CSRP1 belongs to the CSRP family, characterized by cysteine-rich motifs that facilitate interactions with cytoskeletal proteins and signaling pathways. Its molecular weight is approximately 21 kDa, and it localizes to the cytoplasm, nucleus, and cell membrane . Key functions include:
Wnt/JNK Signaling: CSRP1 interacts with Dishevelled 2 (Dvl2) and Diversin (Div), modulating noncanonical Wnt pathways and cytoskeletal remodeling .
Cell Migration: It regulates mesoderm and cardiac progenitor cell migration during zebrafish development, with knockdown causing cardiac bifida .
Immune Regulation: CSRP1 influences immune cell infiltration and checkpoint expression in acute myeloid leukemia (AML) .
The antibody is widely used in:
Detects CSRP1 in human tissues (e.g., colon, lung, prostate) and xenograft models .
Requires antigen retrieval with TE buffer (pH 9.0) or citrate buffer (pH 6.0) .
CSRP1 regulates convergent extension movements and cardiac mesoderm migration. Knockdown leads to midline defects and cardiac bifida, highlighting its role in organogenesis .
Overexpressed in AML samples compared to normal bone marrow .
High expression correlates with poor prognosis and immune checkpoint activation (e.g., PD-1/PD-L1) .
Low CSRP1 expression predicts rapid progression to castration-resistant prostate cancer (CRPC) .
Inhibits tumor growth and enhances androgen dependence in xenograft models .
CSRP1 shows ubiquitous cytoplasmic expression across tissues, including the prostate, colon, and pancreas .
| Tissue | Expression Pattern |
|---|---|
| Prostate | Cytoplasmic, membranous |
| Colon | Strong cytoplasmic staining |
| Pancreas | Membranous and cytoplasmic |
| Adipose Tissue | Weak cytoplasmic signal |
CSRP1 (also known as CRP, CRP1, CSRP, or CYRP) is a member of the cysteine and glycine-rich protein family that plays crucial roles in cell adhesion and cytoskeletal organization . Research suggests it significantly contributes to cell morphology and migration processes, making it essential for tissue development and wound healing . Additionally, CSRP1 may have important functions in neuronal development . The protein contains specific structural domains that facilitate its interaction with other cellular components, particularly those involved in cytoskeletal dynamics. Its regulatory functions appear to be context-dependent, varying across different tissue types and developmental stages.
CSRP1 antibodies are typically generated by immunizing host animals (commonly rabbits) with either recombinant fusion proteins containing specific amino acid sequences of human CSRP1 or with synthesized peptides derived from CSRP1 protein . The most common types available include:
Polyclonal antibodies: Generated in rabbits against various epitopes of CSRP1, these antibodies recognize multiple regions of the target protein. For example, the CAB1071 antibody is produced using a recombinant fusion protein corresponding to amino acids 1-193 of human CSRP1 (NP_004069.1) .
Monoclonal antibodies: Produced from single B-cell clones, providing higher specificity for particular epitopes. Some monoclonal antibodies like clone 2A11 are available for CSRP1 detection .
Antibodies targeting different regions of the CSRP1 protein are available, including those recognizing the internal region (AA 51-100, AA 71-120) and broader regions (AA 1-193, AA 94-192) . These variations allow researchers to select antibodies appropriate for specific experimental requirements and applications.
To ensure experimental rigor, CSRP1 antibody specificity should be validated through multiple complementary approaches:
Western blotting with positive control samples (such as A-549, BT-474, HeLa cells for human samples, or appropriate mouse tissues) . Compare the observed molecular weight with the expected size of CSRP1 (~21 kDa).
Immunoprecipitation followed by mass spectrometry to confirm pull-down of authentic CSRP1 protein.
RNA interference or CRISPR knockout validation, where CSRP1 expression is reduced/eliminated and corresponding reduction in antibody signal is observed.
Cross-reactivity testing against related proteins, particularly other CSRP family members like CSRP2 and CSRP3, to ensure specificity.
Immunohistochemistry with appropriate positive and negative control tissues, comparing staining patterns with known CSRP1 expression profiles.
