The DPYSL2 antibody is a research tool targeting dihydropyrimidinase-like 2 (DPYSL2), also known as collapsin response mediator protein 2 (CRMP2). DPYSL2 is a microtubule-associated protein critical for cytoskeletal dynamics, neurodevelopment, and cancer progression . It regulates axon guidance, cell migration, and signaling pathways such as mTOR and STAT3 . Antibodies against DPYSL2 enable detection, quantification, and functional studies of this protein in diverse biological contexts.
DPYSL2 antibodies are widely used in:
CRISPR/Cas9 knockout validation in cancer and neuronal models
Studying interactions with proteins like JAK1, STAT3, and PKM2 in cancer metastasis
Schizophrenia: Knockout of DPYSL2-B in human iPSC-derived neurons reduced dendritic length by 58% and disrupted mTOR signaling, mirroring transcriptomic changes seen in schizophrenia .
Alzheimer’s Disease: DPYSL2 dysfunction correlates with microtubule instability, a hallmark of neurodegenerative diseases .
Post-Translational Modifications: DPYSL2 phosphorylation at Ser522 regulates its role in cytoskeletal remodeling .
Pathway Modulation:
Therapeutic Targeting: Anti-DPYSL2 strategies (e.g., nanobodies) show promise in glioblastoma and metastatic cancers .
Biomarker Potential: High DPYSL2 expression correlates with poor survival in bladder and breast cancers .
DPYSL2 antibodies remain indispensable for unraveling the protein’s dual roles in neurodevelopment and oncogenesis, offering pathways for therapeutic intervention.
CRMP2 has been implicated in a range of cellular processes, including:
DPYSL2 (dihydropyrimidinase-like 2) is a cytoplasmic protein of 572 amino acid residues with a mass of 62.3 kDa. It belongs to the Hydantoinase/dihydropyrimidinase protein family and plays critical roles in neuronal development and polarity, axon growth and guidance, neuronal growth cone collapse, and cell migration. DPYSL2 is also known by several synonyms including CRMP2, DHPRP2, DRP-2, DRP2, N2A3, ULIP-2, ULIP2, and CRMP-2. Its ubiquitous expression across many tissue types and involvement in multiple cellular processes make it an important target for research in both normal development and disease states .
The protein has gained significant attention due to its emerging roles in various cancer types, including acute myeloid leukemia and bladder cancer, where it has been identified as a potential therapeutic target and prognostic marker .
DPYSL2 antibodies are utilized in multiple experimental applications, with the following being most commonly employed:
Western Blot (WB): The most widely used application for detecting DPYSL2 protein expression levels and post-translational modifications
Enzyme-Linked Immunosorbent Assay (ELISA): For quantitative measurement of DPYSL2 in samples
Immunofluorescence (IF): For visualizing subcellular localization and co-localization studies
Immunohistochemistry (IHC): For examining expression patterns in tissue samples
Immunoprecipitation (IP): For studying protein-protein interactions involving DPYSL2
These techniques have been referenced in over 170 scientific publications, demonstrating their reliability and widespread acceptance in the research community .
When selecting a DPYSL2 antibody for research, consider the following criteria:
Application compatibility: Verify that the antibody has been validated for your specific application (WB, IHC, IF, etc.)
Species reactivity: Ensure cross-reactivity with your experimental model organism (human, mouse, rat, etc.)
Epitope recognition: Select antibodies targeting different epitopes based on:
Full-length protein recognition for expression studies
Isoform-specific epitopes if studying particular variants
Phospho-specific antibodies for studying post-translational modifications
Clonality: Consider monoclonal antibodies for high specificity or polyclonal antibodies for stronger signals
Validation data: Review published literature and manufacturer validation data showing antibody performance
For challenging applications, consider using multiple antibodies recognizing different epitopes to confirm findings and reduce the risk of non-specific binding .
