DPYSL2 Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Applications in Research and Diagnostics

The antibody is employed in diverse experimental workflows to study DPYSL2’s roles in neurobiology and disease.

Primary Applications

  • Western Blot (WB): Detects DPYSL2 in lysates or tissue extracts, often used to analyze phosphorylation states (e.g., Thr514) .

  • Immunohistochemistry (IHC): Localizes DPYSL2 in fixed tissues, aiding in spatial expression profiling .

  • Immunofluorescence (IF): Visualizes DPYSL2 in live or fixed cells, useful for studying subcellular localization .

  • ELISA: Quantifies DPYSL2 levels in biological fluids or lysates, often paired with matched antibodies for sandwich assays .

  • Flow Cytometry: Measures DPYSL2 expression in cell populations .

Key Research Insights

  • Neuronal Polarity and Axon Guidance: DPYSL2 interacts with GSK3-β to regulate neuronal polarization. Nonphosphorylated DPYSL2 promotes axon-like neurite formation, while GSK3-β-induced phosphorylation inhibits it .

  • Neurodegenerative Diseases: DPYSL2 dysregulation is implicated in schizophrenia, with altered expression linked to abnormal axon branching .

  • Semaphorin Signaling: Required for class 3 semaphorin-mediated cytoskeletal remodeling and growth cone collapse .

Validation and Performance Data

Recombinant monoclonal antibodies undergo rigorous validation to ensure specificity and reproducibility.

Validation Examples

  • Proteintech’s 83990-4-PBS:

    • Sandwich ELISA: Detects DPYSL2 in K-562 cell extracts (167.28 ng/mL) and U-87 MG cells (105.42 ng/mL) .

    • Optimized Buffer: PBS-only formulation (BSA/azide-free) for conjugation flexibility .

  • Aviva’s 20D8:

    • IHC: Stains human breast cancer tissues at 1:50 dilution .

    • Flow Cytometry: Detects DPYSL2 in SH-SY5Y neuronal cells .

  • Assay Genie’s CAB14570:

    • KO-Validated: Confirmed specificity in knockout models .

Research Implications and Future Directions

DPYSL2 antibodies enable mechanistic studies of neuronal development and disease. Emerging areas include:

  1. Therapeutic Targeting: Inhibiting DPYSL2 phosphorylation to enhance axon regeneration in spinal cord injuries.

  2. Biomarker Discovery: Quantifying DPYSL2 levels in cerebrospinal fluid to diagnose neurodegenerative disorders.

  3. Multimodal Assays: Pairing recombinant antibodies with CRISPR-edited cell lines for high-throughput screening .

Technical Considerations

  • Storage: -80°C (long-term) or -20°C (short-term) .

  • Optimization: Dilution ranges vary (e.g., 1:50–1:200 for IHC) and require user validation .

  • Conjugation: Unconjugated antibodies allow customization for multiplex assays or mass cytometry .

Product Specs

Buffer
Rabbit IgG in phosphate buffered saline, pH 7.4, 150mM NaCl, 0.02% sodium azide and 50% glycerol.
Description

The DPYSL2 Recombinant Monoclonal Antibody is a meticulously produced reagent. The antibody is generated through a well-defined process involving in vitro cloning of the heavy and light chain genes into expression vectors. These vectors are then introduced into host cells, enabling the recombinant antibody's expression within a cell culture environment. Following expression, the DPYSL2 Recombinant Monoclonal Antibody is purified from the cell culture supernatant using affinity chromatography. This antibody demonstrates high specificity for the human DPYSL2 protein. Its versatility is evident in its suitability for a wide range of applications, including ELISA, IHC, IF, and FC.

DPYSL2 is a multifaceted protein playing a pivotal role in the development and function of the nervous system. Its involvement encompasses neurite outgrowth, axon guidance, synaptic plasticity, neuronal signaling, and neuronal survival. Disruptions in DPYSL2 function are associated with various neurological disorders and conditions.

