CPVL (carboxypeptidase, vitellogenic-like) is a serine carboxypeptidase initially identified in human macrophages, with roles in antigen processing, secretory pathways, and cellular structures like lamellipodia . CPVL antibodies are immunological tools designed to detect and study this protein in research and diagnostic applications. These antibodies enable insights into CPVL's expression, localization, and functional roles in both physiological and pathological contexts.
CPVL antibodies are validated for diverse experimental techniques. Key applications include:
Application | Dilution Range | Reactivity |
---|---|---|
Western Blot (WB) | 1:1,000–1:4,000 | Human, Mouse, Rat |
Immunohistochemistry (IHC) | 1:20–1:200 | Human tissues |
Immunofluorescence (IF/ICC) | 1:50–1:500 | HepG2 cells |
Flow Cytometry (FC) | 0.40 µg/10^6 cells | Human samples |
Immunoprecipitation (IP) | 0.5–4.0 µg/mg lysate | HEK-293 cells |
Source: Proteintech (12548-1-AP, 60429-5-PBS) , Abcam (ab204553)
Mechanism: CPVL promotes glioma cell survival by interacting with BTK and suppressing STAT1 phosphorylation via p300-mediated acetylation .
Clinical Relevance: High CPVL expression correlates with advanced tumor grade and poor prognosis in glioma patients .
Antibody Use: Western blot and IHC confirmed CPVL upregulation in glioma tissues vs. normal brain samples .
Mechanism: CPVL facilitates resistance by negatively regulating PTEN, impacting drug efficacy .
Antibody Use: Silencing CPVL via shRNA reduced tumor growth in xenograft models, validated using qRT-PCR and Western blot .
Antigen Processing: CPVL colocalizes with MHC class I/II in macrophage phagosomes, suggesting a role in peptide trimming for antigen presentation .
Proteintech 12548-1-AP: Rabbit IgG, reacts with human/mouse/rat, detects ~54 kDa band in WB .
Abcam ab204553: Rabbit IgG, validated for IHC and IF in human kidney and CACO-2 cells .
Proteintech 60429 Series: Mouse IgG1/IgG2a pairs (e.g., 60429-1-PBS + 60429-2-PBS) for cytometric bead arrays and ELISAs .
Cancer: CPVL is upregulated in glioma, breast cancer, and liver cancer, linked to therapy resistance and poor survival .
Inflammatory Diseases: CPVL levels correlate with inflammation severity, suggesting utility as a biomarker for infection outcomes .
Genetic Links: CPVL variants are associated with diabetic nephropathy and Wilms tumor .
Specificity: Antibodies are validated using knockout/knockdown controls (e.g., shRNA in glioma studies) .
Storage: Long-term stability at -20°C to -80°C in glycerol or PBS buffers .
CPVL antibodies are critical for ongoing research into:
CPVL (Carboxypeptidase, vitellogenic-like) is a serine carboxypeptidase that was first characterized in human macrophages. This protein appears to have multiple biological functions, including the digestion of phagocytosed particles in lysosomes, participation in inflammatory protease cascades, and trimming of peptides for antigen presentation . CPVL was initially identified in macrophages, and recent single-cell RNA-sequencing data confirms that macrophages in glioma tissues are a major source of CPVL expression . The protein contains a serine carboxypeptidase active site that is critical for its enzymatic functions, as demonstrated by mutagenesis studies of this region .
When conducting literature searches or database queries for CPVL, researchers should be aware of several alternative names and identifiers to ensure comprehensive results. These include: VLP, PSEC0124, UNQ197/PRO223, Probable serine carboxypeptidase CPVL, Vitellogenic carboxypeptidase-like protein, VCP-like protein, and hVLP . Using these alternative identifiers in database searches will help ensure that relevant research is not overlooked due to nomenclature variations across different studies and databases.
When selecting a CPVL antibody, researchers should consider several critical factors based on their specific experimental needs:
Application compatibility: Different antibodies are validated for specific applications. For instance, the rabbit recombinant monoclonal CPVL antibody (ab180147) is suitable for Western blot (WB) and immunohistochemistry on paraffin sections (IHC-P), while the rabbit polyclonal CPVL antibody (ab204553) is validated for IHC-P and immunocytochemistry/immunofluorescence (ICC/IF) .