Peptide competition assays, where pre-incubation with the immunizing peptide should abolish specific antibody binding.
These validation steps are critical for preventing experimental artifacts and ensuring reliable research outcomes.
For optimal Western blotting results with CSRP1 antibodies, researchers should consider the following protocol:
Sample preparation:
Protein separation:
Use 12-15% SDS-PAGE gels (optimal for visualizing CSRP1's ~21 kDa band)
Run at 100-120V for adequate separation
Transfer conditions:
Transfer to PVDF membranes (preferred over nitrocellulose for this protein)
Use wet transfer at 100V for 1 hour or 30V overnight at 4°C
Blocking and antibody incubation:
Block with 5% non-fat milk in TBST for 1 hour at room temperature
Dilute primary CSRP1 antibody at 1:500 to 1:2000 in blocking solution
Incubate overnight at 4°C with gentle rocking
Wash 3-5 times with TBST (5 minutes each)
Incubate with HRP-conjugated secondary antibody (1:5000-1:10000) for 1 hour at room temperature
Wash 3-5 times with TBST
Detection:
Use enhanced chemiluminescence detection
Typical exposure times range from 30 seconds to 5 minutes
The antibody should detect a single band at approximately 21 kDa, corresponding to CSRP1 protein.
Optimization of CSRP1 antibodies for IHC requires careful consideration of several parameters:
Tissue preparation:
Formalin-fixed paraffin-embedded (FFPE) sections (4-6 μm thickness)
Freshly cut sections yield better results than stored slides
Antigen retrieval methods:
Heat-induced epitope retrieval in citrate buffer (pH 6.0) for 20 minutes
Allow cooling to room temperature (approximately 20 minutes)
Blocking steps:
Block endogenous peroxidase with 3% H₂O₂ for 10 minutes
Block non-specific binding with 5-10% normal serum from secondary antibody host species
Consider additional blocking if background is observed (e.g., avidin/biotin blocking)
Antibody dilution and incubation:
Test a range of antibody dilutions (start with manufacturer recommendations)
Incubate primary antibody overnight at 4°C in a humidified chamber
Use appropriate detection systems (e.g., polymer-based or ABC method)
Controls:
Always include positive control tissues with known CSRP1 expression
Include negative controls (omitting primary antibody)
Consider using tissues from CSRP1 knockout models as specificity controls
Counterstaining and mounting:
Light hematoxylin counterstaining provides optimal nuclear detail without obscuring CSRP1 staining
Use permanent mounting media for long-term storage
With proper optimization, CSRP1 antibodies can provide specific staining in various tissue types, enabling both research and potential diagnostic applications.
Several complementary approaches can be employed to investigate CSRP1 protein-protein interactions:
Co-immunoprecipitation (Co-IP):
RNA immunoprecipitation (RIP):
Proximity ligation assay (PLA):
Visualize protein-protein interactions in situ
Requires CSRP1 antibody raised in a different host species than antibodies against potential interacting partners
Provides spatial information about interaction sites within cells
Protein-protein interaction network analysis:
Yeast two-hybrid or mammalian two-hybrid screening:
For discovery of novel CSRP1 interacting partners
Requires construction of CSRP1 fusion proteins as "bait"
These methods, used individually or in combination, can provide comprehensive insights into the interactome of CSRP1 and its functional relevance in different cellular contexts.
Research has established significant correlations between CSRP1 expression and clinical outcomes in AML patients:
Survival impact: High CSRP1 expression is associated with significantly worse prognosis in AML patients, with a hazard ratio of 2.36 (95% CI: 1.53-3.64; P < 0.001) in the TCGA-LAML dataset. This finding has been validated across multiple independent cohorts, including the Beat-AML dataset, ZZU cohort, and GSE12417 dataset .
Multivariate analysis: CSRP1 remains an independent prognostic factor after adjusting for established risk factors, with an adjusted HR of 2.048 (95% CI: 1.313-3.193; P = 0.002) .