A robust experimental design using DPYSL2 antibodies should include the following controls:
Positive controls:
Cell lines or tissues with confirmed DPYSL2 expression (neuronal tissues show high expression)
Recombinant DPYSL2 protein for Western blot
Transfected cells overexpressing DPYSL2
Negative controls:
Specificity controls:
Blocking peptide competition assays
Multiple antibodies targeting different epitopes
Knockout/knockdown validation
These controls help distinguish true signals from background noise and confirm antibody specificity, enhancing the reliability of experimental results .
Optimal sample preparation varies by application:
Extract proteins using RIPA buffer supplemented with protease and phosphatase inhibitors
Include 1% NP-40 or Triton X-100 to enhance solubilization
Sonicate briefly to break down DNA
Heat samples at 95°C for 5 minutes in Laemmli buffer
Load 20-50 μg total protein per lane
Fix tissues in 4% paraformaldehyde for 24 hours
Use citrate buffer (pH 6.0) for antigen retrieval
Block with 5% normal serum from the same species as the secondary antibody
Optimize primary antibody concentration (typically 1:100 to 1:500 dilution)
Include antigen retrieval steps to expose epitopes
Fix cells in 4% paraformaldehyde for 15 minutes at room temperature
Permeabilize with 0.2% Triton X-100 for 10 minutes
Block with 3% BSA for at least 1 hour
Incubate with primary antibody overnight at 4°C
Use fluorophore-conjugated secondary antibodies with minimal spectral overlap if performing co-localization studies
| Issue | Possible Causes | Troubleshooting Approaches |
|---|---|---|
| No signal in Western blot | Insufficient protein, antibody concentration too low, poor transfer | Increase protein loading (50-100 μg), optimize antibody concentration, verify transfer with Ponceau S |
| High background | Non-specific binding, insufficient blocking, excessive antibody | Increase blocking time/concentration, reduce primary antibody concentration, add 0.1% Tween-20 to wash buffers |
| Multiple bands | Cross-reactivity, protein degradation, post-translational modifications | Use freshly prepared samples with protease inhibitors, reduce exposure time, try different antibody clones |
| Inconsistent staining in IHC | Fixation issues, antigen masking, uneven antibody distribution | Optimize fixation protocol, enhance antigen retrieval, ensure even antibody coverage |
| Poor reproducibility | Batch variation in antibodies, inconsistent sample preparation | Use the same antibody lot, standardize sample preparation, include internal controls |
For optimal results, validate new antibody lots by comparing them with previously validated antibodies using the same experimental conditions .
Multiple reference datasets provide valuable information on DPYSL2 expression patterns:
Tissue expression patterns: DPYSL2 is ubiquitously expressed across many tissue types with particularly high expression in neuronal tissues.
Cell line reference data: Various cell line databases document DPYSL2 expression, which can be useful for selecting appropriate positive control cell lines for antibody validation.
Cancer expression databases: Public databases such as GEPIA show that AML patients have higher DPYSL2 mRNA levels than normal samples, providing valuable comparison points for researchers studying DPYSL2 in cancer contexts .
Normal vs. pathological expression: Studies have demonstrated that DPYSL2 expression is upregulated in bladder cancer tissue compared with adjacent normal bladder tissue, and expression correlates with cancer staging .
Subcellular localization: The canonical localization of DPYSL2 is cytoplasmic, though changes in localization can occur under specific cellular conditions .
These reference datasets provide crucial benchmarks for researcher validation and experimental design.
DPYSL2 has emerging roles in cancer biology with significant implications for disease progression:
DPYSL2 antibodies facilitate these discoveries through:
Expression profiling using IHC and Western blot to correlate DPYSL2 levels with clinical outcomes
Co-immunoprecipitation to identify protein interaction partners like PKM2
Immunofluorescence to track subcellular localization changes during disease progression
Phospho-specific antibodies to monitor activation of signaling pathways
These applications have helped establish DPYSL2 as a potential therapeutic target for cancer treatment .
