panel Development:
Survival Analysis Framework:
Analysis Type | Statistical Method | Software Implementation |
---|---|---|
Univariate | Kaplan-Meier | R survival package |
Multivariate | Cox Regression | R survival package |
Nomogram | rms package | R rms package |
Cut-off Determination | maxstat method | R maxstat package |
Validation Methods:
Internal: bootstrap resampling
External: independent cohorts
Calibration plots
Time-dependent ROC curves
Concordance index (C-index)
Ensuring antibody specificity is crucial for reliable research outcomes:
Cross-Reactivity Assessment Protocol:
Test on cells with CRISPR-mediated ERVFRD-1 knockout
Evaluate binding to tissues from other species with known ERVFRD-1 homologs
Screen against related human endogenous retroviral proteins
Absorption Controls Implementation:
Pre-absorb antibody with recombinant ERVFRD-1 protein
Include graduated concentrations of competing antigens
Quantify reduction in signal intensity
Epitope Mapping:
Use overlapping peptide arrays to precisely identify binding epitopes
Select antibodies targeting unique regions not shared with other ERV proteins
Verify epitope conservation across experimental models
Western Blot Validation Standards:
ERVFRD-1 belongs to the Gamma type-C retroviral envelope protein family and shares sequence similarities with other ERV proteins, making specificity validation particularly important . Additionally, post-translational modifications including protein cleavage and glycosylation can affect antibody binding and should be considered when validating specificity .
To investigate evolutionary aspects of ERVFRD-1:
Cross-Species Comparison Methodology:
Test FITC-conjugated human ERVFRD-1 antibodies on tissues from evolutionary related species
Map epitope conservation using sequence alignment tools
Correlate binding affinity with functional conservation
Phylogenetic Analysis Protocol:
Functional Conservation Assessment:
Compare fusion activity in trophoblast models from different species
Correlate expression patterns with placentation strategies
Examine immune regulatory functions across species
This research direction is particularly relevant as ERVFRD-1 gene orthologs have been reported in mouse and chimpanzee species , suggesting evolutionary conservation of function that may provide insights into both normal physiology and pathological roles in cancer.
To explore ERVFRD-1's potential role in predicting immunotherapy responses:
Integrated Biomarker Analysis Workflow:
Quantify ERVFRD-1 expression using FITC-conjugated antibodies via flow cytometry or immunofluorescence
Determine tumor mutation burden (TMB) through next-generation sequencing
Calculate correlation coefficients between ERVFRD-1 expression and TMB
Apply the TIDE algorithm to predict immune checkpoint blockade responses
Patient Stratification Approach:
Divide patients into quadrants based on ERVFRD-1 expression and TMB
Track clinical outcomes and treatment responses
Identify optimal cut-off values for both markers
Multiparametric Flow Cytometry Protocol:
Panel design: FITC-ERVFRD-1, immune checkpoint markers, T-cell activation markers
Gating strategy focused on tumor and immune cell populations
Correlation analysis with genomic markers and clinical outcomes
Research has shown that using the ggstatsplot R package can analyze the correlation between TMB enrichment scores and ERVFRD-1 expression, while the TIDE algorithm can predict potential immunotherapy responses based on ERVFRD-1 expression patterns . These computational approaches can be validated and extended using protein-level analyses with FITC-conjugated antibodies.
To investigate ERVFRD-1's role in signaling pathways:
Phospho-Proteomics Integration:
Sort cells based on ERVFRD-1 expression using FITC-conjugated antibodies
Perform phospho-proteomic analysis on sorted populations
Map activated signaling pathways using pathway enrichment tools
Live Cell Signaling Analysis:
Use FITC-conjugated ERVFRD-1 antibodies alongside calcium indicators or FRET-based reporters
Monitor real-time signaling changes in relation to ERVFRD-1 expression
Quantify signaling dynamics in single cells
Pathway Inhibition Matrix:
Pathway | Inhibitor | Effect on ERVFRD-1 Expression | Effect on Phenotype |
---|---|---|---|
PI3K/AKT | LY294002 | To be determined experimentally | Cell survival impact |
MAPK | PD98059 | To be determined experimentally | Proliferation effects |
JAK/STAT | Ruxolitinib | To be determined experimentally | Immune modulation |
GO, KEGG, and GSEA Analysis Framework:
Perform differential gene expression analysis between high and low ERVFRD-1 expressing cells
Conduct GO term enrichment to identify biological processes
Apply KEGG pathway analysis to map molecular interactions
Execute GSEA to identify enriched gene sets
Previous research using GO, KEGG, and GSEA analyses revealed significant involvement of ERVFRD-1 in tumor immunoregulation in KIRC . These computational findings can be validated at the protein level using FITC-conjugated antibodies in combination with signaling pathway analysis.
