ENO1 Human

Enolase-1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Cancer Progression

ENO1 is overexpressed in multiple cancers, correlating with poor prognosis . Mechanisms include:

  • Metabolic reprogramming: Drives the Warburg effect via hypoxia-inducible factor-1α (HIF-1α) and glycolysis-related gene upregulation .

  • Invasion and metastasis: Binds plasminogen to degrade extracellular matrix and promote cell migration .

  • Exosome-mediated transfer: Exosomal ENO1 upregulates integrin α6β4 and activates FAK/Src-p38MAPK pathways in hepatocellular carcinoma .

Table 2: ENO1 Overexpression in Human Cancers

Cancer TypeAssociated Pathways/EffectsClinical Correlation
Glioblastoma PI3K/AKT activation, chemoresistanceReduced survival
Lung Cancer CDK6-mediated G1/S transition, AKT/p38 activationEnhanced proliferation
Pancreatic Cancer ERK phosphorylation, apoptosis resistanceMetastasis, poor prognosis
Hepatocellular Carcinoma Exosome-mediated integrin signalingTNM stage association

Autoimmunity and Infection

ENO1 serves as an autoantigen in Hashimoto encephalopathy and a plasminogen receptor in bacterial infections (e.g., streptococci) .

Mechanisms of Oncogenesis

  • Transcriptional regulation: The nuclear isoform MBP-1 binds the c-myc promoter, suppressing oncogene expression .

  • Metabolic crosstalk: ENO1 stabilizes choline kinase α (CHKα), enhancing phosphatidylcholine synthesis in glioblastoma .

  • Therapy resistance: ENO1 deletion sensitizes tumors to ENO2 inhibition via synthetic lethality .

Key Findings:

  • Knockout of ENO1 in pancreatic cancer activates oxidative phosphorylation and lipid metabolism, reducing tumorigenesis .

  • Hypoxia-induced ENO1 promotes ERK phosphorylation, enabling apoptosis evasion in pancreatic ductal adenocarcinoma .

Therapeutic Targeting

ENO1 is a promising target due to its surface expression in tumors and role in therapy resistance:

  • Antibody-based inhibition: Humanized anti-ENO1 antibodies reduce lactate production and VEGF secretion in myeloma and prostate cancer .

  • Gene silencing: shRNA-mediated knockdown suppresses proliferation in lung cancer (H1299, H460) and hepatocellular carcinoma .

Table 3: Therapeutic Strategies Targeting ENO1

ApproachModel SystemOutcome
Anti-ENO1 mAb Multiple MyelomaReduced glycolysis, cell migration
Exosome blockade Hepatocellular CarcinomaInhibited lung metastasis
CRISPR knockout Pancreatic CancerMetabolic reprogramming

Research Advancements

Recent studies highlight novel roles:

  1. ENO1-CHKα axis: ENO1 stabilizes CHKα, linking glycolysis to phospholipid metabolism in glioblastoma .

  2. Extracellular regulation: Secreted ENO1 enhances glycolysis via HIF-1α/GLUT1 in myeloma cells, independent of enzymatic activity .

  3. Synthetic lethality: Co-deletion of ENO1 and MIR34A in glioblastoma creates dependency on ENO2, a vulnerability for targeted therapy .