DPYSL2 has been identified as a potential mediator of drug resistance, particularly in the context of homoharringtonine (HHT) resistance in AML. Researchers can effectively use DPYSL2 antibodies to study drug resistance through several approaches:
Expression monitoring: Use Western blot to track DPYSL2 protein levels before and after drug treatment. Studies have shown that DPYSL2 protein levels decreased in three AML cell lines following HHT treatment .
Target validation studies: DPYSL2 was identified as a differentially expressed gene between HHT-resistant and HHT-sensitive cells by RNA-seq. Antibodies can confirm these findings at the protein level .
Mechanism exploration: Auto-docking studies revealed that HHT can target DPYSL2, forming a hydrogen bond with the ARG-173 residue with a binding energy of -3.83. Antibodies specific to this region can help validate structural interactions .
Treatment response monitoring: Dynamic expression analysis using DPYSL2 antibodies can track protein level changes during treatment. mRNA expression analyses have shown that DPYSL2 levels were reduced after complete remission in AML patients with high DPYSL2 expression at diagnosis .
In vivo validation: Antibodies can be used to confirm knockdown efficiency in xenograft models studying the effects of DPYSL2 inhibition on drug sensitivity and tumor growth .
These approaches help elucidate the molecular mechanisms of drug resistance and identify strategies to overcome therapeutic challenges.
To study DPYSL2 protein-protein interactions, researchers should consider the following approaches:
Co-immunoprecipitation (Co-IP):
Lyse cells in a gentle buffer (e.g., 25 mM Tris-HCl pH 7.4, 150 mM NaCl, 1% NP-40, 1 mM EDTA, protease inhibitors)
Pre-clear lysate with protein A/G beads
Incubate with DPYSL2 antibody (5-10 μg per mg of protein) overnight at 4°C
Add protein A/G beads for 2-3 hours
Wash 4-5 times with buffer containing reduced detergent
Elute with Laemmli buffer and analyze by Western blot using antibodies against suspected interaction partners
Proximity Ligation Assay (PLA):
Fix cells with 4% paraformaldehyde
Permeabilize with 0.2% Triton X-100
Incubate with primary antibodies against DPYSL2 and the target protein (from different species)
Apply species-specific PLA probes
Perform ligation and amplification according to manufacturer's protocol
Visualize interaction signals by fluorescence microscopy
Bimolecular Fluorescence Complementation (BiFC):
Clone DPYSL2 and potential interacting protein with split fluorescent protein fragments
Transfect constructs into appropriate cell lines
Analyze reconstituted fluorescence indicating protein-protein interaction
These methods have successfully identified that DPYSL2 interacts with pyruvate kinase M2 (PKM2) in bladder cancer cells, promoting the conversion of PKM2 tetramers to PKM2 dimers, which enhances aerobic glycolysis and epithelial-mesenchymal transition .
DPYSL2 undergoes several post-translational modifications (PTMs) that significantly impact its function in various cellular processes. Studying these modifications requires specialized approaches:
Phosphorylation sites:
Multiple kinases phosphorylate DPYSL2, affecting its roles in cytoskeletal dynamics and signaling
Use phospho-specific antibodies to detect site-specific phosphorylation
Employ phos-tag gels to separate phosphorylated from non-phosphorylated forms
Confirm with mass spectrometry for precise mapping of phosphorylation sites
SUMOylation and ubiquitination:
Affect DPYSL2 stability and subcellular localization
Use co-IP with SUMO or ubiquitin antibodies to detect modified forms
Employ proteasome inhibitors to stabilize ubiquitinated forms
Analyze by Western blot with DPYSL2 antibodies to detect higher molecular weight bands
Glycosylation:
Can impact DPYSL2 folding and function
Treat samples with glycosidases and analyze mobility shifts by Western blot
Use lectin-based enrichment followed by DPYSL2 antibody detection
Methodological approach:
Compare PTM patterns between normal and disease states using phospho-specific antibodies
Use kinase inhibitors to manipulate phosphorylation status
Create site-specific mutants to study functional consequences of specific modifications
Employ mass spectrometry for comprehensive PTM profiling
Understanding these modifications is critical as they may play roles in DPYSL2's involvement in disease processes, including its function in the JAK2/STAT3/STAT5-PI3K P85/AKT/GSK3b signaling axis in AML .
