The C1RL antibody is a rabbit IgG polyclonal antibody designed to bind specifically to the C1RL protein, a serine protease analog involved in immune regulation and cancer biology. Its Y-shaped structure, typical of immunoglobulins, includes:
Fab domains: Antigen-binding regions (arms of the "Y") that interact with C1RL epitopes.
Fc domain: Mediates interactions with effector cells, enabling downstream applications like immunoprecipitation or ELISA .
Application | Dilution Range | Tested Samples |
---|---|---|
Western Blot (WB) | 1:500–1:3000 | Human plasma |
Immunofluorescence (IF/ICC) | 1:200–1:800 | U2OS cells |
ELISA | Variable | Human/mouse lysates |
Purity: Antigen-affinity purified to ensure specificity.
Storage: PBS with 0.02% sodium azide and 50% glycerol (-20°C) .
Cancer Prognosis:
C1RL is overexpressed in glioblastoma and serves as a prognostic marker in hepatocellular carcinoma (HCC) and renal cell carcinoma (RCC). Studies using the C1RL antibody demonstrate its utility in detecting protein levels via WB and immunohistochemistry .
Observed Molecular Weight: ~70 kDa (due to N-glycosylation), despite a calculated weight of 53 kDa .
Epitope Targeting: Binds to a C1RL fusion protein (Ag20324), ensuring specificity .
Immunogen: Recombinant C1RL fusion protein (Ag20324).
Purification: Antigen-affinity chromatography to minimize cross-reactivity .
Positive WB Detection: 70 kDa band in human plasma (source: ).
IF/ICC Validation: Strong nuclear and cytoplasmic staining in U2OS cells .
Broader Context:
Large-scale antibody validation initiatives, such as the Protein Capture Reagent Program (PCRP), emphasize rigorous testing to avoid non-specific binding—a challenge highlighted in antibody characterization crises . The C1RL antibody aligns with these standards by providing application-specific optimization guidelines .
C1RL, also known as C1r-like serine protease analog protein (CLSPa), is a complement system protein containing a CUB domain and a serine protease domain with a characteristic catalytic triad. The protein plays a role in complement activation and has been implicated in several disease processes .
Based on current validation data, C1RL antibodies are suitable for multiple experimental applications:
Application | Recommended Dilution | Notes |
---|---|---|
Western Blotting | 0.04-0.4 μg/mL | Detects band at ~70 kDa |
Immunofluorescence | 0.25-2 μg/mL | Useful for subcellular localization |
Immunohistochemistry | 1:50-1:200 | Effective for tissue sections |
Indirect ELISA | Varies by antibody | Check manufacturer specifications |
The antibodies have demonstrated reactivity with human and mouse samples, making them suitable for comparative studies across these species .
Proper validation is critical for ensuring reliable results. A methodological approach includes:
Positive and negative control samples: Include tissues/cells known to express or lack C1RL expression.
Blocking peptide experiments: Use the immunizing peptide to demonstrate binding specificity.
siRNA knockdown: Confirm antibody specificity by reducing target protein expression.
Multiple antibody comparison: Validate results using antibodies targeting different epitopes of C1RL.
When validating polyclonal antibodies like those against C1RL, researchers should verify batch-to-batch consistency, especially when changing lots in long-term studies .
Recent studies indicate that C1RL is upregulated in glioblastoma (GBM) and has prognostic value in hepatocellular carcinoma and renal cell cancer . Analysis of 2,120 glioma patients across five public datasets revealed significant correlations between C1RL expression and clinical outcomes, suggesting its potential utility as a biomarker .
When designing studies to investigate C1RL in cancer:
Use carefully selected tissue microarrays with adequate controls
Employ standardized scoring systems for immunohistochemistry
Consider dual immunostaining to analyze C1RL in relation to other cancer markers
Correlate protein expression with transcriptomic data when available
The altered expression patterns make C1RL a promising target for prognostic studies, but researchers should establish appropriate thresholds for high versus low expression based on their specific experimental context and sample cohort.
Experimental conditions vary by application and sample type:
For Western Blotting:
Sample preparation should include protease inhibitors to prevent degradation
For glycosylated C1RL detection, avoid harsh reducing conditions
Use 8-10% SDS-PAGE gels for optimal separation
Expected band at 70 kDa (rather than the calculated 53 kDa) due to glycosylation
For Immunohistochemistry/Immunofluorescence:
Antigen retrieval methods: Heat-induced epitope retrieval in citrate buffer (pH 6.0)
Blocking: 5-10% normal serum from the same species as the secondary antibody
Recommended dilution range: 1:50-1:200 for IHC, 0.25-2 μg/mL for IF
Counterstain: Hematoxylin for IHC; DAPI for nuclear visualization in IF
C1RL antibodies can be incorporated into single-cell analysis workflows, particularly with platforms like 10x Genomics' Antibody Capture technology. This approach enables researchers to correlate protein expression with transcriptomic data at the single-cell level .