Form
Liquid
Lead Time
Typically, we can ship products within 1-3 business days after receiving your orders. Delivery timelines may vary based on the purchasing method or location. For specific delivery information, please contact your local distributors.
Synonyms
Dihydropyrimidinase-related protein 2 (DRP-2) (Collapsin response mediator protein 2) (CRMP-2) (N2A3) (Unc-33-like phosphoprotein 2) (ULIP-2), DPYSL2, CRMP2 ULIP2
Target Names
DPYSL2
Uniprot No.

Target Background

Function

DPYSL2 plays a significant role in neuronal development and polarity, as well as in axon growth and guidance, neuronal growth cone collapse, and cell migration. It is essential for signaling by class 3 semaphorins and subsequent remodeling of the cytoskeleton. Additionally, DPYSL2 may play a role in endocytosis.

Gene References Into Functions
  1. Cross-talk between distinct CRMP2 posttranslational modifications is a key factor in determining NaV1.7 trafficking and localization PMID: 27940916
  2. The crystal structure of human tetrameric CRMP-2 has been elucidated. PMID: 28044206
  3. Increased pCRMP2 may underlie the axonal pathology of Lewy body dementias. PMID: 27609071
  4. These data identify a novel oncogenic mechanism where CDK5 activation induces CRMP2A phosphorylation in the nuclei of tumor cells. PMID: 26555036
  5. Polymorphisms of the DPYSL2 gene in humans may be associated with the development of schizophrenia. PMID: 25847191
  6. Functional variants in DPYSL2 sequence increase the risk of schizophrenia and suggest a link to mTOR signaling. PMID: 25416705
  7. Changes for CRMP2, TCP1epsilon, TPM2, and 14-3-3gamma were confirmed in experimental tumors and in a series of 28 human SI-NETs. PMID: 25224486
  8. Reduced CRMP2 expression and elevated expression of nuclear phosphorylated CRMP2 may be associated with breast cancer progression. PMID: 23381229
  9. Levels of total GSK3 were decreased in the Huntington disease-affected frontal cortex, and this correlated with decreased phosphorylated CRMP2. PMID: 24634145
  10. High dihydropyrimidinase-related protein 2 expression is associated with lung cancer. PMID: 24518087
  11. Genetic variants in DPYSL2 may play a role in susceptibility to alcohol dependence. PMID: 23846846
  12. A specific and reversible intermolecular Cys-504-Cys-504 dithiol-disulfide switch in homotetrameric CRMP2 determines two conformations of the quaternary CRMP2 complex that controls axonal outgrowth and thus neuronal development. PMID: 24133216
  13. CRMP-2-dependent regulation of ROCK II activity is mediated through interaction of the CRMP-2L N terminus with the ROCK II catalytic domain as well as by GSK3-dependent phosphorylation of CRMP-2. PMID: 24036111
  14. A novel regulatory mechanism that utilizes CRMP2 SUMOylation to choreograph NaV1.7 trafficking has been identified. PMID: 23836888
  15. New insights into CRMP-2 as a controller of myosin II-mediated cellular functions through the inhibition of ROCK II in nonneuronal cells have been gained. PMID: 22431514
  16. No Alzheimer disease-associated differences in CNP and DPYSL2 promoter DNA methylation were observed. PMID: 22954668
  17. Deletion analysis of CRMP-2 identified a 51 amino acid sequence in the C-terminus that is required for targeting to the basal body and primary cilium. This domain contains GSK-3beta phosphorylation sites. PMID: 23185275
  18. High levels of nuclear phosphorylated CRMP-2 is associated with lung cancer. PMID: 23023514
  19. CRMP2 hyperphosphorylation is specific to Alzheimer's disease and is not a common event in all forms of dementia and neurodegeneration, especially other tauopathies. PMID: 21860090