Clinical characteristics: The table below summarizes key clinical differences between high and low CSRP1 expression groups in AML:
| Characteristic | Low CSRP1 group | High CSRP1 group | P-value |
|---|---|---|---|
| n | 109 | 115 | - |
| Age, mean ± SD | 44.81 ± 16.56 | 44.74 ± 15.61 | 0.975 |
| WBC, median (IQR) | 25.05 (7.6, 62.18) | 23.7 (6.19, 67.19) | 0.625 |
| PB-blasts, median (IQR) | 38.5 (0.84, 80) | 49.5 (8, 85) | 0.089 |
| BM-blasts, median (IQR) | 46.6 (0.83, 72.3) | 59.6 (24.4, 84.6) | 0.021 |
| Risk, n (%) | 0.651 | ||
| Favorable | 24 (22%) | 23 (20%) | |
| Intermediate | 47 (43.1%) | 45 (39.1%) | |
| Adverse | 38 (34.9%) | 47 (40.9%) |
Molecular associations: High CSRP1 expression correlates with increased frequency of DNMT3A mutations .
Predictive value: Time-dependent ROC curve analysis from the TCGA-LAML dataset demonstrates excellent predictive performance of CSRP1 for AML patient survival .
These findings suggest that CSRP1 expression analysis, facilitated by antibody-based detection methods, may have utility in AML risk stratification and treatment decision-making.
Several potential molecular mechanisms may explain CSRP1's role in cancer progression:
Cytoskeletal regulation and cell migration:
Signaling pathway modulation:
Drug sensitivity correlations:
Epigenetic regulation:
Tissue-specific effects:
Understanding these mechanisms requires sophisticated experimental approaches using CSRP1 antibodies for protein detection, localization, and interaction studies. Further research may reveal novel therapeutic opportunities targeting CSRP1 or its associated pathways.
CSRP1 antibodies can be strategically employed to investigate differential expression across cancer types through several methodological approaches:
Tissue microarray (TMA) analysis:
Utilize CSRP1 antibodies for immunohistochemical staining of TMAs containing multiple cancer types
Quantify staining intensity and proportion of positive cells using digital pathology platforms
Correlate expression patterns with clinicopathological features and survival data
This approach could validate the RNA-seq findings showing CSRP1 overexpression in 7 cancers and underexpression in 18 others
Western blot comparative analysis:
Flow cytometry for cancer cell population analysis:
Develop protocols for intracellular staining with CSRP1 antibodies
Analyze heterogeneity within tumor samples at the single-cell level
Combine with other markers to identify specific cellular subsets with altered CSRP1 expression
Subcellular localization studies:
Use immunofluorescence with CSRP1 antibodies to determine whether subcellular localization differs across cancer types
Co-stain with markers for specific cellular compartments to assess potential functional differences
Comparative phosphoproteomic analysis:
Develop or obtain phospho-specific CSRP1 antibodies
Investigate whether post-translational modifications of CSRP1 differ between cancer types
Correlate modification patterns with activation of specific signaling pathways
These approaches allow researchers to move beyond transcriptomic data to validate protein-level differences, potentially revealing new diagnostic markers or therapeutic targets across cancer types.
Researchers frequently encounter several challenges when working with CSRP1 antibodies that can be systematically addressed:
Non-specific binding and background:
Problem: High background or multiple bands in Western blots
Solutions:
Increase blocking time/concentration (try 5% BSA instead of milk for phospho-epitopes)
Optimize antibody dilution (test range from 1:500 to 1:2000)
Increase washing duration and frequency
Pre-adsorb antibody with non-specific proteins
Use more stringent washing buffers (increase Tween-20 concentration slightly)
Inconsistent immunoprecipitation results:
Problem: Poor or variable pull-down efficiency
Solutions:
Poor signal in immunohistochemistry:
Problem: Weak or absent staining
Solutions:
Test multiple antigen retrieval methods (citrate vs. EDTA buffers)
Extend primary antibody incubation time
Use amplification systems (e.g., tyramide signal amplification)
Ensure tissue fixation is optimal (overfixation can mask epitopes)
Try different CSRP1 antibodies targeting different epitopes
Cross-reactivity with CSRP family members:
Problem: Antibody may detect related proteins (CSRP2, CSRP3)
Solutions:
Verify antibody specificity using recombinant CSRP proteins
Use knockout/knockdown controls
Select antibodies targeting unique regions of CSRP1
Confirm results with orthogonal methods
Batch-to-batch variability:
Problem: Inconsistent results between antibody lots
Solutions:
Purchase larger quantities of single lots for long-term projects
Validate each new lot against previous lots
Maintain reference standards and positive controls
Consider monoclonal antibodies for greater consistency
Applying these systematic troubleshooting approaches can significantly improve experimental outcomes with CSRP1 antibodies.
