DCHS1 (Dachsous cadherin related 1) is a calcium-dependent cell-adhesion membrane protein encoded by the DCHS1 gene. In humans, the canonical protein has 3298 amino acid residues with a mass of 346.2 kDa and is localized in the cell membrane. It's significant because it plays critical roles in neuroprogenitor cell proliferation and differentiation, proper morphogenesis of the mitral valve, and is involved in cell adhesion mechanisms . DCHS1 is also known by several synonyms including CDH25, CDHR6, FIB1, MMVP2, MVP2, PCDH16, VMLDS1, and CDH19 . Research interest in DCHS1 has grown due to its association with diseases like Van Maldergem syndrome and mitral valve prolapse .
DCHS1 antibodies are most effectively utilized in:
Western Blot (1:500-1000 dilution): For detecting protein expression levels and molecular weight confirmation
Immunofluorescence (IF): For cellular localization studies (1:50-100 dilution)
Immunohistochemistry (IHC): For tissue expression pattern analysis
Immunocytochemistry (ICC): For subcellular localization (1:50-100 dilution)
The applications should be selected based on your specific research question, with ELISA being preferred for quantification and immunohistochemistry for spatial expression patterns .
Selection of the appropriate DCHS1 antibody should be based on:
Target epitope: Antibodies targeting different regions (e.g., AA 2964-2981) may have different specificities
Species reactivity: Ensure the antibody reacts with your species of interest (human, mouse, rat, etc.)
Clonality: Polyclonal antibodies offer broader epitope recognition, while monoclonal antibodies provide higher specificity
Conjugation requirements: Select based on detection method - unconjugated for standard IHC/WB, or specific conjugates (FITC, Biotin, HRP) for specialized applications
Validation data: Review data showing antibody performance in your intended application
For applications requiring high sensitivity in detecting native protein conformation, antibodies validated for immunofluorescence are recommended .
DCHS1 shows variable expression across normal human tissues:
For accurate assessment of DCHS1 expression:
RNA-level analysis: qPCR with validated primers or RNA-seq analysis compared to reference genes
Protein-level analysis: Immunohistochemistry with properly validated antibodies at 1:100 dilution
Subcellular localization: Immunofluorescence microscopy focusing on membrane localization
When comparing expression between normal and pathological states, researchers should use consistent methodology and include proper controls to account for tissue-specific baseline expression levels .
DCHS1 expression varies significantly across cancer types:
Upregulated in:
Glioblastoma (GBM)
Head and neck squamous cell carcinoma (HNSC)
Kidney renal clear cell carcinoma (KIRC)
Pheochromocytoma and Paraganglioma (PCPG)
Downregulated in:
Cervical squamous cell carcinoma (CESC)
Lung adenocarcinoma (LUAD)
Breast invasive carcinoma (BRCA)
Kidney renal papillary cell carcinoma (KIRP)
Prostate adenocarcinoma (PRAD)
Recommended techniques for assessment:
RNA-seq analysis: Comparing tumor vs. normal tissue from TCGA and GTEx databases
Immunohistochemistry: Using tissue microarrays with scoring systems for intensity (0-3+)
Western blot: For quantitative protein expression analysis
ROC curve analysis: To evaluate diagnostic potential (AUC values ranging from 0.717-0.980 across cancer types)
The combination of these techniques provides comprehensive evidence of DCHS1's differential expression and potential as a diagnostic biomarker in specific cancers .
To investigate DCHS1's role in cell adhesion and migration, researchers can employ:
Gene silencing/overexpression studies:
Functional assays:
Protein stability and dynamics:
In vivo models:
These approaches should be combined to establish comprehensive understanding of DCHS1's functional role in cellular processes .
DCHS1 has been implicated in epithelial-mesenchymal transition (EMT), a process critical in development and cancer progression. To investigate this relationship:
EMT marker profiling following DCHS1 modulation:
Functional assays to assess EMT phenotype:
Cell morphology changes (epithelial to spindle-shaped)
Migration and invasion capacity in Transwell chambers
Resistance to anoikis (suspension culture survival)
Colony formation in 3D matrices
Signaling pathway analysis:
In vitro validation:
Gene Set Enrichment Analysis (GSEA) has revealed significant association between DCHS1 expression and EMT pathways across multiple cancer types, suggesting a mechanistic link that warrants further investigation .
The connection between DCHS1 mutations and mitral valve prolapse (MVP) has been established through complementary genetic and functional approaches:
Genetic evidence:
Protein stability studies:
Animal model validation:
Expression analysis:
This multi-modal evidence establishes DCHS1 as a critical factor in mitral valve development, with mutations leading to MVP through mechanisms involving protein stability and altered developmental processes .
DCHS1 expression shows significant correlation with immune cell infiltration in the tumor microenvironment:
Key immune cell correlations:
Cancer type specificity:
Methodological approaches:
Single-cell RNA sequencing for cell type-specific analysis
Deconvolution algorithms (e.g., CIBERSORT, xCell) for bulk RNA-seq data
Multiplex immunohistochemistry for spatial context
Digital spatial profiling for high-dimensional analysis
Therapeutic implications:
These correlations suggest DCHS1 may influence tumor progression through modulation of the immune microenvironment, particularly through interactions with stromal components like CAFs and ECs .
