ENO2 Human, His refers to recombinant human neuronal enolase (Enolase 2) fused with a hexahistidine (His) tag at the N-terminus. This engineered protein is widely used in biochemical and molecular biology research to study its enzymatic, structural, and functional roles in cellular metabolism, neuroprotection, and disease mechanisms.
ENO2 Human, His catalyzes the reversible conversion of 2-phosphoglycerate to phosphoenolpyruvate in glycolysis, a critical step for ATP production . Its enzymatic site spans most of the protein length, enabling specificity for magnesium-dependent catalysis .
Beyond glycolysis, ENO2 exhibits diverse roles:
Neurotrophic Activity: Binds calcium-dependently to CNS neurons, promoting survival and neurite outgrowth .
Cell Surface Plasminogen Binding: Facilitates extracellular matrix remodeling .
Gene Regulation: Interacts with Hsp70 and VDAC1 to stabilize mitochondrial membrane potential .
ENO2 is primarily cytosolic but can translocate to the nucleus or cell membrane, depending on cellular stress or developmental cues .
ENO2 is upregulated during human cytomegalovirus (HCMV) infection, supporting viral replication by enhancing glycolysis and glucose consumption . Targeting ENO2 with inhibitors (e.g., POMHEX) reduces viral titers by ~85-fold .
ENO2 is a diagnostic biomarker for neurodegenerative diseases (e.g., Creutzfeldt-Jakob disease) and neuroendocrine tumors . Its neuroprotective effects are linked to calcium-dependent binding to neocortical neurons .
The Human ENO2 ELISA Kit (Proteintech, Cat. KE00050) quantifies ENO2 levels in serum, plasma, or cell culture supernatants:
Parameter | Value |
---|---|
Sensitivity | 0.11 ng/mL |
Dynamic Range | 0.313–20 ng/mL |
Reactivity | Human |
Prognostic Value: High ENO2 expression in clear cell renal cell carcinoma (ccRCC) correlates with poor survival and disease-free intervals .
Diagnostic Accuracy: ROC analysis of ENO2 in ccRCC tissues shows an AUC of 0.679 for 10-year survival prediction .
ENO2, also known as gamma-enolase or neuron-specific enolase (NSE), is an enzyme encoded by the ENO2 gene in humans. It functions as a phosphopyruvate hydratase, catalyzing the conversion of 2-phosphoglycerate to phosphoenolpyruvate in the glycolytic pathway . ENO2 is one of three enolase isoenzymes found in mammals, existing as a homodimer primarily in mature neurons and cells of neuronal origin . During neural development, a switch occurs from alpha enolase to gamma enolase in neural tissue of rats and primates . Beyond its metabolic function, ENO2 has emerged as a significant biomarker for neuroendocrine tumors and certain neurological conditions, making it valuable in both research and clinical contexts .
Distinguishing between the three enolase isoforms (alpha, beta, and gamma) requires specific methodological approaches. While alpha-isoform is expressed in most tissues and beta-form predominantly in muscle tissue, gamma-enolase (ENO2) is found specifically in nervous tissue . For precise isoform identification, researchers should employ:
Antibody-based methods: Using monoclonal antibodies with confirmed specificity for ENO2, such as the ENO2/1462 clone, which has been validated against more than 19,000 full-length human proteins via HuProt™ Array .
Expression analysis: qRT-PCR with isoform-specific primers can quantify ENO2 mRNA levels relative to other enolases .
Protein characterization: Western blotting with antibodies that recognize the ~50 kDa ENO2 protein specifically .
Functional assays: Measuring enzymatic activity with substrates that have differential affinity for the isoforms.
When studying mixed tissue samples, a combination of these approaches provides the most reliable differentiation between enolase isoforms.
For precise quantification of ENO2 in research samples, several validated methodological approaches exist:
Enzyme-Linked Immunosorbent Assay (ELISA): Sandwich ELISA kits for human ENO2 offer high sensitivity (0.11 ng/mL) and a detection range of 0.313-20 ng/mL. These assays utilize antibodies specific for ENO2 pre-coated onto microwells, followed by detection with HRP-conjugated antibodies and TMB development . The color intensity, measurable at 450 nm with correction at 630 nm, is proportional to the quantity of bound protein .
