ENO2 Human

Enolase-2 Human Recombinant
Shipped with Ice Packs
In Stock

Description

ENO2 Human Recombinant expressed in E. coli contains 434 amino acids and its Mw is 47 kDa.
The Enolase-2 is purified by proprietary chromatographic techniques.

Product Specs

Introduction
Neuron-specific enolase (NSE), also known as Enolase-2, is a glycolytic isoenzyme primarily found in central and peripheral neurons and neuroendocrine cells. NSE is released into the cerebrospinal fluid (CSF) following neural tissue injury. Additionally, neoplasms originating from neural or neuroendocrine tissue release NSE into the bloodstream. NSE serves as a valuable biomarker for detecting and monitoring various tumors, including neuroblastoma, small cell lung cancer, medullary thyroid cancer, carcinoid tumors, pancreatic endocrine tumors, and melanoma. As one of the three enolase isoenzymes present in mammals, NSE is specifically found in mature neurons and cells of neuronal origin. During the development of neural tissue in rats and primates, a transition from alpha enolase to gamma enolase (NSE) takes place.
Description
Recombinant human ENO2, expressed in E. coli, consists of 434 amino acids and exhibits a molecular weight of 47 kDa. The purification of Enolase-2 is achieved using proprietary chromatographic techniques.
Physical Appearance
Clear, sterile-filtered solution.
Formulation
Enolase 2 is provided in a buffer solution containing 20mM Tris (pH 7.5), 0.1M KCl, and 5mM MgSO4.
Stability
For short-term storage (1-2 weeks), the product should be kept at 4°C. For extended storage, it is recommended to freeze the product at -20°C. Repeated freeze-thaw cycles should be avoided.
Purity
The purity of the product is greater than 95%, as determined by SDS-PAGE analysis.
Synonyms
Gamma-enolase, EC 4.2.1.11, 2-phospho-D-glycerate hydro-lyase, Neural enolase, Neuron-specific enolase, NSE, Enolase 2, ENO2.
Source
Escherichia Coli.
Amino Acid Sequence
MSIEKIWARE ILDSRGNPTV EVDLYTAKGL FRAAVPSGAS TGIYEALELR DGDKQRYLGK GVLKAVDHIN STIAPALISS GLSVVEQEKL DNLMLELDGT ENKSKFGANA ILGVSLAVCK AGAAERELPL YRHIAQLAGN SDLILPVPAF NVINGGSHAG NKLAMQEFMI LPVGAESFRD AMRLGAEVYH TLKGVIKDKY GKDATNVGDE GGFAPNILEN SEALELVKEA IDKAGYTEKI VIGMDVAASE FYRDGKYDLD FKSPTDPSRY ITGDQLGALY QDFVRDYPVV SIEDPFDQDD WAAWSKFTAN VGIQIVGDDL TVTNPKRIER AVEEKACNCL LLKVNQIGSV TEAIQACKLA QENGWGVMVS HRSGETEDTF IADLVVGLCT GQIKTGAPCR SERLAKYNQL MRIEEELGDE ARFAGHNFRN PSVL.

Q&A

What is ENO2 and what are its alternative designations in scientific literature?

ENO2, also known as Enolase 2, is a gamma enolase isoform that is primarily enriched in neuronal tissue. In scientific literature, you may find ENO2 referred to by several alternative designations including Gamma enolase, Neural enolase, Neuron specific enolase (NSE), and formerly as 14-3-2 protein . As a cytoplasmic glycolytic enzyme, ENO2 plays a crucial role in the glycolysis pathway, where it catalyzes the conversion of 2-phosphoglycerate to phosphoenolpyruvate, the ninth and penultimate step of glycolysis. The enzyme is particularly abundant in neurons and neuroendocrine cells, which makes it valuable as a tissue-specific marker in various research applications.

Where is the ENO2 gene located in the human genome and what is its genomic organization?

The gene coding for ENO2 has been mapped to the short arm of human chromosome 12, specifically in the region of pter-p1205 . Cytogenetic analysis using cell hybrids containing various deletions of human chromosome 12 has established the linear order of genes on chromosome 12 as: pter-TPI-GAPD-LDHB-ENO2-centromere-SHMT-PEPB-qter . This precise mapping has important implications for understanding the regulatory mechanisms controlling ENO2 expression and for studying potential genetic alterations affecting this locus in various pathological conditions. The ENO2 gene corresponds to Gene ID 2026 in the NCBI database, which can be used for referencing its sequence information and related genomic data .

How does ENO2 expression differ across normal human tissues and what regulates its expression?