While standard applications for DCHS1 antibodies are well-established, more challenging techniques like ChIP require optimization:
Antibody selection considerations:
Protocol optimization:
Crosslinking: Start with 1% formaldehyde for 10 minutes at room temperature
Sonication: Optimize cycles to achieve 200-500bp DNA fragments
Antibody concentration: Test range from 2-10μg per ChIP reaction
Incubation time: Extend to overnight at 4°C for maximum binding
Washing stringency: Balance between reducing background and maintaining signal
Controls:
IgG negative control: Crucial for determining background signal
Input DNA: Use 5-10% of starting material
Positive control loci: Include known DCHS1 binding regions if available
Sequential ChIP: Consider for protein complex studies
Validation approaches:
qPCR of target regions vs. non-binding regions
Western blot of immunoprecipitated material
Mass spectrometry verification of pulled-down proteins
Given DCHS1's role as a membrane protein, membrane extraction and solubilization steps may require particular attention to ensure efficient immunoprecipitation while maintaining protein-DNA interactions.
Integrating DCHS1 antibodies into super-resolution microscopy requires specialized optimization:
Antibody preparation for super-resolution techniques:
Select high-affinity antibodies with minimal background
Consider directly conjugated antibodies to reduce localization errors
For STORM/PALM: Use photoswitchable fluorophore conjugates
For STED: Select fluorophores with appropriate depletion properties
Antibody concentration: Typically lower than conventional microscopy (1:200-1:500)
Sample preparation considerations:
Fixation: 4% PFA with mild permeabilization to preserve membrane structures
Buffer optimization: Oxygen scavenging systems for STORM
Mounting media: Specialized for each super-resolution technique
Cell culture on specific coverslips (high precision thickness)
Validation approaches:
Correlative microscopy with conventional techniques
Dual-color imaging with known markers of cell membrane or cadherin family
Antibody clustering analysis
Quantitative assessment of labeling density and specificity
Analysis strategies:
Cluster analysis of DCHS1 distribution
Co-localization with interacting partners
Temporal dynamics using live-cell super-resolution
3D reconstruction of membrane distribution patterns
When designing super-resolution experiments, researchers should consider DCHS1's membrane localization and potential clustering properties, which may be particularly well-suited for techniques like PALM or STORM that can resolve nanoscale protein organization .
A multi-modal approach combining antibody detection with genetic manipulation provides powerful insights into DCHS1 function:
Spatiotemporal expression mapping:
Genetic manipulation approaches:
Combinatorial analysis techniques:
Single-cell RNA-seq with antibody-based cell sorting
ATAC-seq combined with DCHS1 ChIP to assess chromatin accessibility
Proximity labeling (BioID, APEX) with DCHS1 antibody validation
Optogenetic control of DCHS1 with antibody-based readouts
In vivo developmental analysis:
Studies with Dchs1 mutant mice have revealed that while no morphological defects are observed in Dchs1+/- mice during early embryonic development (E11.5–E13.5), significant changes in mitral-valve shape become apparent at later timepoints (E15.5–E17.5), with more severe phenotypes in Dchs1-/- animals .
AI and machine learning approaches are revolutionizing antibody design and validation processes for targets like DCHS1:
AI-driven antibody design approaches:
Sequence-based antibody design models like DyAb can predict affinity improvements based on limited training data
Deep learning models can incorporate protein structural information to optimize binding sites
Machine learning algorithms can select combinations of mutations to enhance antibody properties
Performance metrics and validation:
Correlation between predicted and measured improvements in affinity (ΔpKD)
Models have achieved Pearson correlation coefficients of r=0.84 and Spearman coefficients of ρ=0.84 for antibody variant prediction
High expression and binding rates (>85%) for AI-designed antibodies compared to traditional methods
Implementation strategies:
Applications specific to DCHS1:
Epitope optimization targeting crucial functional domains
Cross-reactivity prediction across species for comparative studies
Affinity maturation for improved sensitivity in low-expression contexts
Recent advancements have shown that AI-designed antibodies maintain high expression and binding rates comparable to single point mutants while significantly improving target affinity, suggesting promising applications for DCHS1 research .
Emerging single-cell technologies offer unprecedented insights when combined with DCHS1 antibodies:
Single-cell protein analysis techniques:
Mass cytometry (CyTOF) with DCHS1 antibodies conjugated to metal isotopes
CITE-seq combining antibody detection with transcriptomics
Single-cell Western blotting for protein expression heterogeneity
Microfluidic antibody capture for quantitative single-cell surface protein analysis
Spatial profiling approaches:
Imaging mass cytometry for tissue section analysis with DCHS1 antibodies
Co-detection by indexing (CODEX) for highly multiplexed tissue imaging
Digital spatial profiling combining region selection with molecular quantification
4i (iterative indirect immunofluorescence imaging) for sequential antibody staining
Functional single-cell applications:
Live-cell antibody imaging with microfluidic cell capture
Single-cell secretion assays combined with DCHS1 surface detection
Clonal tracking with DCHS1 expression correlation
Lineage tracing combined with antibody-based detection
Data integration strategies:
Multi-omics integration of DCHS1 protein data with transcriptomics
Trajectory analysis correlating DCHS1 expression with cell state transitions
Cell-cell interaction mapping based on DCHS1 and partner protein expression
Spatial statistics for tissue organization analysis
These techniques are particularly valuable for understanding DCHS1's heterogeneous expression in cancer and its correlation with immune cell infiltration in the tumor microenvironment, potentially revealing new therapeutic targets and prognostic indicators .