Immunohistochemistry: For tissue sections, particularly paraffin-embedded samples, monoclonal antibodies such as ENO2/1462 allow for visualization of ENO2 in its cellular context .
Western Blotting: For protein extract analysis, antibodies recognizing the ~50 kDa ENO2 protein provide semi-quantitative measurement .
qRT-PCR: For mRNA expression analysis, especially when protein is difficult to extract.
Performance metrics for ELISA quantification demonstrate high reliability with recovery rates averaging 87% in human plasma samples and 90% in cell culture supernatants . Intra-assay and inter-assay coefficient of variation (CV) values typically fall below 8%, indicating excellent reproducibility, as shown in the following tables:
Intra-Assay Performance:
Sample | n | mean (ng/mL) | SD | CV% |
---|---|---|---|---|
1 | 20 | 2.3 | 0.1 | 6.0 |
2 | 20 | 7.6 | 0.6 | 7.8 |
3 | 20 | 16.1 | 0.9 | 5.5 |
Inter-Assay Performance:
Sample | n | mean (ng/mL) | SD | CV% |
---|---|---|---|---|
1 | 24 | 2.3 | 0.1 | 5.7 |
2 | 24 | 7.4 | 0.4 | 5.6 |
3 | 24 | 12.9 | 0.8 | 5.8 |
ENO2 plays a critical role in cancer metabolic reprogramming through its function in glycolysis. Research indicates that ENO2 promotes the Warburg effect—a metabolic shift where cancer cells preferentially utilize glycolysis even in the presence of oxygen . By catalyzing the conversion of 2-phosphoglycerate to phosphoenolpyruvate, ENO2 facilitates this aberrant metabolic pathway .
The metabolic impact of ENO2 in cancer cells manifests in several measurable ways:
Increased glucose uptake: ENO2 overexpression correlates with enhanced glucose consumption by tumor cells.
Elevated lactate production: Higher ENO2 levels result in increased lactate secretion, a hallmark of the Warburg effect.
Accelerated tumor growth: The metabolic advantage conferred by ENO2 supports rapid proliferation of cancer cells.
ENO2 upregulation has been documented in multiple cancer types, including head and neck cancer, colorectal cancer, pancreatic cancer, and bladder cancer, consistently associating with more aggressive phenotypes . In clear cell renal cell carcinoma (ccRCC), ENO2 has been shown to promote cancer progression by enhancing cellular glycolysis . Experimental approaches to study this phenomenon include genetic manipulation of ENO2 expression (knockdown/overexpression), metabolic flux analysis, and correlation studies between ENO2 levels and glycolytic markers.
The relationship between hypoxia-inducible factor-1 alpha (HIF-1α) and ENO2 represents a crucial regulatory mechanism in cancer biology, particularly in clear cell renal cell carcinoma (ccRCC). Research has established that ENO2 is regulated by HIF-1α through direct transcriptional activation .
Mechanistically, HIF-1α binds to hypoxia-response elements (HREs) within the ENO2 promoter region. Chromatin immunoprecipitation (ChIP) experiments in A498 cells have confirmed HIF-1α binding to three putative binding sites within the proximal 1000 bp region of the ENO2 promoter . The significance of this regulatory relationship is demonstrated through both molecular and functional experiments:
Genetic manipulation: Knockdown of HIF-1α using siRNA results in decreased ENO2 protein and mRNA levels .
Promoter analysis: The ENO2 promoter contains the consensus 5′-(A/G)CGTG-3′ HRE sequences that HIF-1α recognizes .
Functional consequences: HIF-1α-mediated upregulation of ENO2 contributes to the metabolic adaptation of cancer cells to hypoxic conditions, supporting their survival and growth.
For researchers investigating this relationship, recommended experimental approaches include:
Use of hypoxia chambers to simulate physiological hypoxia
Application of HIF stabilizers (such as cobalt chloride or deferoxamine) to mimic hypoxic signaling
Construction of luciferase reporter assays with wild-type and mutated HRE sites
Comprehensive analysis combining ChIP, qPCR, and western blotting to establish the regulatory relationship
This HIF-1α-ENO2 axis represents a potential therapeutic target, as disrupting this regulatory mechanism could impair cancer cell adaptation to hypoxic microenvironments.
