EVC Antibody

Shipped with Ice Packs
In Stock

Description

Overview of EVC Antibody

Target protein:

  • Recognizes EvC ciliary complex subunit 1 (EVC), a 992-amino acid transmembrane protein with a mass of ~112 kDa .

  • Localizes to cell membranes and cytoplasm, playing a role in Hedgehog (Hh) signaling regulation .

Associated diseases:

  • Linked to Ellis-van Creveld syndrome (chondroectodermal dysplasia) and Weyers acrodental dysostosis .

  • Overexpressed in HTLV-1-infected cells and ATL, contributing to pro-survival signaling .

Synonyms:

  • EVC1, EVCL, DWF-1, Ellis van Creveld protein .

Role in Hedgehog Signaling and Disease

  • Oncogenic function:

    • EVC overexpression in ATL enhances Hedgehog pathway activation, promoting cell survival .

    • Validated via immunohistochemistry in 87.5% of aggressive ATL cases using commercial antibodies .

  • Ciliary regulation:

    • EVC-EVC2 complex stability and ciliary targeting depend on post-translational modifications:

      • Ubiquitination: Reduces protein levels and disrupts ciliary localization .

      • Sumoylation: Enhances interaction with EFCAB7-IQCE complex, improving EvC zone targeting .

Validation and Specificity Data

  • Antibody validation:

    • Western blot: Confirmed specificity in mouse kidney samples and NIH3T3 cells .

    • Immunohistochemistry: Verified in embryonic tissues (vibrissae, vertebrae) and ATL patient samples .

  • Epitope mapping:

    • Commercial antibodies target recombinant human EVC fusion proteins or peptide sequences .

Key Research Findings

Study FocusMethodologyOutcomeSource
EVC in ATLIHC, RT-PCREVC overexpression drives HH signaling
Post-translational modificationsCo-IP, Ubiquitination assaysSUMO3 enhances EFCAB7 binding; Ubiquitination reduces stability
Developmental rolesImmunofluorescenceEVC/LBN colocalization in embryonic heart

Technical Considerations

  • Storage: Stable at -20°C for 12 months; avoid freeze-thaw cycles .

  • Buffer: Phosphate-buffered saline with 50% glycerol .

  • Controls: Include plasmid-transfected cells (e.g., 293T) to confirm antibody specificity .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship the products within 1-3 business days after receiving your order. Delivery time may vary depending on the purchase method or location. Please consult your local distributor for specific delivery timelines.
Synonyms
EVCEllis-van Creveld syndrome protein antibody; DWF-1 antibody
Target Names
EVC
Uniprot No.