Recent research has revealed DPYSL2's unexpected role in cellular metabolism, particularly in cancer. To investigate this function, the following approaches are recommended:
Glucose uptake assays:
Lactate production measurement:
Metabolic enzyme activity assays:
Seahorse metabolic analysis:
Measure oxygen consumption rate (OCR) and extracellular acidification rate (ECAR)
Compare glycolytic capacity and mitochondrial function between control and DPYSL2-modified cells
Metabolomics profiling:
Use mass spectrometry to generate comprehensive metabolite profiles
Compare metabolite levels between control and DPYSL2-knockdown cells
In vivo validation:
Use PET imaging with 18F-FDG to assess glucose uptake in xenograft tumors with modified DPYSL2 expression
These approaches have helped establish that DPYSL2 promotes aerobic glycolysis in cancer cells through its interaction with PKM2, contributing to the Warburg effect that supports cancer progression .
DPYSL2 has been implicated in promoting epithelial-mesenchymal transition (EMT) in cancer cells. To investigate this role, researchers should consider these methodological approaches:
EMT marker analysis:
Use Western blot with antibodies against epithelial markers (E-cadherin, ZO-1) and mesenchymal markers (N-cadherin, Vimentin, Snail, Slug, Twist)
Compare marker expression in cells with normal vs. altered DPYSL2 levels
Perform immunofluorescence to visualize changes in marker localization and expression
Quantitative analysis of these markers can reveal the extent of EMT induction or suppression
Cell migration and invasion assays:
Conduct wound healing/scratch assays to assess migration capacity
Use Transwell chambers with or without Matrigel coating to evaluate invasion potential
Compare migration/invasion between control and DPYSL2-knockdown cells
Time-lapse microscopy can provide dynamic insights into migratory behavior changes
Morphological analysis:
Document and quantify morphological changes associated with EMT using phase-contrast microscopy
Perform cytoskeletal staining (F-actin) to visualize structural reorganization
Transcriptional regulation:
Use ChIP assays to study binding of EMT-related transcription factors to target promoters
Conduct reporter assays to measure activity of EMT-related promoters
Perform RT-qPCR to quantify mRNA levels of EMT markers
Rescue experiments:
These methodologies have demonstrated that DPYSL2 promotes EMT in bladder cancer via its interaction with PKM2, supporting tumor progression and metastasis .
Developing DPYSL2 as a therapeutic target requires systematic validation through multiple experimental approaches:
These approaches have supported DPYSL2 as a promising therapeutic target in both AML and bladder cancer, with potential applications in other cancer types as well .
Comparing DPYSL2 expression across cancer types requires careful methodology to ensure valid comparisons and meaningful integration:
Data normalization and standardization:
Use appropriate housekeeping genes or global normalization methods when comparing qPCR data
Apply batch correction algorithms when integrating data from different sources
Normalize Western blot data to loading controls and reference standards
Meta-analysis approaches:
Integrate data from public repositories (TCGA, GEO, GEPIA)
Apply statistical methods that account for inter-study heterogeneity
Use forest plots to visualize effect sizes across different cancer types
Multi-omics integration:
Correlate DPYSL2 protein expression with mRNA levels
Integrate with methylation, mutation, and copy number variation data
Create comprehensive molecular profiles across cancer types
Comparative analysis examples:
DPYSL2 is overexpressed in both AML and bladder cancer compared to normal tissues
High DPYSL2 expression correlates with poor prognosis in both cancer types, suggesting a conserved role in cancer progression
Different mechanisms may be involved: JAK2/STAT3/STAT5-PI3K P85/AKT/GSK3b in AML vs. PKM2 interaction in bladder cancer
Visualization and analysis tools:
Use heatmaps to display expression patterns across cancer types
Employ principal component analysis to identify cancer clusters based on DPYSL2 and related gene expression
Create Kaplan-Meier plots to compare prognostic impact across different cancers
This integrative approach has revealed that while DPYSL2 may have conserved oncogenic functions across cancer types, the specific molecular mechanisms may differ, requiring cancer-specific therapeutic strategies .