Product Specs

Introduction
ENO1, also known as Enolase-1, is a crucial glycolytic enzyme found in nearly all tissues. This homodimeric protein exists in two forms: a cytoplasmic full-length form and a shorter nuclear form. The full-length ENO1 participates in glycolysis, particularly under hypoxic conditions, and acts as a plasminogen receptor on cell surfaces during tissue invasion. The shorter form is involved in the regulation of the c-myc promoter. Deregulated ENO1 expression is implicated in the progression of several cancers, including breast and lung cancer. Furthermore, ENO1 serves as an autoantigen in autoimmune diseases such as Hashimoto's encephalopathy and severe asthma. In Behcet's disease, ENO1 is targeted by serum anti-endothelial antibodies.
Description
This product consists of the recombinant human ENO1 protein, produced in E. coli. This non-glycosylated protein is a single polypeptide chain with a molecular weight of 47.1kDa, encompassing amino acids 1 to 434.
Physical Appearance
A clear, sterile-filtered solution.
Formulation
The ENO1 protein is provided at a concentration of 1mg/ml in a solution containing 20mM Tris-HCl (pH 7.5), 1mM MgSO4, and 10% glycerol.
Stability
For short-term storage (up to 4 weeks), keep the vial refrigerated at 4°C. For extended storage, freeze the protein at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Repeated freezing and thawing should be avoided.
Biological Activity
The specific activity of this protein is measured as greater than 20,000 pmol/min/ug. This value is determined by monitoring the decrease in NAD absorbance at 340nm, which corresponds to the formation of NADH at pH 6.5 and 37°C.
Purity
The purity of this protein is determined to be greater than 90% using SDS-PAGE analysis.
Synonyms
NNE, PPH, MPB1, MBP-1, ENO1L1, ENO1, Alpha-Enolase, Enolase-Alpha, 2-phospho-D-glycerate hydro-lyase, Non-neural enolase, Enolase 1, MPB-1, Phosphopyruvate hydratase, C-myc promoter-binding protein, Plasminogen-binding protein, MBPB1.
Source
Escherichia Coli.
Amino Acid Sequence
MSILKIHARE IFDSRGNPTV EVDLFTSKGL FRAAVPSGAS TGIYEALELR DNDKTRYMGK GVSKAVEHIN KTIAPALVSK KLNVTEQEKI DKLMIEMDGT ENKSKFGANA ILGVSLAVCK AGAVEKGVPL YRHIADLAGN SEVILPVPAF NVINGGSHAG NKLAMQEFMI LPVGAANFRE AMRIGAEVYH NLKNVIKEKY GKDATNVGDE GGFAPNILEN KEGLELLKTA IGKAGYTDKV VIGMDVAASE FFRSGKYDLD FKSPDDPSRY ISPDQLADLY KSFIKDYPVV SIEDPFDQDD WGAWQKFTAS AGIQVVGDDL TVTNPKRIAK AVNEKSCNCL LLKVNQIGSV TESLQACKLA QANGWGVMVS HRSGETEDTF IADLVVGLCT GQIKTGAPCR SERLAKYNQL LRIEEELGSK AKFAGRNFRN PLAK.

Q&A

What is ENO1 and what are its primary functions in human cells?

The protein exists in multiple forms - cytosolic, membrane-bound, and secreted - each with distinct functions. While cytosolic ENO1 participates primarily in glycolysis, surface and extracellular ENO1 demonstrate unique biological activities. Studies have shown that extracellular ENO1 can enhance glycolytic activity by increasing hypoxia-inducible factor 1-α (HIF-1α) expression, thereby promoting tumor progression .

Importantly, knockout studies have identified ENO1 as an indispensable factor in cancer progression, where its deletion inhibits cell invasion and migration capabilities . This multifunctionality makes ENO1 an attractive target for cancer research and potential therapeutic development.

How is ENO1 expression typically measured in human samples?

Researchers employ multiple complementary techniques to measure ENO1 expression in human samples, each with specific advantages:

Protein-level detection methods:

  • Western blotting: The gold standard for quantifying total cellular ENO1 protein expression and validating knockout or knockdown efficiency

  • Immunohistochemistry: Used for spatial distribution analysis of ENO1 in tissue samples, allowing visualization of expression patterns across different cell types and microenvironments

  • Flow cytometry: Essential for detection and quantification of surface ENO1 on non-permeabilized cells, distinguishing membrane-bound from intracellular protein

  • Antibody labeling assays: Specialized techniques to specifically detect membrane-bound ENO1

Genetic expression methods:

  • RT-qPCR: Enables sensitive quantification of ENO1 mRNA expression, particularly useful for assessing transcriptional regulation

  • RNA-seq: Provides comprehensive transcriptomic profiling, allowing comparative analysis of ENO1 across different conditions

For tissue microarray analysis of ENO1 expression, quantification typically involves measuring the positively stained area, with statistical significance evaluated using appropriate tests such as two-tailed unpaired Student's t-tests . When analyzing ENO1 expression in cancer databases like TIMER and GEPIA, researchers typically use log2(TPM) for log-scale and match TCGA normal and GTEx data .