  20. Findings suggest a major role of phosphorylated CRMP-2 as a mechanism involved in process retraction induced by reactive oxygen species. PMID: 22443207
  21. HTLV-1 has the capacity to influence the CRMP2/PI3K/Akt axis, thus positively controlling cytoskeleton organization and lymphocyte migration. PMID: 22227566
  22. CRMP2 controls dendritic projection; the phosphorylation of CRMP2 at Ser522 is essential for proper dendritic field organization for axonal guidance and spine development. PMID: 22279220
  23. This review discusses the essential biology of CRMP2 in the context of nascent data implicating CRMP2 perturbations as either a correlate of or plausible contributor to diverse neuropathologies. PMID: 21271304
  24. Data support DPYSL2 and the surrounding genomic region as a susceptibility locus for schizophrenia. PMID: 21302347
  25. Data provide the first trafficking regulatory role for Crmp2 in non-neuronal cells and support a model in which Crmp2 is an important endocytic regulatory protein that links MICAL-L1.EHD1-based vesicular transport to dynein motors. PMID: 20801876
  26. DPYSL2 does not have a major function in schizophrenia in Japanese subjects. PMID: 20414250
  27. CRMP2, as a tubulin direct binder, may be a GTPase-activating protein (GAP) of tubulin in neurite formation, and its GAP activity may be regulated by an intramolecular interaction with an N-terminal inhibitory region. PMID: 19666111
  28. Aberrant expression of dihydropyrimidinase-related proteins-2,-3, and -4 in fetal Down syndrome brain. PMID: 11771764
  29. No significant associations were found between five polymorphisms of the DRP-2 gene and Bipolar Disorder, nor were associations detected between either of the polymorphisms and bipolar subtypes I and II. PMID: 12951196
  30. Here we show that glial cell line-derived neurotrophic factor (GDNF) enhances CRMP-2 expression in TGW human neuroblastoma cells via activation of RET receptor tyrosine kinase. PMID: 15207709
  31. A significant decrease of crmp-2 protein may represent or underlie impaired neuronal plasticity, neurodegeneration, and wiring of the brain in mesial temporal lobe epilepsy. PMID: 15672539
  32. CRMP-2 transports the Sra-1/WAVE1 complex to axons in a kinesin-1-dependent manner and thereby regulates axon outgrowth and formation. PMID: 16260607
  33. Collapsin response mediator protein-2 transcriptional activity is inhibited by all-trans-retinoic acid during SH-SY5Y neuroblastoma cell differentiation. PMID: 17229153
  34. These observations implicate hyperphosphorylation of CRMP2 as an early event in the development of AD and suggest that it can be induced by a severe APP over-expression and/or processing defect. PMID: 17683481
  35. Results indicate that CRMP-2 may be a novel colorectal cancer biomarker. PMID: 18203259
  36. Association of neurofibromin and CRMP-2 is essential for neuronal cell differentiation. PMID: 18218617
  37. Relative resistance to phosphatases might be a common feature of Cdk5 substrates and could contribute to the hyperphosphorylation of CRMP2 and Tau observed in Alzheimer disease. PMID: 18460467
  38. Data suggest that collapsin response mediator protein-2 (CRMP-2) is a novel calmodulin-binding protein, and that CaM binding may play an important role in regulating CRMP-2 functions. PMID: 19151921
  39. Given that CRMP-2 is a key regulator of axon elongation, this interference with cytoplasmic dynein function by CRMP-2 might have an important role in axon formation and neuronal development. PMID: 19659462
  40. A significant association was found between a single nucleotide polymorphism of the DRP-2 gene and schizophrenia in a North American sample. PMID: 15858820
Database Links