Species reactivity: Confirm that the antibody reacts with your species of interest. Available CPVL antibodies have been validated for human samples, but cross-reactivity with other species may be predicted based on homology .
Clonality: Monoclonal antibodies like ab180147 offer higher specificity for a single epitope, while polyclonal antibodies like ab204553 recognize multiple epitopes and may provide stronger signals .
Immunogen information: The immunogen used to generate the antibody determines its binding region. For example, ab204553 was generated using a recombinant fragment within human CPVL amino acids 250-350 .
Validating antibody specificity is crucial for reliable experimental results. A multi-approach validation strategy for CPVL antibodies should include:
Western blotting: Verify that the antibody detects a band at the predicted molecular weight of CPVL (approximately 54 kDa) in positive control samples (e.g., HepG2 or 293 cell lysates) . The band pattern should match the expected expression profile of CPVL.
Knockdown/knockout controls: Use CPVL-silenced cells (via shRNA or CRISPR) to confirm signal reduction or elimination. The search results describe CPVL silencing using specific lentiviral shRNAs (shCPVL#1 and shCPVL#2) in U251 and LN382 cells, which could serve as negative controls .
Immunoprecipitation followed by mass spectrometry: This approach can confirm that the antibody is pulling down the correct protein and identify any cross-reactivities .
Immunostaining pattern analysis: Compare the staining pattern with published literature. For example, in immunohistochemical analysis, CPVL should be detected in human spleen tissue and kidney tissue with the expected cellular distribution .
Positive and negative tissue controls: Include tissues known to express or lack CPVL as controls in your experiments.
The choice between monoclonal and polyclonal CPVL antibodies has significant implications for research outcomes:
Recognize a single epitope, providing higher specificity
Offer consistent lot-to-lot reproducibility
Typically show less background staining
May be less sensitive to conformational changes in the protein
Ideal for applications requiring high specificity such as Western blotting and quantitative analyses
Recognize multiple epitopes, potentially providing stronger signals
May be more robust to protein denaturation or fixation
Can detect proteins with minor variations or modifications
Better suited for applications like immunoprecipitation and immunohistochemistry where signal amplification is beneficial
In practice, researchers should consider using both types for complementary approaches. For example, a monoclonal antibody might be preferred for precise localization studies, while a polyclonal antibody might be better for detecting low-abundance CPVL in fixed tissues.
For optimal immunohistochemistry (IHC) results with CPVL antibodies, researchers should follow these methodological guidelines:
For paraffin-embedded tissues, perform heat-mediated antigen retrieval with EDTA buffer at pH 9 before IHC staining
Complete deparaffinization and rehydration steps are essential for consistent results
Incubate at 4°C overnight or at room temperature for 1-2 hours
For ab180147: Use prediluted goat anti-rabbit IgG (HRP) as secondary antibody
Counterstain with hematoxylin for nuclear visualization
Include positive controls (human spleen or kidney) and negative controls (antibody diluent only)
Develop with appropriate substrate (DAB recommended)
Evaluate staining patterns in context of known CPVL expression
Analysis should focus on macrophage-rich regions, as CD68 (macrophage marker) co-staining has demonstrated that macrophages are a major source of CPVL expression in tissues like glioma .
Western blotting for CPVL requires specific optimization steps to achieve reliable results:
Extract proteins from relevant cell lines (HepG2 and 293 cells serve as positive controls)
Use approximately 20 μg of protein lysate per lane
Include protease inhibitors in lysis buffers to prevent CPVL degradation
Use standard SDS-PAGE conditions
Transfer to PVDF or nitrocellulose membranes using standard conditions
Block membranes with 5% non-fat milk or BSA in TBST
Incubate with primary antibody overnight at 4°C
Wash thoroughly with TBST buffer
Incubate with HRP-conjugated secondary antibody (e.g., Goat Anti-Rabbit IgG, (H+L), Peroxidase conjugated) at 1/1000 dilution
Develop using enhanced chemiluminescence (ECL)
Verify band size against molecular weight markers
Consider stripping and reprobing for loading controls (e.g., GAPDH, β-actin)
Multiple bands or unexpected molecular weights may indicate post-translational modifications or protein degradation that should be investigated further.