When designing ENO2 knockout or knockdown experiments, researchers should consider several methodological aspects to ensure valid and interpretable results:
Selection of appropriate model systems:
Knockdown/knockout strategy:
siRNA approach: Multiple siRNA sequences should be tested to identify those with highest knockdown efficiency and specificity .
CRISPR-Cas9: Guide RNA design should minimize off-target effects while maximizing on-target efficiency.
Inducible systems: For genes essential for cell survival, inducible knockdown/knockout systems may be necessary.
Validation of ENO2 reduction:
Controls:
Negative controls: Non-targeting siRNA or scrambled sequences.
Rescue experiments: Reintroduction of ENO2 to confirm phenotype specificity.
Isoform specificity: Verify that other enolase isoforms (ENO1, ENO3) are not affected.
Phenotypic analysis:
Timing considerations:
Acute vs. chronic effects: Short-term knockdown may reveal immediate effects, while stable knockout might show compensatory mechanisms.
Cell cycle synchronization: For investigating cell cycle-dependent effects.
Data interpretation:
Compensatory mechanisms: Consider potential upregulation of other enolase isoforms.
Off-target effects: Validate key findings with alternative knockdown methods.
Dosage effects: Partial vs. complete knockdown may yield different results.
Following these methodological considerations will enhance the robustness and reliability of ENO2 functional studies.
ENO2 serves as a valuable biomarker in both neurological and oncological research contexts. Its effective utilization requires understanding of appropriate methodologies, reference ranges, and interpretation frameworks.
Neurological applications:
ENO2 (NSE) functions as a marker of neuronal damage and can be measured in cerebrospinal fluid (CSF) and serum. For research applications:
Traumatic brain injury models: Monitor ENO2 levels as an indicator of neuronal damage severity.
Neurodegenerative disease studies: Track ENO2 as a potential biomarker of disease progression.
Ischemic models: Use ENO2 elevation as a measure of neuronal stress and damage.
Oncological applications:
ENO2 serves as a neuroendocrine marker and is particularly useful in:
Tumor classification: ENO2 positivity helps identify tumors of neuroendocrine origin .
Prognostic studies: Higher ENO2 levels correlate with more aggressive tumor phenotypes .
Treatment response evaluation: Changes in ENO2 expression can indicate therapeutic efficacy.
Methodological approaches:
For optimal biomarker utilization, researchers should consider:
Sample selection and preparation:
Serum/plasma: Collection timing is critical, with standardized processing protocols.
Tissue: Proper fixation and processing for immunohistochemistry.
CSF: Appropriate collection and storage to prevent enzymatic degradation.
Detection methods:
Interpretation guidelines:
By implementing these methodological approaches, researchers can maximize the utility of ENO2 as a biomarker in their experimental systems.
Researchers face several challenges when comparing ENO2 data across different detection platforms, which can impact data interpretation and reproducibility. Understanding these challenges is essential for accurate cross-study comparisons:
Antibody specificity variations:
Different antibody clones may recognize distinct epitopes of ENO2, potentially leading to variations in detection sensitivity. For instance, some antibodies might preferentially detect specific conformational states or post-translational modifications of ENO2 .
Detection range and sensitivity differences:
ELISA kits may have different lower limits of detection and quantifiable ranges. The Proteintech Human ENO2 ELISA Kit has a sensitivity of 0.11 ng/mL with a range of 0.313-20 ng/mL , while other platforms might offer different specifications, complicating direct comparison of absolute values.
Sample preparation variability:
Differences in extraction methods, buffer compositions, and processing steps can affect ENO2 recovery rates. Recovery studies show that even optimized protocols achieve 87% recovery from human plasma and 90% from cell culture supernatants , indicating some inherent variability.
Matrix effects:
The biological matrix (serum, plasma, cell lysate, tissue extract) can influence assay performance. Components in complex biological samples may cause interference, leading to platform-specific biases.
Standardization issues:
Lack of universal calibrators or reference materials for ENO2 means that absolute values may not be directly comparable between different assay platforms.
Cross-reactivity considerations:
Some detection methods may exhibit cross-reactivity with other enolase isoforms (ENO1, ENO3), particularly in assays using polyclonal antibodies.