HGNC: 3014

OMIM: 602463

KEGG: hsa:1808

STRING: 9606.ENSP00000309539

UniGene: Hs.593187

Protein Families
Metallo-dependent hydrolases superfamily, Hydantoinase/dihydropyrimidinase family
Subcellular Location
Cytoplasm, cytosol. Cytoplasm, cytoskeleton. Membrane. Note=Tightly but non-covalently associated with membranes.
Tissue Specificity
Ubiquitous.

Q&A

What is DPYSL2 and what are its primary cellular functions?

DPYSL2, also known as CRMP2 (Collapsin Response Mediator Protein 2), is a multifunctional protein that plays central roles in nervous system development and function. It forms homo- and hetero-tetramers that facilitate neuron guidance, growth, and polarity establishment. The protein promotes microtubule assembly and is required for Sema3A-mediated growth cone collapse. Additionally, DPYSL2 plays critical roles in synaptic signaling through interactions with calcium channels. At the molecular level, DPYSL2 is involved in neurite outgrowth, axon guidance, synaptic plasticity, neuronal signaling, and neuronal survival mechanisms. Dysregulation of this protein has been implicated in various neurological disorders, including Alzheimer's disease, schizophrenia, and epilepsy .

How is DPYSL2 involved in neuronal stem cell differentiation?

DPYSL2 functions as a key regulator of neural stem cell (NSC) differentiation. Research has identified DPYSL2 as one of the differentially expressed proteins during NSC differentiation, particularly when comparing serum-alone versus PNS (peripheral nervous system) + serum treatment groups. Experimental evidence shows that overexpression of DPYSL2 in NSCs promotes differentiation toward neuronal lineages more significantly than toward astrocytic lineages. Specifically, DPYSL2 overexpression leads to a significant increase in Tuj1-positive cells (neurons) compared to control groups, with a less pronounced but still significant increase in GFAP-positive cells (astrocytes). This indicates that DPYSL2 plays a preferential role in directing NSC differentiation toward neurons, making it a potential target for neuronal regeneration strategies .

What are the structural characteristics of human DPYSL2 protein?

Human DPYSL2 is a 62.3 kDa protein consisting of 572 amino acid residues in its canonical form. The protein is primarily localized in the cytoplasm, though it also associates with membranes and the cytoskeleton. Up to two different isoforms have been reported through alternative splicing. The protein contains the sequence "PFPDFVYKRIKARSRLAELRGVPRGLYDGPVCEVSV TPKTVTPASSAKTSPAKQQAPPVRNLHQSGFSLSGAQIDDNIPRRTTQRIVAPPGGRANI TSLG" between amino acids 473-572, which has been utilized as an immunogen for antibody development. DPYSL2 belongs to the Hydantoinase/dihydropyrimidinase protein family and has been evolutionarily conserved across species, with orthologs reported in mouse, rat, bovine, frog, chimpanzee, and chicken .

How are DPYSL2 recombinant monoclonal antibodies produced?

DPYSL2 recombinant monoclonal antibodies are produced through a meticulously executed process of molecular cloning and cell culture techniques. The process begins with in vitro cloning of genes encoding both the heavy and light chains of the DPYSL2 antibody into appropriate expression vectors. These vectors are then introduced into host cells, creating an environment conducive to recombinant antibody expression within a cell culture setting. Following expression, the DPYSL2 recombinant monoclonal antibody is purified from the cell culture supernatant using affinity chromatography, which yields a highly specific antibody preparation. This recombinant approach ensures consistent production of antibodies with identical binding characteristics across different batches, providing researchers with reliable tools for DPYSL2 detection and analysis .

What are the key differences between polyclonal and recombinant monoclonal antibodies for DPYSL2 research?

Recombinant monoclonal antibodies offer several distinct advantages over polyclonal antibodies for DPYSL2 research. While polyclonal antibodies are derived from multiple B cell clones and recognize different epitopes of DPYSL2, recombinant monoclonal antibodies originate from a single clone and target a specific epitope with high specificity. This epitope specificity is crucial when investigating particular domains or post-translational modifications of DPYSL2. Furthermore, recombinant monoclonal antibodies provide consistent batch-to-batch reproducibility, eliminating the variability inherent to polyclonal antibodies. For DPYSL2 research, where distinguishing between closely related CRMP family members or specific phosphorylated forms is often necessary, the precise epitope recognition of recombinant monoclonal antibodies significantly enhances experimental reliability and interpretation .

What validation methods confirm specificity of DPYSL2 antibodies?

The specificity of DPYSL2 antibodies is confirmed through multiple validation approaches. Knockout (KO) validation represents a gold standard, where the antibody is tested against samples from DPYSL2 knockout models to confirm absence of signal. Western blot analysis demonstrates the antibody recognizes a protein of the expected molecular weight (approximately 62 kDa). Immunofluorescence studies in relevant tissues, such as mouse and rat brain sections, verify appropriate tissue and subcellular localization patterns. Cross-reactivity testing across human, mouse, and rat samples confirms species reactivity. Additionally, immunoprecipitation followed by mass spectrometry can verify the exact protein being recognized. For DPYSL2 antibodies, positive controls typically include HeLa cells, mouse/rat lung, and mouse/rat heart tissues, which express detectable levels of the target protein .