Target Background

Function
EVC is a critical component of the EvC complex, which plays a vital role in positively regulating ciliary Hedgehog (Hh) signaling. It is directly involved in endochondral growth and skeletal development.
Gene References Into Functions
  1. Whole exome sequencing (WES) in a family revealed two homozygous variants in EVC2 (c.30dupC; p.Thr11Hisfs*45) and TMC1 (c.1696-1G>A) genes. In another family, WES identified novel compound heterozygous variants (p.Ser307Pro, c.2894+3A>G) in the EVC gene. PMID: 29321360
  2. The molecular mechanism underlying the development of ventricular septal defect induced by the EVC c.343C>G mutation is likely due to a reduction in the anti-apoptotic and proliferative abilities of cardiomyocytes via downregulation of Hh pathway activity. PMID: 29257216
  3. Two Mexican families with patients diagnosed with Ellis-van Creveld syndrome were studied. In all cases, molecular analysis by Sanger sequencing identified the same homozygous mutation in exon 12 of EVC, c.1678G>T, resulting in a premature stop codon. PMID: 29229899
  4. Two novel nonsense mutations and a partial deletion of EVC/EVC2 were detected in two Vietnamese families with EvC. Additionally, a missense mutation of EFCAB7, a potential modifier gene in EvC and related disorders, was found in one family. PMID: 26748586
  5. Sequence analysis identified a novel nonsense mutation (p.Trp234*) in exon 8 of the EVC2 gene and a 15 bp duplication in exon 14 of the EVC gene in two families. PMID: 26580685
  6. Sequencing of both EVC and EVC2 identified two novel heterozygous splice site mutations c.384+5G>C in intron 3 and c.1465-1G>A in intron 10 in EVC, inherited from the mother and father, respectively. PMID: 26621368
  7. Epigenetically deregulated EVC appears to play a significant role in hedgehog activation. PMID: 24996003
  8. Molecular analysis of the EVC and EVC2 genes is valuable in genetic counseling for cases with prenatally detected postaxial polydactyly, thoracic narrowness, short limbs, and endocardial cushion defects. PMID: 21199751
  9. The first family from Pakistan with a large number of individuals affected by EVC was reported. DNA sequence analysis led to the identification of the fifth missense mutation in the EVC gene. PMID: 19744229
  10. STK32B and EVC yielded consistent evidence from cleft lip, with or without cleft palate, trios in all four populations. PMID: 20087401
  11. The expression of a Weyer variant, but not the expression of a truncated protein mimicking an Ellis-van Creveld syndrome mutation, impairs Hedgehog signal transduction in NIH 3T3 cells, consistent with its dominant effect. PMID: 19810119
  12. CRMP1 and EVC genes are located near WFS1, the Wolfram syndrome type 1 gene. PMID: 15492864
  13. In a consanguineous pedigree diagnosed with EvC and borderline intelligence, a 520-kb homozygous deletion comprising EVC, EVC2, C4orf6, and STK32B, caused by recombination between LINE-1 elements, was detected. PMID: 18454448
  14. EVC mutation is hypomorphic, and such mutations can cause a phenotype of cardiac and limb defects less severe than typical Ellis van Creveld syndrome. PMID: 18947413
  15. EVC and LBN play roles in cardiovascular development and disease. PMID: 19251731

Show More

Hide All

Database Links

HGNC: 3497

OMIM: 193530

KEGG: hsa:2121

STRING: 9606.ENSP00000264956

UniGene: Hs.646899

Involvement In Disease
Ellis-van Creveld syndrome (EVC); Acrofacial dysostosis, Weyers type (WAD)
Subcellular Location
Cell membrane; Single-pass membrane protein. Cytoplasm, cytoskeleton, cilium basal body. Cell projection, cilium. Cell projection, cilium membrane.
Tissue Specificity
Found in the developing vertebral bodies, ribs, upper and lower limbs, heart, kidney, lung.

Q&A

What are EVC antibodies and what are their primary research applications?

EVC antibodies refer to two distinct research contexts:

First, antibodies targeting the Ellis-van Creveld syndrome protein 2 (EVC2), which functions as a component of the EvC complex that positively regulates ciliary Hedgehog (Hh) signaling. EVC2 plays critical roles in bone formation and skeletal development and may be involved in early embryonic morphogenesis. These antibodies are primarily used in developmental biology and genetic disorder research .

Second, antibodies used in extracellular vesicle (EV) research, which are critical tools for the identification, characterization, and isolation of EVs and other extracellular nanoparticles (ENPs). EVs function as natural delivery vectors and mediators of biological signals across tissues .

Primary research applications include:

  • Western blot analysis for protein detection and characterization

  • Flow cytometry for single-vesicle analysis and high-throughput EV characterization

  • Immunohistochemistry to localize proteins in tissue samples

  • Sandwich assays for EV detection and quantification

  • Targeted therapy development, particularly in cancer research using antibody-displaying EVs

How do I select the appropriate EVC antibody for my research?

Selection of appropriate EVC antibodies requires consideration of multiple technical factors:

For EVC2 protein research:

  • Application compatibility: Verify the antibody has been validated for your specific application (WB, IHC-P, etc.)

  • Species reactivity: Ensure compatibility with your experimental model (human, mouse, etc.)