What experimental models are commonly used to study ENO1 function?

Researchers utilize diverse experimental models to investigate ENO1 function across multiple contexts:

Cell line models:

  • Multiple myeloma lines (RPMI-8226, U266, KMS-11): Frequently used to study ENO1's role in hematological malignancies

  • Solid tumor lines (PC-3): Employed to investigate ENO1 function in epithelial cancers

  • These models permit direct manipulation of ENO1 expression and assessment of resulting phenotypes

Genetic manipulation approaches:

  • CRISPR-Cas9 knockout: Generation of stable ENO1-knockout cell lines using lentiviral delivery of sgRNAs targeting the ENO1 gene, followed by antibiotic selection (typically 2mg/L puromycin for 14 days) and monoclonal isolation

  • RNAi-mediated knockdown: Transient reduction of ENO1 expression using targeted siRNAs to assess acute effects

  • Overexpression systems: Introduction of ENO1-expressing plasmids (such as pLenti-C-Myc-DDK-P2A-Puro) using transfection reagents, followed by antibiotic selection to establish stable overexpression models

In vivo models:

  • NOG mice (NOD-SCID IL-2 receptor gamma null): Used for tumorigenicity assays to assess ENO1's influence on tumor growth in vivo

  • Xenograft models: Enable evaluation of ENO1-manipulated cancer cells in a more physiologically relevant environment, including assessment of tumor growth kinetics and metabolic parameters

Biochemical and structural studies:

  • Recombinant protein systems: Production of purified ENO1 for in vitro enzymatic and protein interaction studies

  • Computational modeling: Analysis of ENO1's 3D structure using tools like SiteMap and FTMap to identify potential binding sites for protein-protein interactions

Each model offers distinct advantages, and combining multiple approaches provides comprehensive insights into ENO1's diverse functions.

How does extracellular ENO1 differ functionally from intracellular ENO1?

Extracellular ENO1 (surface-bound or secreted) demonstrates distinct functions from its intracellular counterpart, revealing a sophisticated dual role for this protein in cancer biology:

Functional differences:

Extracellular ENO1 enhances glycolytic activity through an indirect mechanism, primarily by increasing HIF-1α expression, which subsequently upregulates glycolysis-related genes. This represents a unique regulatory pathway distinct from intracellular ENO1's direct enzymatic role in glycolysis .

Specifically, extracellular ENO1 promotes:

  • Enhanced expression of HIF1A, HK2, and GLUT1 mRNA and protein levels

  • Increased lactate production and intracellular LDH activity

  • Elevated cell migration, viability, and tumor-promoting cytokine secretion

  • Enhanced VEGF production (a key mediator of angiogenesis)

These effects can be specifically blocked using ENO1-specific monoclonal antibodies, confirming their dependence on extracellular ENO1 .

Methodological approaches to distinguish these forms:

TechniqueApplicationAdvantage
Flow cytometryDetects surface ENO1 on non-permeabilized cellsQuantitative measurement of membrane-bound protein
Antibody labelingIdentifies membrane-bound ENO1Distinguishes surface from intracellular protein
Recombinant protein treatmentStudies extracellular ENO1 effectsIsolates extracellular functions from intracellular ones
ENO1-specific antibodiesBlocks extracellular ENO1 functionConfirms specificity of observed effects

Knockdown studies have demonstrated that reducing total cellular ENO1 simultaneously decreases surface ENO1 expression, suggesting a dynamic relationship between these pools . This finding highlights the importance of considering both intracellular and extracellular ENO1 when investigating its role in cancer biology.

What are the protein-protein interaction domains of ENO1?