To address these challenges, researchers should:
Include appropriate internal controls and standards across experiments
Perform parallel analyses with multiple detection methods when possible
Report detailed methodological information including antibody clones, detection limits, and assay conditions
Consider relative changes rather than absolute values when comparing across platforms
Validate key findings using orthogonal detection methods
By acknowledging these limitations and implementing appropriate controls, researchers can make more reliable comparisons of ENO2 data across different experimental platforms and studies.
Several promising research directions involving ENO2 merit deeper investigation in the precision medicine field:
Liquid biopsy development:
The quantification of ENO2 in blood samples could serve as a minimally invasive method for monitoring neurological damage or neuroendocrine tumor progression. Given the established sensitivity of current ELISA methods (0.11 ng/mL) , research should focus on developing even more sensitive detection platforms for early disease detection.
Therapeutic targeting:
The role of ENO2 in glycolysis and its regulation by HIF-1α suggests potential for targeted therapy development . Research into small molecule inhibitors specific to ENO2 (versus other enolase isoforms) could yield novel therapeutic approaches for neuroendocrine tumors and other cancers with ENO2 overexpression.
Biomarker panels:
Investigation of ENO2 in combination with other markers could improve diagnostic accuracy. The established practice of using ENO2 alongside Synaptophysin, Chromogranin A, and Neurofilament for neuroendocrine tumors suggests that optimized biomarker panels could be developed for various neurological and oncological conditions.
Metabolic profiling:
Further research into how ENO2 expression patterns correlate with broader metabolic profiles could yield insights into disease mechanisms and potential therapeutic vulnerabilities, particularly in cancers demonstrating the Warburg effect .
Imaging applications:
Development of ENO2-targeted imaging agents could enhance visualization of neuronal damage or neuroendocrine tumors. Research into radiolabeled antibodies or small molecules targeting ENO2 could advance diagnostic imaging capabilities.
These research directions represent significant opportunities for translating our current understanding of ENO2 biology into clinically relevant applications. Methodological approaches combining genomics, proteomics, and metabolomics will be essential for realizing the full potential of ENO2 in precision medicine.
Advances in structural biology offer promising avenues for developing ENO2-specific inhibitors with potential therapeutic applications. The distinctive structural features of ENO2 compared to other enolase isoforms provide opportunities for selective targeting that minimizes off-target effects.
Future research in this area should focus on:
High-resolution structural analysis of ENO2 active sites and binding pockets using X-ray crystallography and cryo-electron microscopy, particularly comparing structures of ENO2 with ENO1 and ENO3 to identify isoform-specific features.
Structure-based virtual screening to identify lead compounds with preferential binding to ENO2 over other enolase isoforms, followed by medicinal chemistry optimization.
Development of allosteric inhibitors that target ENO2-specific regulatory sites rather than the catalytic site that may be more conserved across enolase isoforms.
Investigation of the structural basis for ENO2's role in protein-protein interactions beyond its enzymatic function, potentially revealing additional targetable interfaces.
Analysis of post-translational modifications specific to ENO2 that might affect protein function and offer additional targeting opportunities.
The successful development of ENO2-specific inhibitors could provide valuable tools for both research applications and potential therapeutic interventions in conditions where ENO2 dysregulation contributes to pathology.
The recombinant human Enolase-2 protein is typically produced in E. coli and includes an N-terminal His-tag for purification purposes. The protein sequence ranges from Met1 to Leu434, with an additional N-terminal Met and a 6-His tag . The predicted molecular mass of this recombinant protein is approximately 48 kDa, although it may appear as 46 kDa under reducing conditions in SDS-PAGE .
Enolase-2 is highly active in converting phosphoglyceric acid to phosphoenolpyruvate, with a specific activity greater than 6,000 pmol/min/μg under the described conditions . This enzyme is crucial for energy production in neurons and is often used as a marker for neuronal damage and neuroendocrine tumors .
Recombinant Enolase-2 is widely used in research for various applications, including:
The recombinant Enolase-2 protein is supplied as a 0.2 μm filtered solution in MES, NaCl, KCl, and MgSO₄. It should be stored at -20 to -70°C to maintain stability and avoid repeated freeze-thaw cycles . Under sterile conditions, it remains stable for up to 6 months from the date of receipt and 3 months after opening .