  • Clone type: Consider whether monoclonal (higher specificity) or polyclonal (broader epitope recognition) better suits your needs

  • Validation data: Review available data including predicted band size (148 kDa for EVC2) and positive control samples

For EV research:

  • Consult the EV Antibody Database (https://exrna.org/resources/evabdb/) to identify validated antibodies for your specific target and application

  • Review detailed information on antibody performance across different assay conditions

  • Consider both positive and negative results reported in the database

  • Examine antibodies specifically validated for your technique (western blot, flow cytometry, sandwich assays)

A systematic selection approach includes:

  • Define your target protein/marker and specific application requirements

  • Search specialized databases and literature for validated antibodies

  • Assess performance metrics in relevant applications and conditions

  • Evaluate both successful and unsuccessful applications to anticipate potential challenges

  • Select antibodies with comprehensive validation data matching your experimental conditions

What protocols are recommended for EVC antibody validation?

Validation of EVC antibodies requires multi-faceted approaches to ensure specificity and reproducibility:

For EVC2 protein antibodies:

  • Western blot validation using appropriate positive control tissues (human placenta tissue lysate at 40 μg)

  • Verification of band appearance at the predicted molecular weight (148 kDa)

  • Optimization of antibody dilution (e.g., 1/450 for ab198930) and exposure time (7 minutes)

  • Immunohistochemistry validation on relevant tissues (human colon cancer tissue) with optimized dilutions (1/40)

For EV antibodies:

  • Western blot: Confirm specificity for EV marker proteins with appropriate controls

  • Flow cytometry: Perform antibody titration to determine optimal concentration

  • Instrument calibration: Use standardized reference beads with appropriate fluorescent spectra

  • Spectral calibration: Generate proper compensation matrices for multiplexed measurements

  • Sandwich immunoassays: Validate antibody performance for both capture and detection roles

The EV Antibody Database provides standardized validation protocols for:

  • Western blot applications for EV markers

  • EV flow cytometry with detailed instrument settings

  • EV sandwich assays with comprehensive performance metrics

Researchers should document both positive and negative results during validation to establish the boundaries of antibody performance in specific experimental contexts.

How can EVC antibodies be engineered for targeted drug delivery systems?

Engineering EVC antibodies for targeted drug delivery involves sophisticated molecular design and optimization:

The development of antibody-displaying extracellular vesicles (Fc-EVs) represents a cutting-edge approach:

  • Engineering strategy:

    • Create fusion constructs combining Fc-binding domains with EV-sorting proteins

    • Systematically screen different combinations to identify optimal performance

    • Express fluorescent reporters (e.g., mNeonGreen) to track EV production and targeting

  • Optimization process:

    • Screen different EV-sorting domains: highest expression achieved with tetraspanins (CD9, CD63, CD81), annexin V, or TNFR

    • Evaluate Fc-binding domains: protein A and derived z domains showed superior antibody binding capacity

    • Test different antibody attachment methods to maximize display efficiency

  • Performance assessment:

    • Measure binding using imaging flow cytometry at single-vesicle resolution

    • Evaluate stability in biological fluids (plasma) at different time points (1, 10, 30 minutes)

    • Quantify targeting efficacy (e.g., 339-fold increase in uptake with trastuzumab-displaying EVs)

  • Therapeutic applications:

    • Target HER2-positive cancer cells using trastuzumab-displaying EVs

    • Direct EVs to PD-L1-expressing tumors using atezolizumab

    • Load EVs with therapeutic cargo (e.g., doxorubicin) while maintaining targeting specificity

This modular approach allows flexible targeting to virtually any tissue of interest by simply changing the displayed antibody, creating a versatile platform for precision medicine applications .

What are the methodological considerations for using EVC antibodies in flow cytometry?

Flow cytometry with EVC antibodies requires specialized methodological approaches:

  • Instrument preparation:

    • Calibrate using standardized reference beads for appropriate fluorescent spectra

    • Perform spectral calibration to ensure accurate signal measurement

    • Generate compensation matrices for multiplexed measurements with multiple antibodies

    • Set appropriate trigger thresholds to detect small particles while minimizing background

  • Sample preparation for EV analysis:

    • Isolate EVs using validated methods (ultracentrifugation, size exclusion chromatography)

    • Quantify EV concentration using complementary techniques

    • Prepare appropriate dilutions to avoid swarm detection artifacts

    • Include size calibration standards

  • Antibody optimization:

    • Titrate antibodies to determine optimal concentration for signal-to-noise ratio

    • Validate specificity using appropriate positive and negative controls

    • Assess antibody performance in the presence of biological matrices

  • Data analysis:

    • Implement consistent gating strategies based on size and marker expression

    • Report results as mean fluorescent intensity (MFI) for quantitative comparisons

    • Analyze targeting efficiency (e.g., fold increase in uptake compared to controls)

    • Perform multiparameter analysis to identify specific subpopulations

For antibody-displaying EVs, imaging flow cytometry allows detection of antibody binding at the single-vesicle level, providing higher resolution data on targeting efficiency and heterogeneity .