The identification of ENO1's protein-protein interaction domains, particularly with Hsp70, reveals critical structural insights into its non-glycolytic functions:

Domain identification methods:

Researchers have employed sophisticated in silico approaches to map ENO1's interaction domains:

  • SiteMap analysis identifies binding sites based on parameters including site size, solvent exposure, tightness, and hydrophobic/hydrophilic character

  • FTMap evaluation uses small molecular probes of different sizes, shapes, and polarities to map protein surfaces and identify energetically favorable binding regions

  • 3D protein structure analysis utilizes structures from the Protein Data Bank to conduct computational analyses

Key findings:

Through these approaches, research has identified that the Hsp70-binding domain is localized to an internal region of ENO1, specifically including amino acids from positions 162 to 282 . This region was first-ranked by SiteMap with both the best SiteScore (>0.80 threshold) and DScore (>0.98 threshold as a druggable site) .

The identification of this specific interaction domain is significant because both ENO1 and Hsp70 are multifunctional proteins overexpressed in numerous human cancers . Understanding their interaction provides insights into potential mechanisms of cancer progression and identifies targets for therapeutic intervention.

Validation approaches:

For in vitro confirmation of these computationally predicted interactions, researchers typically employ:

  • Molecular dynamics simulations to analyze the stability and characteristics of the protein complex over time

  • Protein fragment analysis to verify specific binding regions

  • Site-directed mutagenesis to confirm the functional importance of specific residues

This combined computational and experimental approach provides a comprehensive understanding of ENO1's protein interaction capabilities that extend beyond its glycolytic function.

How does ENO1 contribute to cancer metabolism reprogramming?

ENO1 plays multifaceted roles in cancer metabolism reprogramming, extending far beyond its canonical enzymatic function in glycolysis:

Glycolytic enhancement mechanisms:

ENO1 significantly influences cancer metabolism through several distinct mechanisms:

  • Direct enzymatic activity: As a glycolytic enzyme, ENO1 catalyzes the conversion of 2-phosphoglycerate to phosphoenolpyruvate, directly supporting enhanced glycolysis in cancer cells

  • HIF-1α pathway activation: Extracellular ENO1 increases HIF-1α expression, which orchestrates a comprehensive glycolytic program

  • Glycolysis-related gene regulation: ENO1 elevates expression of key glycolytic regulators including HIF1A, HK2, and GLUT1 at both mRNA and protein levels

  • Enhanced lactate production: Both knockdown studies and extracellular ENO1 treatment demonstrate ENO1's critical role in lactate generation, the endpoint of aerobic glycolysis

Experimental evidence:

Multiple experimental approaches have confirmed ENO1's metabolic influence:

  • ENO1 knockout leads to substantial metabolic reprogramming in cancer cells

  • Knockdown of ENO1 expression reduces lactate production, cell viability, and migration capabilities in multiple myeloma and prostate cancer cells

  • Extracellular ENO1 treatment dose-dependently increases lactate secretion and enhances intracellular LDH activity, effects that can be specifically blocked with ENO1 monoclonal antibodies

In vivo significance:

Administration of ENO1-specific antibodies reduces tumor growth and serum lactate levels in multiple myeloma xenograft models, demonstrating that targeting ENO1 can impact cancer metabolism in vivo . This finding highlights ENO1's potential as a therapeutic target, particularly through inhibition of its extracellular functions.

The ability of ENO1 to regulate metabolism through both enzymatic and signaling functions represents a unique dual mechanism that contributes to cancer progression and offers multiple intervention points for therapeutic development.

How can researchers effectively knockout ENO1 in cancer cell lines?

Generating stable ENO1 knockout cell lines requires precise methodology and careful validation:

Step-by-step protocol:

  • Vector construction:

    • Design sgRNA sequences based on the ENO1 gene sequence

    • Clone sgRNAs into an appropriate plasmid (e.g., LentiCRISPR v2) using restriction sites

    • Verify construct by sequencing

  • Lentivirus production:

    • Generate recombinant lentiviruses using packaging systems (e.g., ViraPower Packaging Mix)

    • Collect viral supernatant and filter to remove cell debris

  • Target cell infection:

    • Infect target cancer cells with lentiviruses for approximately 48 hours

    • Apply appropriate antibiotic selection (e.g., 2mg/L puromycin) for approximately 14 days

    • This extended selection period ensures elimination of non-transduced cells

  • Monoclonal isolation:

    • Subculture the selected population to obtain monoclonal cells

    • Screen multiple clones to identify those with complete ENO1 knockout

  • Validation:

    • Confirm ENO1 knockout by detecting protein deficiency using Western blotting

    • Verify functional consequences through glycolytic assays (e.g., lactate production)

Critical considerations:

  • sgRNA design: Create multiple sgRNAs targeting different ENO1 regions to increase knockout efficiency and reduce off-target effects

  • Control selection: Include cells transduced with non-targeting sgRNAs as controls

  • Functional validation: Assess both protein expression and functional consequences (glycolysis, migration, invasion) to confirm complete knockout

  • Compensatory mechanisms: Monitor potential upregulation of other enolase isoforms (ENO2, ENO3) that might compensate for ENO1 loss

  • Cell viability impact: Be aware that complete ENO1 knockout may significantly impact cell viability in some cancer types, potentially requiring alternative approaches like inducible systems

Successful ENO1 knockout has been demonstrated to inhibit cell invasion and migration capabilities, confirming ENO1's role as an indispensable factor in cancer progression . This approach provides a powerful tool for investigating ENO1's diverse functions in cancer biology.

What are the methodological approaches for studying ENO1's role in the HIF-1α pathway?

Investigating ENO1's relationship with the HIF-1α pathway requires multifaceted experimental approaches:

Key methodological strategies:

  • Gene and protein expression analysis:

    • RT-qPCR to measure HIF1A, HK2, GLUT1, and other glycolysis-related genes after ENO1 manipulation

    • Western blotting to detect HIF-1α, HK2, and GLUT1 protein levels following ENO1 treatment

    • These analyses reveal that extracellular ENO1 enhances HIF-1α expression and its downstream targets

  • Functional metabolic assays:

    • Lactate production measurement to assess glycolytic output

    • Intracellular LDH activity assays to evaluate the conversion of pyruvate to lactate

    • Both parameters increase following extracellular ENO1 treatment, demonstrating enhanced glycolytic activity

  • Mechanistic dissection:

    • HIF-1α silencing experiments to determine which ENO1-mediated effects are HIF-1α-dependent

    • Combined treatment with ENO1 and inhibitors of the HIF-1α pathway

    • These approaches reveal that extracellular ENO1-mediated glycolysis, glycolysis-related gene expression, and pro-cancer activities are reduced by HIF-1α silencing, confirming the pathway dependence

  • Extracellular ENO1 intervention:

    • Recombinant ENO1 protein addition to study extracellular effects on the HIF-1α pathway

    • ENO1-specific monoclonal antibody treatments to block extracellular functions

    • The ability of ENO1 mAbs to inhibit ENO1-induced cellular effects demonstrates specificity and therapeutic potential

  • In vivo validation:

    • Administration of ENO1 mAb in xenograft models

    • Measurement of tumor growth and serum lactate levels

    • These studies confirm that targeting ENO1 reduces both tumor growth and glycolytic activity in vivo

This methodological framework enables researchers to establish not only the correlation between ENO1 and HIF-1α but also the causal relationship and dependency, providing insights into a novel regulatory mechanism with therapeutic implications.

What statistical approaches should be used when analyzing ENO1 expression data?

Recommended statistical methods by experimental design:

  • For comparing two groups:

    • Two-tailed unpaired Student's t-tests for independent samples (e.g., comparing ENO1 expression between tumor and normal tissues)

    • Two-tailed paired Student's t-tests for matched samples (e.g., comparing treatment effects in the same cell line)

    • Wilcoxon test for evaluating differential expression between cancer and normal tissues in database analyses

  • For multiple group comparisons:

    • One-way analysis of variance (ANOVA) followed by Tukey's post hoc test for individual group comparisons

    • This approach is essential when comparing effects across multiple conditions or cell lines

  • For survival analysis:

    • Kaplan-Meier curves with log-rank tests to evaluate ENO1 expression's prognostic value

    • Patient stratification using automated "best cutoff" value selection for optimal group separation

  • For gene expression analysis:

    • Log2 transformation of expression data (TPM or TPM+1) for normalization

    • Appropriate housekeeping genes for RT-qPCR normalization

Data presentation standards:

  • Present data as mean ± SEM from at least three separate experiments to account for biological variability

  • Use box plots for comparing expression levels between groups in database analyses

  • Apply consistent P-value thresholds (typically P<0.05 is considered statistically significant)

  • Include appropriate sample sizes with power calculations where possible

Software recommendations:

Researchers commonly utilize statistical packages such as:

  • SPSS for comprehensive statistical analyses

  • R for advanced bioinformatic analyses and visualization

  • GraphPad Prism for biomedical research data analysis and presentation

Adhering to these statistical best practices ensures robust, reproducible findings when analyzing ENO1 expression and its functional implications in cancer research.

How can ENO1-specific antibodies be developed and validated for cancer therapy?

Developing ENO1-specific antibodies for cancer therapy requires systematic validation across multiple experimental systems:

Development strategy:

  • Target identification:

    • Focus on extracellular/surface ENO1, which promotes glycolysis and pro-cancer activities in multiple myeloma via the HIF-1α pathway

    • This approach targets cancer-promoting functions while potentially sparing intracellular enzymatic activities

  • Antibody generation:

    • Develop monoclonal antibodies against purified recombinant ENO1

    • Screen for antibodies that specifically recognize native conformations of ENO1

    • Select candidates based on binding affinity and specificity

  • In vitro validation:

    • Confirm antibody specificity through immunoblotting, ELISA, and immunoprecipitation

    • Evaluate functional effects on cancer cells:

      • Inhibition of extracellular ENO1-mediated glycolysis (lactate production)

      • Reduction of HIF-1α expression and downstream glycolysis-related genes

      • Suppression of cell migration, viability, and tumor-promoting cytokine secretion

  • In vivo efficacy assessment:

    • Test antibodies in xenograft models using appropriate cancer cell lines

    • Monitor tumor growth kinetics over time

    • Measure serum lactate levels as a biomarker of glycolytic activity

    • ENO1 mAb administration has been shown to successfully reduce tumor growth and serum lactate levels in multiple myeloma xenograft models

Efficacy parameters:

Research has demonstrated that ENO1-specific antibodies can effectively counteract multiple extracellular ENO1-induced effects:

  • Inhibition of enhanced lactate production

  • Reduction of HIF-1α, HK2, and GLUT1 expression

  • Suppression of cell viability increases

  • Blockade of VEGF production enhancement

These findings highlight the therapeutic potential of ENO1-specific antibodies, particularly through glycolysis inhibition, and warrant further studies in other cancer types . The approach represents a targeted strategy that exploits ENO1's unique extracellular functions in cancer promotion.

What are potential biomarker applications of ENO1 in cancer diagnostics?

ENO1 shows significant potential as a cancer biomarker across multiple applications:

Expression patterns in cancer:

ENO1 expression is frequently elevated in multiple cancer types compared to normal tissues, as demonstrated through comprehensive database analyses:

  • TIMER and GEPIA database analyses reveal significant differential expression between cancers and paired normal tissues

  • Tissue microarray studies show increased ENO1 positivity in multiple myeloma compared to normal bone marrow, quantifiable through immunohistochemical staining

Prognostic value:

ENO1 expression levels correlate with clinical outcomes in various cancers:

  • Kaplan-Meier Plotter analyses demonstrate prognostic value across multiple cancer types

  • Higher ENO1 expression is generally associated with poorer outcomes, reflecting its role in promoting cancer progression

Multi-platform detection approaches:

Detection MethodSample TypeClinical Application
ImmunohistochemistryTissue biopsiesDiagnostic classification, prognostic stratification
Western blottingTissue lysatesProtein level quantification
RT-qPCRTissue or liquid biopsiesmRNA expression analysis
Flow cytometryCell suspensionsSurface ENO1 detection

Methodological considerations:

For optimal biomarker development:

  • Standardize cutoff values using approaches like "best cutoff" methods employed in prognostic databases

  • Validate findings across independent cohorts using consistent methodologies

  • Consider ENO1 in multi-biomarker panels for improved specificity and sensitivity

  • Account for cancer-specific contexts when interpreting ENO1 expression

The dual role of ENO1 as both a glycolytic enzyme and regulator of cancer-promoting pathways makes it particularly valuable as a biomarker that reflects fundamental aspects of cancer metabolism and progression. Further clinical validation studies are warranted to establish standardized assays for routine clinical application.