ENO2 exhibits a distinctive tissue-specific expression pattern, with highest levels observed in neuronal tissues. Unlike alpha-enolase, which is expressed in most tissues, and beta-enolase, which is predominantly expressed in muscle tissue, gamma-enolase (ENO2) shows a more restricted expression profile . The differential expression of enolase isoforms across tissues suggests tissue-specific regulatory mechanisms.

Research indicates that ENO2 expression can be modulated in various pathophysiological conditions, particularly in cancer where altered metabolic states and the Warburg effect lead to significant changes in glycolytic enzyme expression. The transcriptional regulation of ENO2 involves specific promoter regions that can be analyzed using luciferase reporter assays to determine the impact of various factors on gene expression . Understanding these regulatory mechanisms has important implications for both basic research and potential therapeutic targeting strategies.

What are the most reliable methods for detecting and quantifying ENO2 in human samples?

Several methods are available for detecting and quantifying ENO2 in human samples, with Enzyme-Linked Immunosorbent Assay (ELISA) being one of the most widely used and reliable approaches. The solid phase sandwich ELISA technique provides high sensitivity and specificity for ENO2 detection. In this method, an antibody specific for ENO2 is pre-coated onto microwells, allowing for capture of ENO2 protein from samples during incubation. After washing, a second ENO2-specific antibody is added, followed by a horseradish peroxidase (HRP)-conjugated antibody and TMB reagent for signal development .

When selecting a detection method, researchers should consider the sample type, required sensitivity, and available equipment. For protein-level detection, Western blotting can complement ELISA results, while RT-PCR and RNA sequencing provide valuable information about ENO2 gene expression at the transcript level. Immunohistochemistry is particularly useful for visualizing ENO2 distribution in tissue sections. For high-throughput screening, protein microarrays may offer advantages in terms of sample conservation and parallel analysis of multiple markers.

What are the technical specifications of ENO2 ELISA kits and what factors affect assay performance?

Commercial ENO2 ELISA kits demonstrate high sensitivity and reproducibility for research applications. Key technical specifications include:

  • Sensitivity: 0.11 ng/mL

  • Detection range: 0.313-20 ng/mL

  • Sample compatibility: Serum, Plasma, Cell culture supernatants

  • Recovery rates: 87% for human plasma (range 71%-118%) and 90% for cell culture supernatants (range 70%-110%)

The intra-assay and inter-assay variation metrics are crucial for evaluating assay reliability:

IntraAssay Precision:

SamplenMean (ng/mL)SDCV%
1202.30.16.0
2207.60.67.8
32016.10.95.5

InterAssay Precision:

SamplenMean (ng/mL)SDCV%
1242.30.15.7
2247.40.45.6
32412.90.85.8

These performance metrics indicate excellent reproducibility with coefficient of variation (CV) values consistently below 8% . To ensure optimal assay performance, researchers should carefully follow manufacturer protocols regarding sample handling, reagent preparation, and incubation times. Special attention should be paid to potential interfering substances and sample matrix effects that could affect ELISA results.

What are the best sample preparation practices for ENO2 analysis in different specimen types?

Proper sample preparation is critical for accurate ENO2 quantification. For blood-derived specimens, researchers should consider the following best practices:

  • Serum collection: Allow blood to clot completely at room temperature (30-45 minutes), then centrifuge at 1000-2000×g for 10 minutes to separate serum.

  • Plasma collection: Use appropriate anticoagulants (EDTA, heparin, or citrate) followed by centrifugation at 1000-2000×g for 10 minutes within 30 minutes of collection.

  • Sample storage: Aliquot samples to avoid freeze-thaw cycles and store at -80°C for long-term preservation.

For cell culture supernatants, researchers should remove cells by centrifugation and avoid media components that might interfere with the assay. When working with tissue specimens, standardized homogenization protocols with appropriate buffer systems are essential to ensure consistent protein extraction and preservation of ENO2 integrity .

All samples should be thoroughly mixed after thawing and before analysis. Determining the appropriate dilution factor for each sample type is critical for ensuring measurements fall within the assay's linear range (0.313-20 ng/mL for typical ELISA kits) . When comparing different sample types, researchers should be aware of potential matrix effects and validate their preparation protocols accordingly.

How does ENO2 contribute to cancer metabolism through the Warburg effect?