Key implementation steps:
Ensure your C1RL antibody is compatible with oligonucleotide conjugation
Include C1RL in your Feature Reference CSV file with "Antibody Capture" as the feature_type
Analyze data through visualization tools like Loupe Browser
Use log-transformed antibody counts for dimensionality reduction analysis
When interpreting results, be aware of potential aggregate formation, which can be identified through metrics provided in the aggregate_barcodes.csv output file from Cell Ranger analysis .
C1RL has been identified as having proteolytic activity within the complement cascade. Research has shown that prohaptoglobin is proteolytically cleaved in the endoplasmic reticulum by C1RL . This activity suggests C1RL plays an important role in protein processing beyond traditional complement activation.
In disease contexts, particularly cancer, C1RL may contribute to immunomodulation through:
Alteration of complement activation patterns
Potential involvement in creating immunosuppressive tumor microenvironments
Interaction with other proteases in the tumor milieu
For functional studies, researchers should consider both gain-of-function approaches (overexpression of C1RL) and loss-of-function studies (siRNA, CRISPR-Cas9) to elucidate its mechanistic contributions to disease processes.
To effectively study C1RL's prognostic value:
Patient cohort selection: Include adequate sample sizes with well-documented clinical data and follow-up
Tissue microarray design: Ensure representation of tumor heterogeneity with multiple cores per patient
Standardized IHC protocols: Use validated antibodies with consistent staining and scoring systems
Statistical analysis: Employ Kaplan-Meier survival analysis with multivariate Cox regression to account for confounding factors
Research shows C1RL may have particular relevance in glioma, where immunological and clinicopathological characteristics have been associated with patient outcomes . When designing these studies, consider:
Stratification by molecular subtypes
Integration with other established biomarkers
Correlation with treatment response data when available
To investigate protein-protein interactions involving C1RL:
Co-immunoprecipitation: Use C1RL antibodies to pull down protein complexes
Proximity ligation assay: Visualize and quantify protein interactions in situ
FRET/BRET: Examine real-time interactions in living cells
Yeast two-hybrid screening: Identify novel interaction partners
When designing antibody-based interaction studies, consider the epitope location in relation to potential binding domains. The immunogen sequence "NVLPVCLPDNETLYRSGLLGYVSGFGMEMGWLTTELKYSRLPVAPREACNAWLQKRQRPEVFSDNMFCVGDETQ" used for some commercial antibodies should be evaluated to ensure it doesn't interfere with interaction surfaces.
Researchers frequently encounter several challenges when working with C1RL antibodies:
Background signal in immunostaining:
Increase blocking time and concentration
Use more stringent washing protocols
Consider tissue-specific autofluorescence quenching for IF
Inconsistent molecular weight detection:
Tissue-specific optimization requirements:
Different fixation protocols may be needed for various tissues
Antigen retrieval conditions may require optimization
Antibody concentration should be titrated for each application
Antibody selection significantly influences experimental outcomes. For C1RL studies, consider:
Polyclonal vs. monoclonal: Current commercial C1RL antibodies are primarily polyclonal , offering broad epitope recognition but potential batch variation
Species reactivity: Verify cross-reactivity with your model system (human and mouse reactivity confirmed for some antibodies)
Storage and handling:
Store at recommended temperatures (-20°C for many C1RL antibodies)
Avoid repeated freeze-thaw cycles
Use storage buffers appropriate for the antibody (PBS or glycerol formulations)
Application-specific validation: An antibody performing well in Western blot may not be optimal for IHC or IF applications
Recent advances in computational antibody design offer opportunities for improving C1RL-targeted reagents:
Computational methods can predict antibody-antigen interactions, allowing researchers to:
Design antibodies with improved specificity for C1RL
Predict cross-reactivity with related proteins
Optimize antibody stability and expressibility
The AbDesign algorithm represents one such approach, using a three-stage process to optimize both antibody stability and binding energy . Such computational methods can help address challenges in antibody design, particularly for targeting nonideal features like those in C1RL.
For researchers working with existing antibodies, structural modeling of C1RL-antibody interactions can help predict epitope accessibility in different experimental conditions and applications.
Several cutting-edge technologies show promise for advancing C1RL research:
Single-cell proteogenomics: Integration of antibody-based detection with transcriptomic analysis at single-cell resolution
Spatial transcriptomics with protein co-detection: Analyzing C1RL protein expression in spatial context alongside gene expression data
Advanced microscopy techniques:
Super-resolution microscopy for detailed subcellular localization
Intravital imaging for studying C1RL dynamics in vivo
Antibody engineering approaches:
Nanobodies for improved tissue penetration
Bispecific antibodies for functional studies
Site-specific conjugation techniques for improved imaging probes
Current evidence suggests several promising avenues for C1RL research:
Cancer biomarker development: C1RL upregulation in glioblastoma, hepatocellular carcinoma, and renal cell cancer points to potential diagnostic and prognostic applications
Immunomodulation: As a complement-related protein, C1RL may influence tumor immune microenvironments
Therapeutic targeting: Understanding C1RL function could reveal new therapeutic opportunities, particularly in cancers where it serves as a prognostic marker
Complement system biology: Further characterization of C1RL's role may provide insights into novel complement activation pathways
Future research should focus on validating C1RL's functional significance through carefully designed in vitro and in vivo models, with particular attention to its role in modulating immune responses in disease contexts.