What are the optimal experimental conditions for DPYSL2 antibody applications in Western blotting?

For Western blot applications using DPYSL2 recombinant monoclonal antibodies, several parameters require optimization. The recommended dilution range is 1:500 to 1:5000, with optimal dilution determined through titration experiments for each specific antibody and sample type. Samples should be prepared in a buffer containing protease inhibitors to prevent degradation of DPYSL2, which has a calculated molecular weight of 62 kDa. When performing SDS-PAGE, 10-12% gels are typically sufficient for resolving DPYSL2. During transfer, standard PVDF or nitrocellulose membranes are suitable. Blocking should be performed with 5% non-fat dry milk or BSA in TBST, and primary antibody incubation is recommended overnight at 4°C. For detection, standard ECL systems are appropriate, with exposure times adjusted according to signal intensity. Positive control samples include HeLa cells, mouse lung, mouse heart, rat lung, and rat heart tissues, which all express detectable levels of DPYSL2 .

How should immunofluorescence protocols be optimized for DPYSL2 detection in neural tissues?

Immunofluorescence detection of DPYSL2 in neural tissues requires specific protocol optimization. The recommended antibody dilution range is 1:50 to 1:200, with the optimal concentration determined empirically for each tissue type. For tissue preparation, 4% paraformaldehyde fixation followed by either frozen sectioning or paraffin embedding is suitable, though antigen retrieval may be necessary for paraffin sections. Permeabilization with 0.1-0.3% Triton X-100 facilitates antibody access to cytoplasmic DPYSL2. Blocking with 10% normal serum from the species of the secondary antibody for 1-2 hours helps reduce background. Primary antibody incubation should be conducted overnight at 4°C, followed by appropriate fluorophore-conjugated secondary antibody incubation. DAPI counterstaining helps visualize nuclei. Confocal microscopy at 40x magnification has been successfully used to detect DPYSL2 expression in both mouse and rat brain tissues. Cytoplasmic, membrane, and cytoskeletal localization patterns should be evident in properly optimized experiments .

What considerations are important when using DPYSL2 antibodies for studying neural stem cell differentiation?

When using DPYSL2 antibodies to study neural stem cell (NSC) differentiation, several methodological considerations are crucial. First, temporal dynamics must be accounted for, as DPYSL2 expression changes significantly during differentiation processes. Sampling at multiple time points (typically days 1, 3, 7, and 14 after induction of differentiation) provides comprehensive data. Second, co-immunostaining with lineage markers such as Tuj1 (for neurons) and GFAP (for astrocytes) enables correlation between DPYSL2 expression and cell fate. Third, quantification approaches should include both the percentage of DPYSL2-positive cells and the intensity of DPYSL2 staining. Research has demonstrated that DPYSL2 overexpression promotes NSC differentiation particularly toward neuronal lineages (0.525 ± 0.02 Tuj1-positive cells in DPYSL2 overexpression group versus 0.271 ± 0.05 in control group, p<0.05). Finally, experimental manipulations of DPYSL2 expression through overexpression or knockdown approaches can establish causative relationships between DPYSL2 levels and differentiation outcomes .

How do post-translational modifications of DPYSL2 affect antibody recognition?

Post-translational modifications (PTMs) significantly impact DPYSL2 antibody recognition and have crucial biological implications. DPYSL2 undergoes extensive phosphorylation, particularly by kinases including GSK3β, CDK5, and ROCK, which regulate its function in neurite outgrowth and axon guidance. Hyperphosphorylation of DPYSL2 has been implicated in Alzheimer's disease pathogenesis. When selecting antibodies, researchers must determine whether they need antibodies that recognize total DPYSL2 regardless of phosphorylation state, or phospho-specific antibodies that detect particular phosphorylated residues. Antibodies raised against sequences containing phosphorylation sites may show differential binding depending on the phosphorylation status. For comprehensive analysis, complementary approaches using both total and phospho-specific antibodies are recommended. Additionally, treatment with phosphatases before immunodetection can help distinguish whether observed changes in DPYSL2 detection are due to altered expression or modified phosphorylation states .

What strategies enable distinction between DPYSL2 and other CRMP family members?