ENO2 plays a significant role in cancer metabolism through its involvement in the Warburg effect, a phenomenon where cancer cells predominantly produce energy through glycolysis even in oxygen-rich conditions. Research has demonstrated that ENO2 can enhance glycolysis levels in cancer cells, thereby promoting tumor growth and progression . In clear cell renal cell carcinoma (ccRCC), ENO2 has been shown to affect glucose utilization, lactate production, and intracellular ATP generation, all of which are critical aspects of altered energy metabolism in cancer cells .

The mechanistic contribution of ENO2 to the Warburg effect involves:

  • Acceleration of glycolytic flux, allowing cancer cells to rapidly generate ATP

  • Enhancement of lactate production, creating an acidic microenvironment favorable for tumor invasion

  • Redirection of glycolytic intermediates to biosynthetic pathways supporting rapid cellular proliferation

  • Modulation of metabolic signaling networks that interface with growth and survival pathways

Experimental techniques to assess ENO2's metabolic impact include glucose consumption assays, lactate production measurements, ATP detection, and metabolic flux analysis using isotope-labeled glucose . These methodological approaches provide valuable insights into how ENO2 reprograms energy metabolism in cancer cells and offer potential avenues for therapeutic intervention.

What evidence supports ENO2 as a diagnostic and prognostic biomarker in clear cell renal cell carcinoma?

Substantial evidence supports ENO2 as a valuable diagnostic and prognostic biomarker in clear cell renal cell carcinoma (ccRCC). Through comprehensive bioinformatic analysis of multiple gene expression datasets (GSE36895, GSE66272, and GSE71963) from the Gene Expression Omnibus database, researchers have identified ENO2 as significantly differentially expressed between normal kidney and ccRCC tissues . Validation using the Oncomine dataset has further confirmed ENO2's potential as a key parameter for ccRCC diagnosis and management.

Kaplan-Meier survival analysis and Cox proportional hazards regression analysis have established that ENO2 can independently predict clinical prognosis in ccRCC patients . Higher ENO2 expression levels correlate with poorer survival outcomes, suggesting its value as a prognostic indicator. Functional experiments both in vitro and in vivo have substantiated ENO2's role in promoting ccRCC progression.

The diagnostic utility of ENO2 is enhanced by its involvement in glycolysis, which represents a critical pathway in ccRCC pathogenesis as demonstrated through KEGG pathway enrichment and Gene Ontology functional annotation analyses . This metabolic connection provides a biological rationale for ENO2's role as a ccRCC biomarker and potential therapeutic target.

How does ENO2 influence the epithelial-mesenchymal transition and immune microenvironment in cancer?

ENO2 has been implicated in modulating the epithelial-mesenchymal transition (EMT) process in renal cell carcinoma, a critical step in cancer progression and metastasis . The EMT process enables cancer cells to lose their epithelial characteristics and acquire mesenchymal properties, enhancing their migratory and invasive capabilities. Research indicates that ENO2 affects this transition, though the exact molecular mechanisms require further elucidation.

Additionally, ENO2 participates in regulating the tumor immune microenvironment , which has profound implications for cancer immunotherapy approaches. The tumor microenvironment consists of various immune cells, stromal cells, and soluble factors that collectively influence tumor growth, progression, and response to therapy. ENO2's regulatory role may involve:

  • Modulation of immune cell infiltration patterns

  • Influence on cytokine and chemokine production

  • Alteration of immune checkpoint expression

  • Metabolic reprogramming of immune cells within the tumor microenvironment

These findings suggest that ENO2 not only serves as a biomarker but also actively contributes to cancer progression through multiple mechanisms beyond its canonical role in glycolysis. Understanding these broader functions could lead to more comprehensive therapeutic strategies targeting ENO2 in cancer treatment.

What gene silencing approaches have been most effective for studying ENO2 function in cancer models?

Several gene silencing approaches have proven effective for investigating ENO2 function in cancer models. RNA interference (RNAi) using small interfering RNA (siRNA) has been successfully employed in studies of clear cell renal cell carcinoma, where ENO2 siRNA was transfected into cancer cells using standard transfection reagents like Lipofectamine 3000 . This approach allows for transient knockdown of ENO2 expression to study acute effects on cancer cell behavior.

For longer-term studies, stable knockdown of ENO2 using short hairpin RNA (shRNA) delivered via lentiviral vectors has proven effective, particularly for in vivo experiments. This approach was employed in animal models where A498 cells with stable ENO2 knockdown were subcutaneously injected into athymic nude mice to assess tumor growth . The use of appropriate control cells (scrambled shRNA) is essential for proper experimental design.

