The ENO3 antibody is a specialized immunological tool targeting the beta-enolase (ENO3) protein, encoded by the ENO3 gene. This antibody is widely used in research to study ENO3's role in glycolysis, muscle development, and diseases such as metabolic myopathies and cancers . ENO3, a glycolytic enzyme, catalyzes the conversion of 2-phosphoglycerate to phosphoenolpyruvate and is predominantly expressed in skeletal and cardiac muscles . Antibodies against ENO3 enable precise detection, quantification, and localization of the protein in experimental models, making them critical for understanding its biological and pathological functions.
ENO3 antibodies have been instrumental in uncovering the protein’s dual role in tumor progression:
Tumor Suppression in HCC: Overexpression of ENO3 in hepatocellular carcinoma (HCC) cells suppresses proliferation, migration, and invasion by inhibiting the Wnt/β-catenin pathway . Studies using ENO3-specific antibodies demonstrated reduced epithelial-mesenchymal transition (EMT) markers, such as E-cadherin upregulation and N-cadherin downregulation .
Oncogenic Role in ccRCC: In clear cell renal cell carcinoma (ccRCC), ENO3 promotes the Warburg effect (aerobic glycolysis) by stabilizing mRNA via NSUN5-mediated 5-methylcytosine modification. Knockdown of ENO3 using siRNA reduced glucose uptake and lactate production, highlighting its metabolic role .
Glycogen Storage Disease XIII: ENO3 antibodies aid in diagnosing this rare myopathy caused by ENO3 mutations. Immunoblotting reveals reduced ENO3 expression in muscle biopsies of affected individuals .
Non-Alcoholic Steatohepatitis (NASH): In murine NASH models, ENO3 antibodies detected elevated protein levels correlated with ferroptosis suppression and lipid accumulation .
ENO3 antibodies undergo rigorous validation:
Western Blot: Recognizes bands at 42–47 kDa in human skeletal muscle, liver (HepG2), and mouse brain tissues .
Immunohistochemistry: Strong staining in human heart and skeletal muscle sections, with antigen retrieval optimizing signal clarity .
Knockout/Knockdown Validation: Specificity confirmed using ENO3-deficient cell lines .
ENO3 antibodies are pivotal in developing targeted therapies:
Diagnostic Biomarker: Low ENO3 expression in HCC correlates with poor prognosis, making it a potential biomarker for patient stratification .
Therapeutic Target: In cancers with dysregulated glycolysis, ENO3 inhibition via monoclonal antibodies or siRNA shows promise in preclinical models .
Research priorities include:
ENO3 (Enolase 3, beta, muscle) encodes the β-subunit of enolase, which is distributed across various tissues including liver, lung, skeletal muscle, and heart. This protein plays crucial roles in both glycogen and cholesterol metabolism . The functional protein has a calculated molecular weight of 47 kDa (434 amino acids) but is typically observed at 42-47 kDa in experimental conditions . ENO3 is particularly important in metabolic processes, as its deficiency has been linked to metabolic myopathies . Additionally, ENO3 accelerates hepatic cholesterol ester accumulation through the mediation of cholesteryl ester generation .
ENO3 antibodies are versatile research tools validated for multiple applications including:
When selecting an ENO3 antibody, researchers should consider the specific experimental requirements and validate the antibody in their particular system, as optimal dilutions may vary based on the sample type and detection method employed .
The choice between monoclonal and polyclonal ENO3 antibodies depends on your experimental goals:
Monoclonal ENO3 Antibodies:
Offer high specificity to a single epitope (e.g., mouse monoclonal IgG2b)
Provide consistent lot-to-lot reproducibility
Ideal for applications requiring high specificity and minimal background
Available options include those reactive to specific regions (e.g., AA 228-277 or other defined sequences)
Polyclonal ENO3 Antibodies:
Often provide stronger signals by binding multiple sites
Typically generated in rabbits against recombinant fusion proteins
Can be more forgiving in applications where protein conformation may vary
For novel research applications, testing both antibody types may help determine which provides optimal results for your specific experimental conditions.
To maintain ENO3 antibody integrity and functionality, proper storage is essential:
Antibodies are typically supplied in PBS with 0.02% sodium azide and 50% glycerol at pH 7.3
Most are stable for one year after shipment when stored properly
Avoid repeated freeze/thaw cycles that can degrade antibody quality
For antibodies provided in small volumes (e.g., 20μl), some formulations contain 0.1% BSA for stabilization
Aliquoting is generally recommended for antibodies without glycerol, but may be unnecessary for those in 50% glycerol storage buffer
Following these storage guidelines will help ensure experimental reproducibility and extend the usable life of your ENO3 antibodies.