How do you troubleshoot non-specific binding in EVC antibody applications?

Non-specific binding represents a significant challenge in antibody applications. Systematic troubleshooting approaches include:

  • Comprehensive controls:

    • Include isotype controls matching the primary antibody's species and isotype

    • Use FMO (fluorescence minus one) controls for multiparameter flow cytometry

    • Test antibodies on samples lacking the target protein/marker

    • For antibody-displaying EVs, include EVs without antibody display and control antibodies not targeting your protein of interest

  • Blocking optimization:

    • Test different blocking agents (BSA, serum, commercial blockers)

    • Optimize blocking duration and temperature

    • Consider pre-adsorption of antibodies with relevant tissues/cells

  • Antibody concentration adjustment:

    • Perform detailed titration experiments to identify optimal concentrations

    • Balance signal intensity against background levels

    • Consider using higher antibody quality (affinity-purified) if persistent issues occur

  • Validation through competitive binding:

    • Pre-treat target cells with naked antibody to demonstrate specificity

    • For example, pre-treatment with trastuzumab should decrease uptake of trastuzumab-displaying EVs, confirming specificity of interaction

  • Buffer and washing optimization:

    • Test different buffer compositions to reduce non-specific interactions

    • Optimize washing steps (number, duration, buffer composition)

    • Consider additives to reduce hydrophobic interactions

When troubleshooting, adopt a systematic approach by changing one variable at a time and thoroughly documenting results to identify the source of non-specific binding.

How can EVC antibodies be effectively used in sandwich assays?

Sandwich assays provide a powerful approach for EV detection and characterization:

  • Assay design principles:

    • Select validated antibody pairs targeting EV surface proteins

    • Choose capture antibodies with high binding capacity and specificity

    • Select detection antibodies with minimal background and high signal generation

    • Consider using commercially validated ECL-based sandwich immunoassay systems

  • Protocol optimization:

    • Optimize coating concentration of capture antibody

    • Determine optimal blocking conditions to minimize background

    • Establish appropriate sample dilutions

    • Titrate detection antibody concentration

    • Optimize incubation times and washing procedures

  • Performance assessment:

    • Calculate signal-to-background ratios under different conditions

    • Determine detection limits and linear range

    • Evaluate reproducibility across multiple assays

    • Compare performance metrics of different antibody pairs

The EV Antibody Database provides detailed guidance on:

  • Capture antibody selection and optimization

  • Detector antibody performance metrics

  • Data interpretation and calculations for sandwich assays

  • Comparison of different antibody combinations for the same target

This information enables researchers to select optimal antibody pairs and assay conditions for specific EV populations and research questions.

What are the latest developments in antibody-displaying extracellular vesicles for cancer therapy?

Antibody-displaying extracellular vesicles represent a cutting-edge approach for targeted cancer therapy:

  • Engineering achievements:

    • Development of EVs decorated with antibody-binding moieties specific for the Fc domain

    • Creation of a modular system allowing decoration with different IgG antibodies

    • Optimization through systematic screening of Fc-binding and EV-sorting domains

    • Engineering stability in biological fluids for in vivo applications

  • Targeting capabilities:

    • HER2-targeted delivery: 339-fold increase in EV uptake by HER2-positive breast cancer cells when guided by trastuzumab

    • PD-L1-targeted delivery: Increased uptake in melanoma cells when guided by atezolizumab

    • Specific targeting demonstrated through competitive binding experiments

  • Therapeutic applications:

    • Loading with chemotherapeutic agents (doxorubicin)

    • Demonstration of lower tumor burden and extended survival in mouse melanoma models

    • Sustained and enriched accumulation in tumor tissue following systemic injection

  • Future directions:

    • Adaptation to display other Fc-fused proteins

    • Compatibility with bispecific antibodies for dual targeting

    • Integration with antibody-drug conjugates for enhanced therapeutic potential

    • Development of combination therapies targeting multiple cancer pathways

This technology combines the targeting precision of therapeutic antibodies with the natural delivery capabilities of EVs, potentially improving therapeutic outcomes while reducing off-target effects .