Successful immunofluorescence experiments with CPVL antibodies require attention to several technical aspects:
For cell lines: Fix with 4% paraformaldehyde and permeabilize with Triton X-100
For tissues: Consider using frozen sections to preserve epitopes that may be sensitive to paraffin embedding
Polyclonal antibody ab204553 has been validated for immunofluorescence at 4 μg/mL
For co-staining experiments with other mouse mAbs, consider using directly conjugated anti-CPVL antibodies (e.g., Alexa-Fluor 488 conjugated)
Include appropriate negative controls (secondary antibody only, isotype controls)
For subcellular localization studies, include organelle markers (e.g., lysosomal markers)
Counterstain nuclei with DAPI
For cytoskeletal studies, TRITC-phalloidin can be used to visualize actin filaments
Optimize laser power and detector settings to avoid saturation
Use sequential scanning to prevent fluorophore crosstalk
Consider acquiring Z-stacks for detailed localization analysis
CPVL has been observed in specific subcellular compartments, including possible association with membrane ruffles and actin structures, so high-resolution imaging is recommended for detailed localization studies .
Investigating CPVL's protein interactions requires sophisticated approaches combining antibody-based techniques with molecular methods:
Use anti-CPVL antibodies to precipitate CPVL and associated proteins from macrophage lysates
Follow with Western blotting for suspected interaction partners
Based on the research results, interaction with Bruton's tyrosine kinase (BTK) would be a primary target for investigation
Utilize primary antibodies against CPVL and potential interacting proteins
This technique allows visualization of protein interactions in situ with subcellular resolution
Particularly useful for confirming CPVL-BTK interactions in specific cellular compartments
Perform immunopurification with anti-CPVL antibodies followed by mass spectrometry analysis
This approach has successfully identified CPVL's interaction with BTK
Compare results between resting and activated macrophages to identify context-specific interactions
Use GST-tagged CPVL in pull-down assays to confirm direct interactions
This approach has been used to validate CPVL-BTK interactions
When designing these experiments, consider that CPVL interactions may be transient or context-dependent. Research has shown that CPVL physically interacts with BTK and influences STAT1 phosphorylation through promoting p300-mediated STAT1 acetylation .
Given CPVL's potential role in trimming peptides for antigen presentation , several methodologies can be employed to investigate this function:
Establish CPVL knockdown and overexpression in antigen-presenting cells (macrophages, dendritic cells)
Measure presentation efficiency using T-cell activation assays
Compare presentation of different antigens to identify specificity patterns
Isolate MHC-bound peptides from cells with modified CPVL expression
Analyze peptide repertoire using mass spectrometry
Compare N-terminal and C-terminal peptide characteristics to identify CPVL-specific processing signatures
Use anti-CPVL antibodies in combination with markers for antigen-processing compartments
Perform immuno-electron microscopy to precisely localize CPVL in relation to MHC loading compartments
Track antigen trafficking in the presence and absence of functional CPVL
Express and purify recombinant CPVL using bacterial expression systems similar to those described in the search results
Test its capacity to process synthetic peptides in vitro
Analyze cleavage products by mass spectrometry
These approaches should be complemented with functional immunological assays to connect biochemical findings with physiological outcomes in antigen presentation.
Recent research has identified CPVL as a regulator of STAT1 signaling in glioma, suggesting important methodological approaches for further investigation :
Use phospho-specific antibodies to detect STAT1 phosphorylation levels by Western blotting
Compare STAT1 phosphorylation in CPVL-silenced versus control cells
Include time-course experiments following stimulation with IFN-γ or other STAT1 activators
Investigate STAT1 acetylation using acetyl-specific antibodies
Perform immunoprecipitation with anti-STAT1 antibodies followed by Western blotting with anti-acetyl lysine antibodies
Compare acetylation levels between CPVL-expressing and CPVL-silenced cells
Use BTK inhibitors in combination with CPVL manipulation
Measure effects on downstream STAT1 target genes
Determine if BTK inhibition mimics CPVL silencing effects
Perform ChIP with anti-STAT1 antibodies in CPVL-manipulated cells
Assess STAT1 binding to promoters of known target genes
Correlate binding with transcriptional output using qRT-PCR
Utilize STAT1-responsive promoter constructs
Compare reporter activity in CPVL-manipulated versus control cells
Include rescue experiments with CPVL cDNA to confirm specificity
These methodologies will help delineate the mechanisms by which CPVL regulates STAT1 signaling and identify potential intervention points for therapeutic development.