When selecting an ENO3 antibody, species cross-reactivity is a critical consideration:
For comparative studies across species, select antibodies with validated cross-reactivity for all target organisms. Western blot validation has confirmed ENO3 antibody detection in various samples including HeLa cells, HEK-293 cells, HepG2 cells, Jurkat cells, and skeletal muscle tissue from multiple species . Always perform preliminary validation tests when using antibodies in species not explicitly listed in the manufacturer's documentation.
Rigorous antibody validation is essential for generating reliable research data. For ENO3 antibodies, consider this comprehensive validation approach:
Positive and negative controls:
Cross-validation with multiple techniques:
Peptide competition assay:
Pre-incubate the antibody with immunizing peptide
Compare signal reduction in samples with and without peptide blocking
Orthogonal validation:
Correlate protein detection with mRNA expression data
Use mass spectrometry to confirm protein identity in immunoprecipitated samples
Reproducibility assessment:
Test antibody performance across multiple batches of samples
Evaluate consistency across different lots of the antibody if possible
Document all validation steps thoroughly as supporting evidence for publications and maintain validated antibodies under optimal storage conditions to ensure consistent performance.
Recent research has implicated ENO3 in cancer metabolism, particularly through its relationship with the Warburg effect. When designing experiments to investigate this connection:
Expression correlation studies:
Metabolic pathway analysis:
Clear cell renal cell carcinoma (ccRCC) studies:
Post-translational modification examination:
Functional studies:
Combine antibody-based detection with knockdown/overexpression experiments
Assess changes in cancer cell metabolism, proliferation, and migration upon modulation of ENO3 levels
This methodological approach enables comprehensive investigation of ENO3's role in cancer metabolism while maximizing the utility of ENO3 antibodies in uncovering novel biological insights.
ENO3 deficiency has been linked to metabolic myopathies , making it an important research target. To optimize antibody-based investigations:
Tissue-specific analysis protocol:
Use skeletal muscle biopsies from patients with suspected metabolic myopathies
Compare with appropriate control samples from healthy individuals
Employ both frozen and paraffin-embedded tissue preparations for comprehensive analysis
Staining optimization for muscle tissue:
Quantitative analysis approaches:
Employ western blotting with gradient gels to resolve potential ENO3 isoforms
Consider using automated image analysis software for quantification
Normalize ENO3 expression to appropriate housekeeping proteins specific to muscle tissue
Physiological correlation:
Correlate ENO3 antibody staining patterns with clinical parameters
Document glycogen accumulation using PAS staining in parallel sections
Assess mitochondrial function markers to understand the metabolic consequences
Genetic correlation:
Combine ENO3 protein analysis with genotyping for known ENO3 mutations
Investigate protein expression patterns in carriers versus affected individuals
This methodological framework enables researchers to effectively use ENO3 antibodies when investigating the molecular basis of metabolic myopathies.
When encountering inconsistent results with ENO3 antibodies, follow this systematic troubleshooting approach:
Sample preparation issues:
Ensure complete protein denaturation for western blotting
Verify protein extraction efficiency from different tissue types
For skeletal muscle samples, pay special attention to myofibrillar protein extraction protocols
Antibody-specific optimization:
Detection system verification:
Evaluate secondary antibody specificity
For fluorescence-based detection, check for autofluorescence in muscle samples
Optimize exposure times or gain settings to prevent signal saturation
Technical controls:
Include recombinant ENO3 protein as a positive control
Use loading controls appropriate for the experimental context
Perform parallel experiments with alternative antibodies targeting different ENO3 epitopes
Documentation and standardization:
Record all experimental conditions in detail
Standardize protocols across experiments
Consider developing a laboratory-specific validated protocol once optimal conditions are determined
By systematically addressing these factors, researchers can improve the consistency and reliability of experiments using ENO3 antibodies.
Investigating post-translational modifications (PTMs) of ENO3 requires specialized approaches:
Identification of potential PTM sites:
Modification-specific detection strategies:
Use antibodies raised against specific ENO3 PTMs when available
Employ two-dimensional gel electrophoresis to separate modified forms
Consider enrichment strategies for phosphorylated or acetylated proteins
Immunoprecipitation-based approaches:
Use ENO3 antibodies for immunoprecipitation followed by:
Western blotting with modification-specific antibodies
Mass spectrometry for comprehensive PTM profiling
Functional assays to determine effects on enzymatic activity
Contextual analysis:
Compare PTM patterns across different physiological conditions
Investigate tissue-specific modification patterns
Assess changes in PTMs during disease progression, particularly in cancer models
Functional correlation:
Correlate PTM changes with ENO3 enzymatic activity
Investigate how modifications affect protein-protein interactions
Determine if modifications alter subcellular localization
This methodological framework enables researchers to leverage ENO3 antibodies for detailed investigation of post-translational regulation mechanisms that may be critical in both normal physiology and disease states.