FITC-conjugated KALRN antibodies are pivotal in studying KALRN’s role in cellular signaling and disease:
Neuronal Development: KALRN regulates actin dynamics and dendritic spine formation, impacting synaptic plasticity .
Cancer Research: KALRN mutations correlate with enhanced antitumor immunity and response to immunotherapy, as shown in TCGA cohort analyses .
Immunofluorescence: Used to visualize KALRN localization in cultured cells (e.g., HeLa) and tissue sections (e.g., brain, colon) .
Recent studies leveraging FITC-conjugated KALRN antibodies include:
Mechanistic Insights: KALRN deficiency in tumor cells increases PD-L1 expression and CD8+ T-cell infiltration, sensitizing cancers to immune checkpoint inhibitors .
Cytoskeletal Regulation: KALRN activates Rho GTPases (e.g., Rac1), influencing cell motility and neuronal morphology .
Validation in Models: In vivo experiments using KALRN-depleted tumors demonstrated enhanced immune cell recruitment and therapeutic response to PD-1 inhibitors .
Commercial FITC-conjugated KALRN antibodies vary in immunogen design and validation:
Supplier | Catalog # | Immunogen Region | Applications | Reactivity |
---|---|---|---|---|
Bioss | bs-11861R-FITC | 1401–1500/2985 | IF, IHC, ICC | Human, Mouse, Rat, Dog |
Abbexa | abxxxxxx | 2410–2661 AA | IF, WB | Human |
GlycoTech | CSB-PA011981 | Not specified | IF, IHC | Human |
KALRN (kalirin RhoGEF kinase) is a protein that activates specific Rho GTPase family members to regulate neuronal function and the actin cytoskeleton . This gene exhibits mutations across diverse cancer types, including melanoma, lung cancer, uterine corpus endometrial carcinoma (UCEC), glioblastoma multiforme (GBM), and colorectal cancer (COAD) . While historically understudied, recent evidence demonstrates that KALRN mutations significantly enhance antitumor immunity and promote favorable responses to immune checkpoint blockade therapy . The mechanism appears to involve compromised DNA damage repair pathways, leading to increased tumor mutation burden and subsequent enhancement of immune recognition .
When selecting KALRN Antibody, FITC Conjugated for experimental applications, researchers should evaluate the following specifications:
Parameter | Specification Details |
---|---|
Source | KLH conjugated synthetic peptide derived from human KALRN/Duo |
Host | Rabbit |
Clonality | Polyclonal |
Immunogen Range | 1401-1500/2985 |
Isotype | IgG |
Standard Concentration | 1μg/μl |
Storage Buffer | Aqueous buffered solution containing 0.01M TBS (pH 7.4) with 1% BSA, 0.03% Proclin300 and 50% Glycerol |
Subcellular Localization | Cytoplasm |
Validated Applications | IF(IHC-P), IF(IHC-F), IF(ICC) |
Cross-Reactivity | Human, Mouse, Rat, Dog, Pig, Horse, Chicken |
These specifications ensure optimal performance across various experimental contexts while minimizing non-specific binding and background issues .
KALRN Antibody, FITC Conjugated requires specific storage conditions to maintain its structural integrity and fluorescent signal. The antibody should be stored at -20°C and divided into multiple aliquots to prevent repeated freeze-thaw cycles that could lead to protein degradation . Light exposure should be minimized due to FITC's photosensitivity. Working solutions should be prepared fresh and used within 24 hours. When handling the antibody, researchers should wear appropriate PPE to prevent contamination and degradation of both the antibody and the handler.
When employing KALRN Antibody, FITC Conjugated for tumor microenvironment studies, protocol optimization should address several critical parameters:
Fixation method selection: For KALRN detection in paraffin-embedded sections, antigen retrieval methods should be carefully optimized, as KALRN epitopes may be sensitive to overfixation with certain fixatives.
Blocking optimization: Given KALRN's cytoplasmic localization, use 5-10% normal serum from the same species as the secondary antibody (though unnecessary with direct conjugates) plus 0.1-0.3% Triton X-100 for permeabilization.
Antibody concentration titration: Perform serial dilutions (1:50 to 1:500) of the antibody to determine optimal signal-to-noise ratio for each tissue or cell type.
Multiplexing considerations: When co-staining with markers of tumor-infiltrating lymphocytes, consider the spectral overlap between FITC (excitation: 495nm, emission: 519nm) and other fluorophores.
Validation controls: Include KALRN-mutated and KALRN-wildtype samples as positive and comparative controls, respectively, based on the differential expression patterns observed in research studies .
To effectively investigate relationships between KALRN mutation status and immune infiltration, researchers should implement a multi-modal analytical approach:
Sequential immunofluorescence: Perform KALRN antibody staining followed by immune cell marker staining (CD8, NK cells, M1/M2 macrophages) on sequential sections to correlate expression patterns.
Quantitative image analysis: Employ digital pathology software to quantify:
KALRN expression intensity
Immune cell density (cells/mm²)
Spatial relationships between KALRN-expressing cells and immune infiltrates
Correlation with genomic data: Integrate immunofluorescence findings with genomic analyses of KALRN mutation status.
Functional validation: Conduct in vitro co-culture experiments similar to those described in research studies, where NK cells co-cultured with KALRN-knockdown tumor cells demonstrated significantly stronger proliferation capacity than those with KALRN-wildtype cells .
This multi-dimensional approach allows researchers to establish meaningful connections between KALRN mutations and antitumor immune responses.
High background fluorescence can significantly compromise data quality in KALRN immunofluorescence studies. Implement these evidence-based troubleshooting approaches:
Optimize antibody concentration: Titrate the antibody (typically starting at 1:100 and adjusting as needed) to determine the minimal concentration that maintains specific signal while reducing background.