Researchers may encounter several challenges when performing Western blots for CPVL detection:
Issue: No band detected
Potential solutions:
Verify sample preparation: CPVL is expressed in specific cell types; confirm expression in your samples
Increase protein loading: Try 20-30 μg total protein as used in published protocols
Optimize antibody concentration: Test a range around the recommended 1/1000 dilution
Extend exposure time: CPVL may be expressed at low levels in some samples
Try alternative lysis buffers: CPVL may require specific extraction conditions
Issue: Multiple bands or unexpected molecular weight
Potential solutions:
Verify antibody specificity: Compare with CPVL knockdown controls
Consider post-translational modifications: CPVL may be glycosylated or proteolytically processed
Optimize sample preparation: Include protease inhibitors to prevent degradation
Check running conditions: Ensure proper SDS-PAGE setup for the expected 54 kDa protein
Issue: High background
Potential solutions:
Increase blocking time/concentration: Use 5% BSA or milk in TBST
Dilute primary antibody further
Increase washing steps: More extensive washing with TBST
Use freshly prepared buffers
Consider alternative secondary antibodies with lower background
Issue: Inconsistent results between experiments
Potential solutions:
Standardize lysate preparation methods
Prepare larger batches of antibody dilutions to use across experiments
Control for protein loading using housekeeping proteins
Achieving specific CPVL staining in immunohistochemistry requires troubleshooting several common issues:
Issue: Non-specific background staining
Potential solutions:
Optimize antibody dilution: Try a range around the recommended 1/250 (monoclonal) or 1/1000 (polyclonal) dilutions
Enhance blocking: Use 3-5% BSA or serum matching the species of the secondary antibody
Add 0.1-0.3% Triton X-100 to reduce hydrophobic interactions
Include avidin/biotin blocking steps if using biotin-based detection systems
Consider using monoclonal antibodies which typically provide higher specificity
Issue: Weak or absent staining
Potential solutions:
Optimize antigen retrieval: Use EDTA buffer at pH 9 as specified for CPVL detection
Extend primary antibody incubation time to overnight at 4°C
Use signal amplification systems such as tyramide signal amplification
Ensure tissue fixation is optimal (overfixation can mask epitopes)
Verify CPVL expression in the selected tissue (human spleen is a good positive control)
Issue: Difficult to distinguish specific from non-specific staining
Potential solutions:
Include appropriate negative controls (isotype control or primary antibody omission)
Use CPVL-silenced tissues or cells as negative controls when possible
Perform dual staining with cell-type markers (e.g., CD68 for macrophages)
Compare staining patterns with published results
Consider using a different CPVL antibody that recognizes a different epitope
Issue: Inconsistent staining between samples
Potential solutions:
Standardize fixation and processing methods
Process and stain all samples in a single batch
Use automated staining platforms if available
Include internal control tissues on each slide
Determining the precise subcellular localization of CPVL can be challenging due to technical and biological factors:
Issue: Conflicting localization patterns between studies
Potential approaches:
Use multiple antibodies targeting different CPVL epitopes
Combine antibody detection with CPVL-EGFP fusion protein expression
Compare fixation methods (paraformaldehyde vs. methanol)
Use super-resolution microscopy for precise localization
Issue: Difficulty distinguishing specific compartments
Potential approaches:
Perform co-localization studies with established organelle markers for:
Lysosomes (LAMP1, LAMP2)
ER (Calnexin, PDI)
Golgi (GM130, TGN46)
Endosomes (EEA1, Rab proteins)
Use immuno-electron microscopy for ultrastructural localization
Perform subcellular fractionation followed by Western blotting
Consider live cell imaging with CPVL-EGFP to track dynamic localization
Issue: Changes in localization under different conditions
Potential approaches:
Systematically compare resting vs. activated macrophages
Examine localization after phagocytosis or inflammation
Study time-course of localization during cellular processes
Investigate if CPVL localization changes in disease states
Issue: Contradictions between imaging and biochemical evidence
Potential approaches:
Combine imaging with proximity labeling techniques (BioID, APEX)
Validate with orthogonal techniques (e.g., PLA, FRET)
Perform subcellular fractionation with high resolution (e.g., density gradient centrifugation)
Consider that multiple pools of CPVL may exist within different compartments
The search results indicate CPVL may be associated with actin structures in lamellipodia/membrane ruffles, suggesting dynamic localization patterns that require careful experimental design .