What are the best practices for western blot analysis using EVC antibodies?

Western blot analysis with EVC antibodies requires specific technical considerations:

For EVC2 protein detection:

  • Sample preparation:

    • Use appropriate positive control tissues (e.g., human placenta)

    • Prepare consistent protein amounts (40 μg recommended)

    • Include molecular weight markers covering the expected range (148 kDa for EVC2)

  • Gel electrophoresis:

    • Use appropriate percentage gels for the target protein size (6% SDS-PAGE recommended for EVC2)

    • Run at consistent voltage following standardized protocols

    • Ensure complete protein transfer to membrane

  • Antibody incubation:

    • Use optimized dilutions (1/450 for ab198930)

    • Maintain consistent incubation times and temperatures

    • Implement thorough washing between steps to reduce background

  • Detection and analysis:

    • Use appropriate exposure times (7 minutes recommended for EVC2)

    • Verify band appearance at expected molecular weight

    • Include appropriate loading controls

    • Consider quantitative analysis when comparing samples

For EV marker antibodies:

  • Sample considerations:

    • Isolate EVs using consistent, validated methods

    • Compare with whole cell lysates as controls

    • Normalize loading based on particle number or protein concentration

  • Optimization strategies:

    • Titrate antibody concentration for optimal signal-to-noise ratio

    • Adjust exposure times based on signal intensity

    • Validate specificity through appropriate controls

The EV Antibody Database contains detailed protocols and validation data for western blot analysis of various EV markers, providing valuable reference for optimization .

How do you quantitatively assess targeting efficiency of antibody-displaying EVs?

Quantitative assessment of targeting efficiency requires multiple complementary approaches:

  • In vitro targeting metrics:

    • Fold increase in uptake: Calculate the ratio of target cell uptake between antibody-displaying EVs and control EVs (e.g., 339-fold increase observed with trastuzumab-guided EVs in HER2+ cells)

    • Mean fluorescent intensity (MFI): Measure fluorescence intensity of cells after incubation with labeled EVs

    • Competitive binding analysis: Quantify decrease in uptake after pre-treatment with naked antibody

    • Time-course analysis: Measure uptake kinetics to determine optimal incubation periods

  • Flow cytometry quantification:

    • Single-vesicle analysis: Use imaging flow cytometry to detect antibody binding at individual EV level

    • Population statistics: Calculate percentage of EVs displaying antibodies

    • Binding capacity: Determine antibody density on EV surface

  • In vivo targeting assessment:

    • Biodistribution analysis: Measure accumulation in target vs. non-target tissues

    • Pharmacokinetic profile: Determine circulation half-life and clearance rates

    • Tumor accumulation: Quantify sustained enrichment in tumor tissue following systemic injection

  • Functional endpoints:

    • Therapeutic cargo delivery: Measure drug concentration in target tissues

    • Biological effects: Assess tumor burden reduction

    • Survival analysis: Evaluate extension of survival in animal models

Standardization is critical - researchers should normalize data appropriately, include relevant controls, and apply consistent analysis methods across experiments to enable meaningful comparisons between different targeting strategies.

What statistical approaches are recommended for analyzing EVC antibody performance data?