Enhance blocking protocols: Utilize 5-10% serum from the species unrelated to the primary antibody plus 1% BSA to minimize non-specific binding.
Address autofluorescence: For tissue samples (particularly those with high collagen content), consider:
Brief treatment with 0.1% Sudan Black B in 70% ethanol
10mM CuSO₄ in 50mM ammonium acetate buffer (pH 5.0) treatment for 10-30 minutes
Photobleaching by pre-exposure to light or treatment with 0.3% H₂O₂
Optimize washing procedures: Implement extended washing periods (minimum 3 washes, 5-10 minutes each) with 0.05-0.1% Tween-20 in PBS.
Consider sample-specific factors: Different fixation protocols may be required for optimal results across diverse tissue/cell types.
Rigorous control implementation is critical for ensuring result validity when using KALRN Antibody, FITC Conjugated:
Implementing this comprehensive control strategy ensures that observed signals accurately represent KALRN protein expression rather than technical artifacts.
When analyzing KALRN expression in relation to tumor-infiltrating lymphocytes, researchers should implement a systematic quantitative approach:
Multi-parameter image segmentation:
Define regions of interest (tumor nests, invasive margins, stromal compartments)
Apply automated cell identification algorithms for KALRN+ cells and immune cell subpopulations
Calculate density metrics (cells/mm²) for each cell population
Spatial relationship analysis:
Measure distances between KALRN+ cells and nearest CD8+ T cells or NK cells
Generate spatial correlation indices to quantify co-localization patterns
Develop neighborhood enrichment metrics to identify significant spatial associations
Expression intensity quantification:
Measure mean fluorescence intensity of KALRN staining
Categorize cells based on expression levels (negative, low, medium, high)
Correlate KALRN expression intensity with immune cell infiltration metrics
Research has shown that KALRN-mutated tumors display significantly higher immune infiltration, with elevated ratios of immune-stimulatory to immune-inhibitory signatures (CD8+/CD4+ regulatory T cells, M1/M2 macrophages, and proinflammatory/anti-inflammatory cytokines) .
To effectively interpret KALRN expression data for immunotherapy response prediction, researchers should employ a multi-dimensional analytical framework:
Integrative biomarker assessment: Analyze KALRN expression in conjunction with established immunotherapy response biomarkers:
Outcome correlation analysis: Multiple studies demonstrate that KALRN mutations correlate with significantly higher immunotherapy response rates across multiple cancer cohorts:
Mechanistic interpretation: Consider how KALRN mutations compromise DNA damage repair pathways, leading to increased neoantigen load and enhanced immune recognition .
This comprehensive interpretive approach allows researchers to contextualize KALRN expression data within the broader immunological landscape of tumor microenvironments.
For comprehensive immune profiling using multiplex immunofluorescence incorporating KALRN Antibody, FITC Conjugated:
Panel design considerations:
Pair KALRN (FITC) with complementary markers in non-overlapping channels:
CD8 (for cytotoxic T cells)
CD56 (for NK cells)
CD68/CD163 (for macrophage polarization)
PD-L1 (for checkpoint expression)
Include nuclear counterstain (DAPI/Hoechst) in far blue spectrum
Sequential staining protocols:
Implement tyramide signal amplification (TSA) for signal enhancement
Use appropriate spectral unmixing algorithms for resolving overlapping emissions
Consider cyclical immunofluorescence for expanded marker panels
Analytical workflow:
Develop computational pipelines for cell phenotyping
Employ machine learning algorithms for pattern recognition
Generate high-dimensional data visualizations (tSNE, UMAP)
This multiplex approach enables researchers to simultaneously assess KALRN expression, immune cell composition, and functional states within the tumor microenvironment.
To investigate functional relationships between KALRN status and DNA damage repair mechanisms, researchers should consider these experimental designs:
KALRN knockdown/knockout models:
Generate stable KALRN knockdown cell lines using shRNA or CRISPR-Cas9
Validate reduced KALRN expression using the FITC-conjugated antibody
Assess DNA damage repair efficiency through comet assays, γH2AX foci formation, and homologous recombination reporter assays
DNA damage induction experiments:
Treat KALRN-wildtype and KALRN-deficient cells with DNA-damaging agents
Monitor repair kinetics using time-course immunofluorescence with KALRN antibody and DNA damage markers
Quantify persistent damage foci as indicators of repair deficiency
Mechanistic studies:
Investigate KALRN interaction with Rho GTPases involved in DNA repair using co-immunoprecipitation
Assess chromosome instability through metaphase spreads and FISH analysis
Monitor DNA repair protein recruitment to damage sites in KALRN-manipulated cells
Research has demonstrated that KALRN mutations compromise the function of KALRN in targeting Rho GTPases for the regulation of DNA damage repair pathways, leading to increased mutation burden and enhanced immunogenicity .
Several promising research directions could be enhanced through application of KALRN Antibody, FITC Conjugated:
Biomarker development for immunotherapy stratification: Given the strong correlation between KALRN mutations and immunotherapy response across multiple cancer cohorts , developing KALRN-based predictive assays could significantly improve patient selection.
Combination therapy approaches: Investigating how KALRN status affects response to combined immunotherapy and DNA damage repair-targeted therapies could reveal novel therapeutic strategies.
Single-cell analysis of KALRN expression heterogeneity: Applying KALRN antibody in single-cell imaging or flow cytometry could reveal intratumoral heterogeneity in KALRN expression and its relationship to immune evasion mechanisms.
Development of KALRN-targeting therapeutic approaches: Understanding how KALRN mutations enhance antitumor immunity could inform the development of therapeutic strategies mimicking these effects in KALRN-wildtype tumors.