Optimization strategies for CSRP1 antibody protocols should be tailored to specific sample types:
Cell lines:
Lysis buffer: Standard RIPA buffer works well for most applications
Protein amount: 20-30μg for Western blot
Fixation for ICC: 4% paraformaldehyde (10 minutes) preserves cytoskeletal elements
Antibody dilution: Start with 1:1000 for Western blots and 1:200 for ICC
Positive controls: A-549, BT-474, and HeLa cells show reliable CSRP1 expression
Primary tissue samples:
Lysis buffer: Consider gentler options like NP-40 buffer to preserve protein complexes
Homogenization: Optimize mechanical disruption to maintain protein integrity
Protein amount: Increase to 40-50μg for Western blots due to cellular heterogeneity
Fixation for IHC: 10% neutral buffered formalin (24-48 hours) followed by careful paraffin embedding
Antibody dilution: May require higher concentrations (1:100-1:500)
Controls: Include adjacent normal tissue when available
Bone marrow samples (relevant for AML research):
Sample preparation: Red blood cell lysis before protein extraction
Protein isolation: Consider nuclear/cytoplasmic fractionation
Antibody concentration: May need higher concentrations (1:200-1:500)
Background reduction: Extended blocking (2 hours) to minimize non-specific binding
Controls: Compare with normal bone marrow samples
Applications: Flow cytometry protocols can be developed for analyzing CSRP1 in specific cell populations
Liquid biopsies (serum/plasma):
Pre-clearing: Remove abundant proteins (albumin, immunoglobulins) before analysis
Concentration: May require sample concentration techniques
Detection methods: Consider using more sensitive detection methods (e.g., chemiluminescent substrates with extended exposure)
Validation: Always confirm with cellular/tissue samples when possible
Mouse models:
Cross-reactivity: Confirm antibody reactivity with mouse CSRP1
Tissue processing: Optimize fixation time based on specific tissues
Background reduction: Use mouse-on-mouse blocking kits if using mouse monoclonal antibodies
Controls: Include tissues from CSRP1 knockout mice when available
Each sample type presents unique challenges requiring systematic optimization to achieve reliable and reproducible results with CSRP1 antibodies.
Several innovative applications of CSRP1 antibodies could significantly advance our understanding of disease mechanisms:
Single-cell protein analysis:
Apply CSRP1 antibodies in mass cytometry (CyTOF) or imaging mass cytometry
Investigate heterogeneity of CSRP1 expression at single-cell resolution within tumors
Correlate with other markers to identify specific cellular subpopulations with unique CSRP1 expression patterns
This could reveal previously unrecognized cellular states in diseases like AML
Proximity-dependent labeling:
Use CSRP1 antibodies conjugated to enzymes like APEX2 or BioID
Map the proximal proteome around CSRP1 in different cellular contexts
Identify context-specific interaction networks that could explain differential functions across tissue types
This approach could elucidate why CSRP1 shows opposing expression patterns across cancer types
Spatial transcriptomics combined with protein detection:
Integrate in situ hybridization with immunofluorescence using CSRP1 antibodies
Correlate protein expression with transcriptional profiles in the spatial context of tissues
Map CSRP1 expression to specific tissue microenvironments
This could reveal niche-dependent regulation of CSRP1 in development and disease
Antibody-based therapeutic targeting:
Develop conjugated antibodies targeting CSRP1 for potential therapeutic applications
Explore selective delivery of cytotoxic agents to CSRP1-overexpressing AML cells
Investigate whether CSRP1-targeting can sensitize cells to conventional therapies
This translational approach could leverage the finding that high CSRP1 expression predicts poor outcomes in AML
Mechanisms of drug sensitivity modulation:
Use CSRP1 antibodies to track changes in expression, localization, and interactions following drug treatment
Investigate the molecular basis for differential drug sensitivity (e.g., higher sensitivity to 5-fluorouracil but lower sensitivity to fludarabine)
Develop predictive models for personalized therapeutic approaches based on CSRP1 status
These novel applications could transform our understanding of CSRP1's role in disease and potentially lead to new diagnostic and therapeutic strategies.