Recent advances in CRISPR-Cas9 genome editing technology offer additional precision for studying ENO2 function through complete gene knockout or targeted modifications of regulatory regions. When designing such experiments, researchers should consider:

  • Validation of knockdown/knockout efficiency at both mRNA and protein levels

  • Assessment of off-target effects through appropriate controls

  • Rescue experiments to confirm specificity of observed phenotypes

  • Selection of cell lines that exhibit ENO2 dependency for most informative results

The choice of silencing method should be guided by specific research questions, required duration of effect, and downstream experimental applications.

How can researchers effectively design studies to elucidate the relationship between ENO2 and tumor immunology?

Designing studies to elucidate the relationship between ENO2 and tumor immunology requires a multifaceted approach combining molecular, cellular, and in vivo techniques. Researchers should consider the following methodological framework:

  • Transcriptomic analysis: Perform RNA-seq or microarray analysis of ENO2-manipulated cancer cells to identify changes in immunomodulatory gene expression. Bioinformatic tools like ClueGo in Cytoscape can help identify enriched immune-related pathways .

  • Immune cell co-culture systems: Establish co-culture models of cancer cells with various immune cell populations (T cells, macrophages, NK cells) to assess how ENO2 manipulation affects immune cell recruitment, activation, and function.

  • Cytokine/chemokine profiling: Measure secreted factors in conditioned media from ENO2-modified cells using multiplex assays to identify changes in the immunomodulatory secretome.

  • Flow cytometry: Analyze immune cell infiltration and phenotypic changes in tumor models with differential ENO2 expression.

  • Syngeneic mouse models: Utilize immunocompetent mouse models with ENO2-manipulated cancer cells to study the full spectrum of tumor-immune interactions in vivo.

  • Correlation with immune checkpoint markers: Assess relationships between ENO2 expression and immune checkpoint molecules (PD-L1, CTLA-4) in patient samples to identify potential therapeutic implications.

  • Single-cell RNA sequencing: Apply this technique to tumor samples to resolve heterogeneity in ENO2 expression and its correlation with immune cell populations at single-cell resolution.

Through careful experimental design and integration of multiple approaches, researchers can unravel the complex relationships between ENO2, cancer metabolism, and the immune microenvironment.

What are the most promising approaches for targeting ENO2 therapeutically and what challenges must be overcome?

The therapeutic targeting of ENO2 represents a promising avenue for cancer treatment, particularly for malignancies like clear cell renal cell carcinoma where ENO2 plays a significant role in disease progression. Several approaches show potential:

  • Small molecule inhibitors: Development of specific ENO2 inhibitors that exploit structural differences between ENO2 and other enolase isoforms to achieve selectivity.

  • RNA-based therapeutics: siRNA or antisense oligonucleotides delivered via nanoparticles or lipid formulations could downregulate ENO2 expression in tumor cells.

  • Metabolic combination therapy: Combining ENO2 inhibition with other metabolic pathway modulators to achieve synergistic anti-tumor effects by comprehensively disrupting cancer energy metabolism.

  • Antibody-drug conjugates: ENO2-targeted antibodies linked to cytotoxic payloads could deliver therapeutic agents specifically to ENO2-expressing cells.

  • Selectivity: Designing inhibitors that specifically target ENO2 without affecting other enolase isoforms necessary for normal cellular function.

  • Target accessibility: Determining whether ENO2 is sufficiently exposed in cancer cells for antibody-based approaches.

  • Delivery systems: Developing effective delivery systems for RNA-based therapeutics that can achieve adequate tumor penetration.

  • Resistance mechanisms: Identifying and addressing potential compensatory pathways that may emerge following ENO2 inhibition.

  • Patient stratification: Establishing predictive biomarkers to identify patients most likely to benefit from ENO2-targeted therapies.

Research using preclinical models, including cell lines and mouse xenografts as described in the literature , will be essential for addressing these challenges and advancing ENO2-targeted therapies toward clinical applications.

What critical controls should be included in experimental designs studying ENO2 function?

When designing experiments to study ENO2 function, researchers should implement a comprehensive set of controls to ensure reliable and interpretable results:

  • Positive and negative tissue/cell controls: Include samples known to express high levels of ENO2 (neuronal tissue/cells) as positive controls and those with minimal expression (e.g., some epithelial cell lines) as negative controls.

  • Knockdown/overexpression validation: For functional studies using gene manipulation, validate the efficiency of ENO2 knockdown or overexpression at both mRNA (qRT-PCR) and protein (Western blot, ELISA) levels .

  • Rescue experiments: Following ENO2 knockdown, perform rescue experiments by re-expressing ENO2 to confirm that observed phenotypes are specifically due to ENO2 depletion rather than off-target effects.