Recent research has revealed significant roles for CPVL in cancer, particularly in glioma progression:
CPVL is significantly upregulated in glioma tissues compared to normal brain tissues
CPVL silencing inhibits proliferation and promotes apoptosis of glioma cells in vitro
CPVL knockdown suppresses tumor growth in xenograft mouse models
Mechanistically, CPVL physically interacts with BTK and downregulates STAT1 phosphorylation
Cell line models:
Animal models:
Clinical samples:
These models should be used in combination with CPVL antibodies for mechanistic studies and therapeutic target validation.
Investigating CPVL's enzymatic activity requires specialized biochemical approaches:
Express CPVL without its 21 amino acid signal sequence using bacterial expression systems
PCR amplify CPVL using specific primers with appropriate restriction sites (e.g., NheI, HindIII)
Clone into expression vectors like pET-28a(+)
Transform competent bacterial cells (e.g., BL21-DE3) and induce with IPTG
Purify inclusion bodies by sonication and washing with detergents
Use synthetic peptide substrates with fluorogenic or chromogenic leaving groups
Test activity under various pH conditions (lysosomal enzymes typically function at acidic pH)
Include specific protease inhibitors to confirm serine carboxypeptidase activity
Compare wild-type CPVL with mutants of the serine carboxypeptidase active site (mutagenesis of the region between 100-400 bp has been used)
Perform proteomics-based substrate screens
Use peptide libraries to determine sequence preferences
Investigate physiologically relevant proteins based on CPVL's proposed functions in:
Antigen processing
Inflammatory cascades
Phagocytosed particle digestion
Compare substrate processing in cells with and without CPVL expression
Use mass spectrometry to identify cleaved products
Correlate enzymatic activity with biological outcomes (e.g., antigen presentation)
These approaches will help determine if CPVL functions as an active carboxypeptidase and identify its specific substrates and biological relevance.
The search results suggest CPVL may serve as a potential prognostic biomarker and therapeutic target, particularly in glioma . Investigating its therapeutic potential requires systematic approaches:
Perform CPVL knockdown in relevant disease models using:
Assess phenotypic outcomes in multiple model systems
Determine if CPVL inhibition reverses disease-associated phenotypes
Design small molecule inhibitors targeting the serine carboxypeptidase active site
Screen existing serine protease inhibitor libraries
Consider antibody-based therapeutic approaches
Explore targeted protein degradation strategies (PROTACs)
Test CPVL inhibition in combination with standard-of-care treatments
For glioma, combine with temozolomide or radiation therapy
Investigate synergy with BTK inhibitors, given the CPVL-BTK interaction
Target multiple nodes in the CPVL-BTK-STAT1 signaling axis
Correlate CPVL expression with patient outcomes in clinical datasets
Develop companion diagnostics using CPVL antibodies
Explore biomarkers that predict response to CPVL-targeted therapies
Design rational clinical trial strategies based on mechanistic insights
Determine effects of CPVL inhibition on normal macrophage functions
Assess potential immunological consequences given CPVL's role in antigen presentation
Evaluate off-target effects of CPVL inhibition strategies
These methodological approaches will help establish whether CPVL represents a viable therapeutic target and guide the development of targeting strategies.