How does ENO1 knockout impact tumor progression in animal models?

ENO1 knockout demonstrates significant tumor-suppressive effects in preclinical models:

Experimental approaches:

Researchers have employed multiple strategies to study ENO1 knockout effects in vivo:

  • Subcutaneous transplantation of ENO1-knockout cells into immunocompromised mice (NOG mice)

  • Assessment of tumor growth kinetics through regular measurement of tumor dimensions

  • Calculation of tumor volume using the formula V = L0.5, where L and W represent the largest and smallest diameters, respectively

Key findings:

ENO1 knockout substantially impacts tumor progression through multiple mechanisms:

  • Reduced tumor growth: Animal studies demonstrate significantly decreased tumor volume in ENO1-knockout models compared to controls

  • Metabolic reprogramming: ENO1 knockout leads to fundamental changes in cancer cell metabolism that impair tumor progression

  • Decreased glycolytic activity: Measurement of serum lactate levels (the endpoint of glycolysis) reveals reduced glycolytic output in ENO1-targeted models

  • Impaired invasive capacity: ENO1 knockout inhibits the invasive and migratory capabilities essential for tumor progression

Complementary approaches:

In addition to genetic knockout, antibody-based targeting provides further evidence of ENO1's role:

  • Administration of ENO1-specific monoclonal antibodies reduces tumor growth in xenograft models

  • This approach specifically targets extracellular/surface ENO1 functions while potentially preserving intracellular activities

These findings collectively identify ENO1 as an indispensable factor in tumor progression across multiple cancer types, validating its potential as a therapeutic target. The dual approach of genetic knockout and antibody targeting provides complementary evidence for ENO1's critical role in cancer biology and offers multiple intervention strategies for potential clinical development.

What are emerging areas of ENO1 research with therapeutic potential?

Several cutting-edge research directions are expanding our understanding of ENO1 and revealing new therapeutic opportunities:

Extracellular ENO1 targeting:

Recent discoveries have highlighted previously unrevealed roles of extracellular ENO1 in promoting glycolysis and pro-cancer activities. This discovery represents a significant paradigm shift, as ENO1 enzymatic activity was previously considered exclusive to the cytosolic form . Key research areas include:

  • Development of antibodies specifically targeting extracellular/surface ENO1

  • Investigation of extracellular ENO1's signaling mechanisms beyond HIF-1α

  • Exploration of ENO1's role in tumor microenvironment modulation

ENO1-Hsp70 interaction targeting:

The identification of specific interaction domains between ENO1 and Hsp70 has opened new avenues for therapeutic development:

  • The ENO1 region spanning amino acids 162-282 has been identified as the Hsp70-binding domain

  • This interaction between two multifunctional proteins overexpressed in numerous human cancers represents a promising therapeutic target

  • Structure-based design of small molecules or peptides that disrupt this interaction could provide cancer-specific interventions

Metabolic vulnerability exploitation:

ENO1's critical role in cancer metabolism creates targetable dependencies:

  • ENO1 knockout leads to metabolic reprogramming that impairs tumor progression

  • Identifying synthetic lethal interactions with ENO1 inhibition could reveal combination therapy approaches

  • Understanding compensatory mechanisms following ENO1 targeting will be crucial for developing effective therapeutic strategies

Precision medicine applications:

Emerging research is exploring ENO1's role in predicting treatment responses:

  • Correlation between ENO1 expression and sensitivity to specific therapies

  • Development of ENO1-based companion diagnostics for treatment selection

  • Identification of cancer subtypes particularly dependent on ENO1 function

These research directions collectively represent promising frontiers in translating our understanding of ENO1 biology into effective cancer therapies. The multifunctional nature of ENO1 provides multiple intervention points, from enzymatic activity to protein-protein interactions to extracellular signaling functions.