Statistical analysis of EVC antibody performance requires rigorous approaches appropriate for the specific application:

  • Western blot quantification:

    • Normalization to loading controls

    • Multiple technical and biological replicates (minimum n=3)

    • Paired statistical tests when comparing treatments on the same samples

    • Non-parametric tests when normality cannot be assumed

  • Flow cytometry analysis:

    • Minimum event counts to ensure statistical power

    • Comparison of mean/median fluorescence intensity (MFI)

    • Population analysis through clustering algorithms

    • Variance analysis across technical and biological replicates

  • Targeting efficiency statistics:

    • Calculate fold-increase in uptake with 95% confidence intervals

    • Use appropriate statistical tests to compare targeting between different antibodies

    • Account for multiple comparisons when screening multiple antibodies

    • Regression analysis for dose-dependent effects

  • In vivo studies:

    • Power analysis to determine appropriate sample sizes

    • Survival analysis using Kaplan-Meier curves and log-rank tests

    • Area under the curve (AUC) analysis for pharmacokinetic/biodistribution data

    • Mixed-effects models for longitudinal data

  • Validation metrics:

    • Sensitivity and specificity calculations

    • Receiver operating characteristic (ROC) curve analysis

    • Concordance metrics between different detection methods

    • Inter-assay and intra-assay coefficient of variation

When reporting results, researchers should clearly state statistical methods, sample sizes, replicate structure, and significance thresholds to enable reproducibility and proper interpretation.

How are EVC antibodies contributing to advancements in precision medicine?

EVC antibodies are driving significant advancements in precision medicine through multiple innovative approaches:

  • Targeted cancer therapies:

    • Development of antibody-displaying EVs for precise delivery of therapeutics to specific cancer types

    • Targeting of clinically relevant markers like HER2 and PD-L1 with remarkable specificity

    • Creation of modular platforms that can be adapted to various tumor types by simply changing the displayed antibody

    • Potential for synergistic effects by combining targeting antibodies with therapeutic cargo

  • Personalized therapeutic approaches:

    • EVs decorated with patient-specific antibodies targeting unique tumor markers

    • Flexibility to combine multiple antibodies for multitargeted approaches

    • Compatibility with various therapeutic modalities including chemotherapeutics, biologics, and gene therapy agents

  • Improved treatment efficacy:

    • Enhanced delivery of therapeutic agents to target tissues (339-fold increase in uptake demonstrated)

    • Reduced off-target effects through precise targeting

    • Lower tumor burden and extended survival demonstrated in preclinical models

    • Potential to improve response rates to immunotherapy, which currently benefits only a subset of patients

  • Future clinical applications:

    • Potential adaptation to display bispecific antibodies for dual targeting

    • Integration with antibody-drug conjugates for enhanced therapeutic potential

    • Development of multifunctional therapeutic platforms

    • Applications beyond cancer to other diseases requiring targeted intervention

The EV Antibody Database further supports these advances by providing validated resources for EV research, facilitating more rapid translation of basic discoveries into clinical applications .

What are the current technical limitations of EVC antibody research and potential solutions?

Despite significant advances, EVC antibody research faces several technical challenges:

  • Standardization challenges:

    • Variability in EV isolation methods affecting antibody performance

    • Lack of universally accepted reference standards

    • Inconsistent reporting of methods and validation data

    Potential solutions:

    • Development of standardized EV preparations for antibody validation

    • Expansion of resources like the EV Antibody Database to include more comprehensive validation data

    • Implementation of minimum information reporting requirements

  • Targeting efficiency limitations:

    • Non-specific binding reducing targeting precision

    • Competitive binding in complex biological environments

    • Variability in target expression across patient samples

    Potential solutions:

    • Optimization of Fc-binding domains through systematic screening

    • Development of higher-affinity targeting antibodies

    • Implementation of dual-targeting strategies

  • Manufacturing challenges:

    • Consistency in EV production and antibody display

    • Scalability limitations for clinical applications

    • Stability concerns in storage and administration

    Potential solutions:

    • Development of stable cell lines with consistent EV production

    • Optimization of storage conditions to maintain antibody display

    • Creation of standardized manufacturing protocols

  • In vivo performance challenges:

    • Immune clearance of antibody-displaying EVs

    • Limited tissue penetration

    • Competitive binding with endogenous antibodies

    Potential solutions:

    • Engineering EVs to evade immune recognition

    • Optimization of antibody density on EV surface

    • Development of administration strategies to enhance tissue delivery

Addressing these limitations requires collaborative efforts across multiple disciplines, including antibody engineering, EV biology, drug delivery, and clinical translation, supported by resources like the EV Antibody Database to facilitate standardization and method optimization .

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.