When encountering signal issues with FITC-conjugated ERVFRD-1 antibodies:
Signal Optimization Protocol:
Titrate antibody concentration (1:50, 1:100, 1:200, 1:500)
Extend incubation time (1 hour, 2 hours, overnight at 4°C)
Test different fixation methods (paraformaldehyde, methanol, acetone)
Implement antigen retrieval (citrate buffer pH 6.0, EDTA buffer pH 9.0)
Autofluorescence Reduction Techniques:
Treat sections with 0.1% Sudan Black B in 70% ethanol (20 minutes)
Use commercially available autofluorescence quenchers
Employ spectral unmixing during imaging
Antibody Quality Control Checklist:
Tissue-Specific Optimization:
For KIRC samples: Implement extended antigen retrieval
For placental tissue: Reduce antibody concentration due to high expression
For cultured cells: Optimize fixation to preserve membrane localization
Since ERVFRD-1 has generally low expression in KIRC tumors compared to normal tissue , signal optimization is particularly important when studying this protein in cancer contexts.
To ensure antibody specificity:
Comprehensive Validation Framework:
Multi-platform Confirmation Approach:
Compare FITC-antibody results with RNA-seq data
Validate with alternative antibody clones targeting different epitopes
Confirm with orthogonal techniques (Western blot, ELISA)
Cross-reactivity Assessment Matrix:
Test | Method | Expected Result | Troubleshooting |
---|---|---|---|
Epitope blocking | Pre-incubation with peptide | Signal elimination | Increase peptide concentration |
Secondary-only | Omit primary antibody | No signal | Check secondary antibody specificity |
Isotype control | Irrelevant antibody, same isotype | No signal | Reduce antibody concentration |
Knockout validation | CRISPR-Cas9 | No signal | Verify knockout efficiency |
Western Blot Confirmation Standards:
Proper validation is essential since ERVFRD-1 belongs to a family of related endogenous retroviral proteins with potential for cross-reactivity .
Emerging approaches for ERVFRD-1 research include:
Advanced Imaging Methodologies:
Super-resolution microscopy: Resolve ERVFRD-1 localization at nanometer scale
Light-sheet microscopy: 3D imaging of ERVFRD-1 in tumor spheroids
Expansion microscopy: Physical tissue expansion for improved resolution
Live-cell lattice light-sheet: Dynamic visualization of ERVFRD-1 trafficking
Single-Cell Multi-omics Integration:
CITE-seq combining ERVFRD-1 antibody detection with transcriptomics
Single-cell Western blot for protein validation
Spatial proteomics to map ERVFRD-1 interactions
CRISPR-Based Functional Screens:
CRISPRa/CRISPRi libraries targeting ERVFRD-1 regulatory elements
CRISPR base editing to study specific mutations
Prime editing for precise genomic modifications
In Situ Sequencing Applications:
Visualize ERVFRD-1 mRNA alongside protein using padlock probes
Multiplex RNA and protein detection in tissue sections
Spatial transcriptomics correlation with protein expression
These technologies could help clarify ERVFRD-1's role in regulating immunological activity within the tumor microenvironment and its potential as a biomarker for diagnosis, immunotherapy, and prognosis assessment of KIRC .
Based on current knowledge, potential therapeutic applications include:
Immunotherapy Target Development:
Design chimeric antigen receptor T cells (CAR-T) targeting ERVFRD-1
Develop bispecific T-cell engagers (BiTEs) linking T cells to ERVFRD-1+ cells
Create antibody-drug conjugates for targeted delivery
Evaluate immune checkpoint inhibitor combinations
Epigenetic Modulation Approach:
Target methylation status of ERVFRD-1 regulatory regions
Develop small molecules to modulate ERVFRD-1 expression
Test combination with existing epigenetic drugs
Precision Medicine Framework:
Functional Screening Pathway:
High-throughput drug screening in models with varied ERVFRD-1 expression
Synthetic lethality approaches with ERVFRD-1 expression status
Identification of downstream vulnerabilities in ERVFRD-1-regulated pathways