How might ENO1 research contribute to understanding cancer metabolism heterogeneity?

ENO1 research offers unique insights into the complex metabolic heterogeneity of cancer:

Mechanisms of metabolic adaptation:

Studies of ENO1 have revealed sophisticated mechanisms through which cancer cells adapt their metabolism:

  • ENO1 knockout leads to metabolic reprogramming, demonstrating the plasticity of cancer metabolism

  • Extracellular ENO1 enhances glycolysis through HIF-1α pathway activation, providing an additional regulatory layer beyond direct enzymatic functions

  • This dual mechanism allows cancer cells to modulate glycolysis through both intracellular enzymatic activity and extracellular signaling

Tumor microenvironment influence:

ENO1 research illuminates how cancer metabolism interacts with the tumor microenvironment:

  • Extracellular ENO1 promotes VEGF production, a key mediator of angiogenesis, linking metabolism to vascular remodeling

  • ENO1-induced secretion of tumor-promoting cytokines shapes the immune landscape

  • These findings highlight how metabolic enzymes can influence the tumor microenvironment beyond their canonical functions

Inter- and intra-tumoral heterogeneity:

Comprehensive analysis of ENO1 expression across cancer types reveals patterns of metabolic heterogeneity:

  • Database analyses using TIMER and GEPIA demonstrate differential ENO1 expression patterns across cancer types

  • ENO1's prognostic significance varies between cancer types, reflecting underlying biological differences

  • Single-cell approaches examining ENO1 expression could further reveal intratumoral metabolic heterogeneity

Therapeutic implications:

Understanding ENO1's contribution to metabolic heterogeneity has direct therapeutic relevance:

  • Identification of cancer types particularly dependent on ENO1 function

  • Development of biomarkers to predict response to metabolism-targeting therapies

  • Creation of combination strategies addressing multiple metabolic vulnerabilities

By studying ENO1's multifaceted roles in different cancer contexts, researchers can gain deeper insights into the metabolic adaptability that challenges current therapeutic approaches. These insights may guide the development of more effective metabolism-targeting strategies that account for cancer's metabolic complexity and adaptability.

Product Science Overview

Structure and Function

Enolase-1 is a multifunctional enzyme with a molecular mass of approximately 49.3 kDa . It catalyzes the conversion of 2-phosphoglycerate to phosphoenolpyruvate, a critical step in glycolysis. Beyond its glycolytic function, Enolase-1 also acts as a plasminogen receptor on the cell surface, particularly in tumors, where it contributes to cancer cell proliferation, migration, invasion, and metastasis .

Biological Significance

Enolase-1 is involved in various physiological and pathological processes. It is predominantly expressed during the early stages of embryonic development . In cancer biology, Enolase-1 overexpression and post-translational modifications are of diagnostic and prognostic value in many cancer types . The enzyme’s role in anaerobic metabolism under hypoxic conditions makes it a critical player in tumor progression and survival .

Recombinant Enolase-1

Recombinant human Enolase-1 is produced using DNA sequences encoding the human ENO1 gene, typically expressed in Escherichia coli . The recombinant protein is often tagged with polyhistidine for purification purposes and is verified for purity using techniques like SDS-PAGE and HPLC . It is commonly used in research to study its biochemical properties, interactions, and potential as a therapeutic target.

Applications in Research

Recombinant Enolase-1 is valuable in various research applications, including:

  • Cancer Research: Studying its role in tumor biology and as a potential biomarker for cancer diagnosis and prognosis .
  • Immunology: Investigating its interaction with immune cells, such as CD4+ T cells, and its impact on immune responses .
  • Metabolic Studies: Exploring its function in glycolysis and its regulation under different physiological conditions .
Storage and Stability

Recombinant Enolase-1 is typically lyophilized and stored at -20°C to -80°C to maintain its stability and activity. It is recommended to avoid repeated freeze-thaw cycles to preserve its integrity .

Enolase-1’s multifunctional nature and involvement in critical biological processes make it a significant focus of research, particularly in understanding cancer biology and developing potential therapeutic interventions.

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.