Rigorous statistical analysis is essential for interpreting CPVL expression data from immunohistochemistry studies:
Scoring Method | Description | Advantages | Limitations |
---|---|---|---|
H-score | Combines intensity (0-3) and percentage of positive cells | Provides continuous variable | Subjective intensity assessment |
Allred score | Sum of proportion score (0-5) and intensity score (0-3) | Established in clinical practice | Less sensitive to small changes |
Digital image analysis | Automated quantification of staining intensity and area | Objective, reproducible | Requires standardized image acquisition |
Cell counting | Manual/automated counting of positive/negative cells | Direct measure of expression frequency | Time-consuming, sampling bias concerns |
Use non-parametric tests (Mann-Whitney, Kruskal-Wallis) for comparing CPVL expression between groups
For survival analysis, use Kaplan-Meier with log-rank test and Cox proportional hazards models
Include multivariate analysis to control for confounding factors
Calculate intra- and inter-observer variability for manual scoring methods
Determine appropriate sample sizes through power analysis
Document detailed protocols for tissue processing and staining
Include representative images of different staining intensities
Report both raw data and derived scores
Specify statistical software and version used
Include 95% confidence intervals in addition to p-values
These approaches will enhance the rigor and reproducibility of CPVL expression analysis in immunohistochemistry studies, a crucial aspect given CPVL's potential role as a prognostic biomarker in diseases like glioma .
Resolving contradictory findings about CPVL function requires systematic methodological approaches:
Investigate context-dependency:
Systematically compare experimental conditions (cell types, disease models)
Determine if differences in CPVL function relate to cellular context
Consider tissue-specific factors that may influence CPVL activity
Resolve technical differences:
Compare antibody specificity and epitope mapping
Evaluate knockdown/overexpression efficiency across studies
Standardize functional assays and readouts
Examine isoform-specific effects:
Integrate multi-omics data:
Combine transcriptomic, proteomic, and functional data
Look for consistent patterns across data types
Use systems biology approaches to model CPVL in biological networks
Explicitly acknowledge contradictory findings
Discuss methodological differences that might explain discrepancies
Present multiple interpretations of data when conclusive evidence is lacking
Propose specific experiments to resolve contradictions
Consider collaborative approaches with laboratories reporting different results
By systematically addressing contradictions, researchers can develop a more nuanced understanding of CPVL's biological functions and disease relevance.
Several cutting-edge technologies hold promise for elucidating CPVL's biological roles:
Single-cell RNA-sequencing to map CPVL expression patterns across cell types
Single-cell proteomics to correlate CPVL protein levels with other markers
Spatial transcriptomics to visualize CPVL expression in tissue context
These approaches can build upon initial findings from the Single cell Glioma RNA-sequence database that identified macrophages as a major source of CPVL
Super-resolution microscopy for precise subcellular localization
Correlative light and electron microscopy (CLEM) to connect functional data with ultrastructure
Expansion microscopy for enhanced visualization of CPVL in cellular compartments
Proximity labeling (BioID, APEX) to map CPVL's protein interaction network
Cross-linking mass spectrometry to identify direct binding partners
Hydrogen-deuterium exchange mass spectrometry to study CPVL structural dynamics
These methods can extend findings on CPVL's interaction with BTK and effects on STAT1
CRISPR activation/interference for precise CPVL expression modulation
CRISPR screens to identify genetic modifiers of CPVL function
Base editing to introduce specific mutations in the carboxypeptidase active site
Prime editing for precise genetic modifications of CPVL regulatory elements
Cryo-electron microscopy of CPVL complexes
X-ray crystallography of CPVL alone and with binding partners
AlphaFold and other AI-based structure prediction to guide functional studies
These emerging technologies will provide unprecedented insights into CPVL's molecular functions and regulatory mechanisms.
Despite recent advances, several fundamental questions about CPVL remain unresolved:
Substrate specificity:
What are the natural substrates of CPVL's carboxypeptidase activity?
How does substrate recognition occur?
Are there tissue-specific differences in CPVL's enzymatic targets?
Physiological functions:
What is CPVL's precise role in antigen presentation pathways?
How does CPVL contribute to normal macrophage functions?
What are the consequences of CPVL knockout in animal models?
Regulation mechanisms:
How is CPVL expression regulated at transcriptional and post-transcriptional levels?
What signals induce CPVL upregulation in cancer and inflammatory conditions?
Are there endogenous inhibitors of CPVL activity?
Pathological relevance:
Therapeutic applications:
Is CPVL a viable therapeutic target in glioma or other diseases?
What strategies could effectively inhibit CPVL function?
Are there potential side effects of CPVL inhibition on immune function?
Addressing these questions will require integrative approaches combining biochemical, cellular, and in vivo studies using the methodologies discussed throughout this document.