DPYSL2 undergoes extensive phosphorylation that regulates its function. When studying these modifications, researchers should consider:
Phospho-specific antibody selection:
Choose antibodies that specifically recognize phosphorylated forms of DPYSL2 at key residues
Validate specificity using phosphatase treatment controls
Consider using multiple phospho-specific antibodies to study different phosphorylation sites simultaneously
Sample preparation:
Include phosphatase inhibitors (sodium orthovanadate, sodium fluoride, β-glycerophosphate) in lysis buffers
Harvest samples quickly to preserve phosphorylation status
Consider using phospho-enrichment techniques for low-abundance phosphorylated forms
Experimental design:
Include positive controls (samples treated with phosphatase inhibitors) and negative controls (samples treated with phosphatases)
Use phos-tag gels to separate phosphorylated from non-phosphorylated forms
Consider kinase inhibitor treatments to identify responsible kinases
Interpretation challenges:
Phosphorylation can change rapidly in response to experimental conditions
Multiple phosphorylation sites may have different functional effects
Temporal dynamics of phosphorylation may be critical
Relevance to disease mechanisms:
These considerations ensure accurate detection and interpretation of DPYSL2 phosphorylation in both normal and pathological contexts.
Multiplex imaging allows simultaneous visualization of multiple proteins, providing valuable insights into complex cellular processes involving DPYSL2. Key methodological considerations include:
Antibody selection for multiplexing:
Choose DPYSL2 antibodies from different host species than other target antibodies
Validate each antibody individually before multiplexing
Ensure specificity through appropriate controls
Select antibodies with compatible fixation and antigen retrieval requirements
Sequential immunostaining approaches:
Perform cyclic immunofluorescence with antibody stripping between rounds
Use tyramide signal amplification (TSA) to allow use of antibodies from the same species
Document complete antibody removal between cycles
Spectral considerations:
Select fluorophores with minimal spectral overlap
Use spectral unmixing for fluorophores with partial overlap
Include single-stain controls for accurate spectral separation
Quantitative analysis:
Use image analysis software for co-localization quantification
Perform spatial relationship analysis between DPYSL2 and interacting partners
Consider using algorithms for cell phenotyping based on multiple markers
Applications in DPYSL2 research:
These approaches allow researchers to study DPYSL2 in its native cellular context while preserving spatial information and relationships with other proteins of interest.
While DPYSL2 is primarily a cytoplasmic protein, flow cytometry can be valuable for studying its expression in different cell populations. The following protocol optimizations are recommended:
Cell preparation:
Fix cells with 4% paraformaldehyde for 15 minutes at room temperature
Permeabilize with 0.1% saponin or 0.1% Triton X-100 to access intracellular DPYSL2
Block with 3-5% BSA or normal serum for 30 minutes
Antibody selection and staining:
Choose antibodies validated for flow cytometry applications
Titrate antibody concentration to determine optimal signal-to-noise ratio
Include isotype controls at the same concentration as the primary antibody
For multicolor panels, include fluorescence minus one (FMO) controls
Protocol specifics:
Incubate with primary DPYSL2 antibody for 30-60 minutes at room temperature
Wash 2-3 times with buffer containing 0.1% saponin to maintain permeabilization
Incubate with fluorophore-conjugated secondary antibody for 30 minutes
Wash thoroughly before analysis
Analysis considerations:
Applications in DPYSL2 research:
This approach enables quantitative assessment of DPYSL2 protein levels at the single-cell level, providing insights into expression heterogeneity within cell populations.