The integration of CSRP1 antibodies into clinical diagnostics represents a promising frontier for improving cancer patient care:
Prognostic and predictive biomarker development:
Standardize immunohistochemical protocols for CSRP1 detection in diagnostic pathology
Develop scoring systems correlating CSRP1 expression levels with clinical outcomes
Create predictive algorithms incorporating CSRP1 status alongside established risk factors
This could enhance current risk stratification systems for AML, which research shows "remain insufficient to select the best schedules"
Companion diagnostics for targeted therapies:
Develop CSRP1 antibody-based assays to identify patients likely to respond to specific treatments
Leverage findings that high CSRP1 expression correlates with differential drug sensitivities
Create standardized testing platforms suitable for clinical laboratory implementation
Validate cutoff values for treatment decisions in prospective clinical trials
Minimal residual disease (MRD) monitoring:
Develop highly sensitive flow cytometry protocols using CSRP1 antibodies
Create multiplexed panels incorporating CSRP1 alongside established MRD markers
Evaluate whether CSRP1 detection can improve MRD sensitivity in AML
This application could be particularly valuable given CSRP1's association with poor prognosis
Liquid biopsy applications:
Develop techniques to detect CSRP1 protein in circulating tumor cells or extracellular vesicles
Create sensitive immunoassays for CSRP1 detection in blood samples
Correlate circulating CSRP1 levels with disease status and treatment response
This non-invasive approach could facilitate longitudinal monitoring
Multiparameter diagnostic platforms:
Integrate CSRP1 antibodies into multiplexed immunofluorescence panels
Combine with other prognostic markers for comprehensive tumor profiling
Develop image analysis algorithms to quantify CSRP1 expression patterns
This approach could capture the complex biological context in which CSRP1 functions
Implementation of these approaches would require rigorous clinical validation, standardization across laboratories, and demonstration of clinical utility beyond existing diagnostic methods.
Computational approaches can significantly enhance CSRP1 antibody-based research, particularly in advancing precision medicine:
Automated image analysis for immunohistochemistry:
Develop deep learning algorithms for quantitative assessment of CSRP1 staining
Enable standardized scoring across laboratories
Identify subtle staining patterns not apparent to human observers
Integrate with digital pathology platforms for clinical implementation
This approach could standardize the prognostic value of CSRP1 in AML and other cancers
Multi-omics data integration:
Correlate CSRP1 protein levels (detected by antibodies) with transcriptomic, genomic, and epigenomic data
Build predictive models incorporating CSRP1 status with other molecular features
Identify patient subgroups with distinct biological and clinical characteristics
Similar to methods used in the study that identified correlations between CSRP1 and DNMT3A mutations
Network-based analyses:
Expand protein-protein interaction networks identified through CSRP1 antibody-based methods
Apply graph theory algorithms to identify critical nodes and potential therapeutic targets
Integrate with knowledge bases of biological pathways
This could build upon the PPI network analysis that identified 19 genes in the most significant module associated with CSRP1 in AML
Drug sensitivity prediction:
Develop machine learning models using CSRP1 antibody-based assay results to predict therapeutic responses
Implement algorithms similar to the OncoPredict algorithm used to explore correlations between CSRP1 and drug resistance
Create decision support tools for clinicians selecting treatment regimens
This approach could leverage findings that high CSRP1 expression correlates with differential drug sensitivities
Virtual screening for targeted therapeutics:
Use structural data and protein interaction networks identified through antibody-based methods
Apply molecular docking simulations to identify compounds targeting CSRP1 or its interaction partners
Prioritize candidate compounds for experimental validation
This approach could advance the suggestion that "CSRP1 may serve as a potential therapeutic target for AML in the future"