  • Isotype controls: For immunoassays and flow cytometry, include appropriate isotype controls to account for non-specific antibody binding.

  • Vehicle controls: In inhibitor studies, include proper vehicle controls containing the same solvent used to dissolve the inhibitor at the same concentration.

  • Housekeeping gene/protein controls: Use validated housekeeping genes or proteins as internal controls for normalization in expression studies, ensuring they remain stable under your experimental conditions.

  • Time course controls: For processes that change over time, include appropriate time-matched controls to distinguish ENO2-specific effects from temporal changes.

  • In vivo controls: For animal studies, include both wild-type animals and those injected with control cells (e.g., cells transfected with scrambled shRNA) alongside the experimental group with ENO2-manipulated cells .

How should researchers interpret contradictory findings regarding ENO2 expression across different cancer types?

Interpreting contradictory findings regarding ENO2 expression across different cancer types requires careful consideration of several factors:

To reconcile contradictory findings, researchers should systematically compare methodologies, validate key findings using multiple techniques, and contextualize results within the specific biological setting of each cancer type.

What are the key considerations for validating ENO2 as a clinically relevant biomarker?

Validating ENO2 as a clinically relevant biomarker requires a rigorous, multi-phase approach addressing several critical considerations:

  • Analytical validation: Establish precise, accurate, and reproducible assays for ENO2 measurement. This includes determining sensitivity, specificity, and reproducibility metrics as demonstrated in the ELISA performance data showing inter- and intra-assay CV% values consistently below 8% .

  • Clinical validation: Confirm the association between ENO2 levels and specific clinical outcomes through appropriate statistical analyses. This should include:

    • Receiver Operating Characteristic (ROC) analysis to determine optimal cutoff values

    • Survival analyses like Kaplan-Meier and Cox proportional hazards regression to assess prognostic value

    • Multivariate analyses to establish ENO2 as an independent predictor rather than a confounding factor

  • Sample size considerations: Ensure adequate statistical power through appropriate sample size calculations for both discovery and validation cohorts.

  • Biological plausibility: Establish a mechanistic understanding of ENO2's role in the disease process, as demonstrated through functional experiments linking ENO2 to glycolysis and the Warburg effect in cancer cells .

  • Clinical utility assessment: Determine whether ENO2 measurement provides actionable information that improves clinical decision-making beyond existing biomarkers.

  • Standardization: Develop standardized protocols for sample collection, processing, storage, and analysis to ensure consistency across laboratories.

  • External validation: Confirm findings in independent patient cohorts from different institutions to ensure generalizability.

  • Comparative effectiveness: Compare ENO2's performance against existing biomarkers to demonstrate added value in clinical settings.

Following these comprehensive validation steps will help establish whether ENO2 can transition from a research-focused biomarker to a clinically useful tool for patient stratification, treatment selection, or monitoring disease progression.

Product Science Overview

Structure and Function

Enolase-2 is composed of two subunits and has a molecular weight of approximately 46 kDa under reducing conditions . The enzyme is highly specific to neurons and neuroendocrine cells, making it a valuable marker for neuronal differentiation and neuroendocrine tumors .

Recombinant Production

Recombinant Human Enolase-2 is typically produced in Escherichia coli (E. coli) expression systems. The recombinant protein is often tagged with a histidine (His) tag to facilitate purification. The protein is purified to a high degree, with a purity greater than 95% as determined by SDS-PAGE and visualized with silver staining .

Applications

Recombinant Enolase-2 is used in various research applications, including:

  • Biochemical studies: Understanding the enzyme’s role in glycolysis and gluconeogenesis.
  • Clinical diagnostics: Serving as a biomarker for neuronal damage and neuroendocrine tumors.
  • Drug development: Screening for potential inhibitors or modulators of enolase activity.
Storage and Stability

The recombinant protein is typically supplied as a 0.2 μm filtered solution in a buffer containing MES, NaCl, KCl, and MgSO4. It is recommended to store the protein at -20 to -70 °C to maintain its stability. Avoiding repeated freeze-thaw cycles is crucial to preserve its activity .

Assay Procedure

The activity of recombinant Enolase-2 can be measured by its ability to convert phosphoglyceric acid to phosphoenolpyruvate. The specific activity is typically greater than 6,000 pmol/min/μg under the described conditions .

Enolase-2’s role in glycolysis and its specificity to neurons make it a significant enzyme for both basic research and clinical applications. Its recombinant form allows for detailed studies and potential therapeutic developments.

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 2024 Thebiotek. All Rights Reserved.