Distinguishing DPYSL2 (CRMP2) from other CRMP family members (CRMP1, CRMP3, CRMP4, and CRMP5) requires careful antibody selection and experimental design. Despite sharing approximately 50-75% sequence homology, these proteins have distinct functions in neuronal development. For antibody-based discrimination, selecting antibodies raised against regions with maximal sequence divergence is essential. The C-terminal region (amino acids 473-572) contains distinctive sequences often used for generating DPYSL2-specific antibodies. Western blotting can distinguish family members partly by molecular weight differences: DPYSL2 (62 kDa), CRMP1 (62 kDa), CRMP3 (64 kDa), CRMP4 (65 kDa), and CRMP5 (68 kDa). Isoelectric focusing combined with Western blotting offers additional separation based on differing isoelectric points. For transcriptional analysis, designing PCR primers specifically in non-homologous regions ensures amplification of only the target CRMP. When possible, validation experiments should include positive controls for each CRMP family member to confirm specificity .

How can DPYSL2 antibodies be effectively employed in studying neurodegenerative diseases?

DPYSL2 antibodies provide valuable tools for investigating neurodegenerative mechanisms, particularly in Alzheimer's disease where DPYSL2 hyperphosphorylation appears to play a pathogenic role. When studying neurodegenerative conditions, researchers should employ a multi-faceted approach. First, analyzing both total and phosphorylated DPYSL2 levels in patient-derived tissues or appropriate disease models is essential, as changes in phosphorylation often exceed changes in total protein levels. Second, co-immunoprecipitation studies using DPYSL2 antibodies can reveal altered protein interactions in disease states, such as disrupted binding to tubulin or increased association with tau. Third, immunohistochemistry in brain sections can demonstrate altered subcellular localization or inclusion formation. Fourth, longitudinal studies in animal models using DPYSL2 antibodies can track progressive changes during disease development. Finally, therapeutic intervention studies targeting DPYSL2 phosphorylation should incorporate antibodies that specifically recognize the relevant phosphorylation sites to monitor treatment efficacy .

What are the common sources of background and non-specific binding when using DPYSL2 antibodies?

Non-specific binding and background issues with DPYSL2 antibodies can arise from several sources that require systematic troubleshooting. Cross-reactivity with other CRMP family members represents a primary concern due to sequence homology. This can be addressed by using antibodies raised against unique epitopes and validating with known positive and negative controls. Secondary antibody cross-reactivity, particularly in multi-labeling experiments, can be minimized by using highly cross-adsorbed secondary antibodies and implementing appropriate blocking steps. Endogenous peroxidase or phosphatase activity in tissues can generate false-positive signals in enzyme-based detection systems; this requires quenching steps with H₂O₂ or levamisole, respectively. Autofluorescence, especially in neural tissues with lipofuscin, can be reduced using Sudan Black B treatment or spectral unmixing during confocal microscopy. Finally, overfixation of tissues may mask epitopes; titrating fixation conditions and implementing appropriate antigen retrieval methods can restore antibody accessibility to DPYSL2 epitopes .

How can researchers optimize co-immunoprecipitation experiments with DPYSL2 antibodies?

Successful co-immunoprecipitation (co-IP) of DPYSL2 and its interaction partners requires optimization of several parameters. Lysis buffer composition is critical—mild non-ionic detergents like NP-40 or Triton X-100 (0.5-1%) preserve protein interactions, while phosphatase inhibitors maintain phosphorylation-dependent interactions. Pre-clearing lysates with protein A/G beads reduces non-specific binding. For the antibody-antigen interaction, using 2-5 μg of DPYSL2 antibody per 500 μg of total protein typically yields good results. Incubation should occur overnight at 4°C with gentle rotation to preserve fragile protein complexes. When investigating interactions with cytoskeletal components, adding stabilizing agents like taxol (for microtubules) or phalloidin (for actin) to the lysis buffer can preserve these associations. For detecting transient interactions, chemical crosslinking prior to lysis using membrane-permeable crosslinkers like DSP (dithiobis(succinimidyl propionate)) may be necessary. Finally, validation of co-IP results using reciprocal co-IP (using antibodies against the interaction partner) provides stronger evidence for genuine protein-protein interactions .