Recent research has revealed several non-canonical functions of DPYSL2 that merit further investigation:
Metabolic regulation:
DPYSL2's interaction with PKM2 and its role in promoting aerobic glycolysis in cancer cells represents a significant departure from its traditionally understood neuronal functions
Researchers should investigate whether DPYSL2 interacts with other metabolic enzymes beyond PKM2
The metabolic impact of DPYSL2 in non-cancer contexts remains largely unexplored
Cancer progression mechanisms:
Drug resistance pathways:
DPYSL2's role in HHT resistance in AML suggests potential involvement in drug resistance mechanisms
Researchers should investigate whether DPYSL2 contributes to resistance to other therapeutic agents
The structural basis for HHT-DPYSL2 interaction provides a starting point for developing targeted inhibitors
Signaling pathway integration:
Therapeutic target development:
Structure-based drug design targeting the DPYSL2-PKM2 interaction or the HHT binding site could yield novel therapeutic approaches
DPYSL2 antibody-drug conjugates represent an unexplored therapeutic strategy
These emerging roles suggest that DPYSL2 functions extend well beyond its canonical role in neuronal development and axon guidance, opening new avenues for therapeutic intervention.
Emerging technologies are expanding the capabilities of antibody-based research for DPYSL2 studies:
Super-resolution microscopy techniques:
Single-cell proteomics:
Mass cytometry (CyTOF) can simultaneously measure DPYSL2 and dozens of other proteins at the single-cell level
Single-cell Western blot technology allows protein analysis in individual cells
These approaches can reveal heterogeneity in DPYSL2 expression and modification within cell populations
Spatial transcriptomics and proteomics:
Combine DPYSL2 antibody staining with spatial transcriptomics to correlate protein localization with gene expression
Digital spatial profiling can map DPYSL2 distribution in tissue contexts with high precision
These technologies are particularly valuable for studying DPYSL2 in tumor microenvironments
Proximity-based labeling:
High-throughput screening platforms:
Antibody arrays and reverse phase protein arrays enable rapid profiling of DPYSL2 across multiple samples
CRISPR screens combined with DPYSL2 antibody detection can identify genes affecting DPYSL2 expression or function
These approaches accelerate discovery of pathways regulating DPYSL2 in disease contexts
These advanced technologies promise to deepen our understanding of DPYSL2's roles in normal physiology and disease, potentially leading to novel therapeutic strategies.
Integrating multi-omics data with antibody-based experiments provides a comprehensive understanding of DPYSL2 biology:
Correlative analysis approaches:
Compare DPYSL2 protein levels (detected by antibodies) with mRNA expression (from RNA-seq)
Correlate post-translational modifications with proteomic datasets
Integrate epigenetic data to understand regulatory mechanisms of DPYSL2 expression
Network analysis:
Place DPYSL2 in protein-protein interaction networks using antibody-based interaction studies
Overlay transcriptomic data to identify co-expressed genes
Identify regulatory modules controlling DPYSL2 expression and function
Methodological approaches:
Use DPYSL2 antibodies for ChIP-seq to identify genomic binding sites (if DPYSL2 has DNA-binding capacity)
Combine proteomics with DPYSL2 immunoprecipitation to identify interaction partners
Validate RNA-seq findings with protein-level measurements using DPYSL2 antibodies
Applications in cancer research:
RNA-seq analysis from TCGA database combined with immunohistochemistry has revealed the JAK2/STAT3/STAT5-PI3K P85/AKT/GSK3b axis as a critical pathway in DPYSL2-mediated AML development
Integration of proteomic and metabolomic data with DPYSL2 antibody studies has helped elucidate its role in regulating PKM2 and aerobic glycolysis in bladder cancer
Data visualization and analysis tools:
Use pathway analysis software to integrate antibody-based protein data with transcriptomic findings
Employ machine learning approaches to identify patterns across multi-omics datasets
Create integrative visualizations that connect DPYSL2 protein expression with other molecular features
This integrative approach provides a systems-level understanding of DPYSL2's role in health and disease, enabling more effective therapeutic targeting strategies.