What controls should be included when quantifying DPYSL2 expression in comparative studies?

Robust quantification of DPYSL2 expression in comparative studies requires implementation of several control measures. Loading controls are essential: for Western blotting, housekeeping proteins like GAPDH, β-actin, or α-tubulin should be used, while normalizing to total protein loading (assessed by Ponceau S or similar stains) provides an alternative approach less susceptible to experimental manipulations. Calibration curves using purified recombinant DPYSL2 protein enable absolute quantification rather than relative comparisons. For immunohistochemistry/immunofluorescence studies, standardized image acquisition parameters and systematic sampling approaches prevent selection bias. Technical replicates (minimum of three) account for methodological variation, while biological replicates address sample-to-sample variability. For experiments manipulating DPYSL2 expression, both positive controls (known to increase DPYSL2) and negative controls (known to decrease DPYSL2) provide reference points. When comparing phosphorylated and total DPYSL2, analyzing the phospho-to-total ratio rather than absolute values often provides more meaningful biological insights .

How should researchers analyze DPYSL2 expression across different neural cell populations?

Analyzing DPYSL2 expression across diverse neural cell populations requires integrating multiple analytical approaches. Flow cytometry offers quantitative single-cell analysis when combined with permeabilization protocols suitable for detecting cytoplasmic DPYSL2 alongside surface markers of neural subtypes. Fluorescence-activated cell sorting (FACS) followed by Western blotting or qPCR enables examination of DPYSL2 expression in isolated neural subpopulations. For tissue sections, multiplexed immunofluorescence combining DPYSL2 antibodies with markers for neurons (NeuN, Tuj1), astrocytes (GFAP), oligodendrocytes (MBP, Olig2), and neural stem cells (Nestin, Sox2) reveals cell type-specific expression patterns. Laser capture microdissection coupled with proteomics or transcriptomics provides region-specific analysis. Single-cell RNA sequencing data can be mined to examine DPYSL2 expression variability across neural subtypes. Quantitative analysis should include both the percentage of DPYSL2-positive cells within each population and the intensity of expression, as both metrics provide complementary information about regulation patterns .

What bioinformatic approaches help interpret DPYSL2 antibody-based proteomics data?

Bioinformatic analysis enhances interpretation of DPYSL2 antibody-based proteomics data by revealing functional networks and regulatory mechanisms. Pathway enrichment analysis using databases like KEGG, Reactome, or GO identifies biological processes associated with DPYSL2 and its interacting partners. Protein-protein interaction network analysis through tools like STRING (9606.ENSP00000309539) places DPYSL2 in its functional context, with visualization software like Cytoscape enabling graphical representation of these networks. Motif analysis around phosphorylation sites recognized by phospho-specific antibodies can reveal regulatory kinases. Comparative analysis across species leveraging orthologous relationships (with DPYSL2 orthologs identified in mouse, rat, bovine, frog, chimpanzee, and chicken) highlights evolutionarily conserved functions. Integration of antibody-derived proteomics data with transcriptomics datasets through correlation analysis can identify potential transcriptional regulators of DPYSL2. When analyzing disease-related changes, comparison with existing databases of disease-associated proteins helps contextualize findings within broader pathological mechanisms .

How can contradictory results between different DPYSL2 antibodies be reconciled?

Contradictory results between different DPYSL2 antibodies represent a significant research challenge requiring systematic investigation. Epitope mapping should be performed to determine precisely which regions of DPYSL2 each antibody recognizes, as differences may reflect epitope availability rather than actual protein abundance. Post-translational modifications, particularly phosphorylation, may mask epitopes recognized by certain antibodies while leaving others unaffected. Alternative splicing of DPYSL2 can generate isoforms lacking specific epitopes, leading to discrepant results between antibodies targeting different regions. Cross-reactivity with other CRMP family members should be evaluated through controls using recombinant proteins. Methodological differences in sample preparation (extraction buffers, fixation protocols) may differentially expose epitopes. For definitive resolution, orthogonal techniques such as mass spectrometry can provide antibody-independent protein identification and quantification. When publishing results, transparent reporting of the specific antibody used (including catalog number and lot), experimental conditions, and observed molecular weight is essential for result interpretation and reproducibility .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.