ENGASE1 Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
Preservative: 0.03% ProClin 300; Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
14-16 weeks (Made-to-order)
Synonyms
ENGASE1 antibody; ENGASE85A antibody; At5g05460 antibody; K18I23.27Cytosolic endo-beta-N-acetylglucosaminidase 1 antibody; ENGase 1 antibody; EC 3.2.1.96 antibody; Endo-beta-N-acetyglucosaminidase 85A antibody; AtENGase85A antibody
Target Names
ENGASE1
Uniprot No.

Target Background

Function
Endoglycosidase ENGASE1 cleaves the β-1,4-glycosidic bond within the N,N'-diacetylchitobiose core of N-glycans, releasing them from glycoproteins. This enzyme plays a crucial role in the biosynthesis of high-mannose type N-glycans during plant development and fruit maturation.
Gene References Into Functions
Further research supports this role: Analysis of an *Arabidopsis thaliana* mutant with suppressed expression of two putative ENGase genes (PMID: 21597164).
Database Links

KEGG: ath:AT5G05460

STRING: 3702.AT5G05460.1

UniGene: At.32993

Protein Families
Glycosyl hydrolase 85 family
Subcellular Location
Cytoplasm, cytosol.

Q&A

What is ENO1 and why is it a significant target for antibody development?

ENO1 (alpha-enolase) is a multifunctional protein that serves as both a glycolytic enzyme and a plasminogen receptor when expressed on cell surfaces. Its significance as an antibody target stems from its involvement in multiple pathological processes. In cancer, ENO1 is highly expressed on cell membranes, in the cytoplasm, and nuclei of tumor cells, where it promotes migration, invasion, and tumorigenesis. Upon oncogenic or inflammatory stimulation, tumor cells or immune cells express cell surface ENO1 as a plasminogen receptor to facilitate their migration via plasmin activation . Additionally, ENO1 has been found to play important roles in autoimmune diseases and can cause neurological damage when autoantibodies against it are present . This multifaceted involvement in disease processes makes ENO1 an ideal target for therapeutic antibody development.

What are the different types of ENO1 antibodies available for research purposes?

Based on the research literature, there are several types of ENO1 antibodies that have been developed for research applications:

  • Monoclonal ENO1 antibodies (ENO1mAb): These are highly specific antibodies developed using hybridoma technology that target specific epitopes on the ENO1 protein. Examples include HuL227, a humanized ENO1 mAb that has shown promising results in prostate cancer research .

  • ENO1-specific autoantibodies (ENO1Ab): These are naturally occurring or experimentally induced antibodies against ENO1 that have been studied in the context of autoimmune diseases and neurological damage .

  • Conjugated ENO1 antibodies: Various conjugated forms have been developed for specific research applications, similar to how other antibodies are modified (though specific ENO1 conjugations weren't detailed in the provided search results).

The selection of the appropriate antibody type depends on the specific research question, experimental design, and intended application.

How does ENO1 antibody function in blocking cancer cell migration and invasion?

ENO1 antibody functions in blocking cancer cell migration and invasion through several mechanisms:

  • Inhibition of the ENO1/plasminogen axis: When expressed on cell surfaces, ENO1 acts as a plasminogen receptor. The binding of plasminogen to ENO1 facilitates its conversion to plasmin, which degrades extracellular matrix components and promotes cell migration. ENO1 antibodies block this interaction, thereby inhibiting the migration and invasion capabilities of cancer cells .

  • Suppression of tumor microenvironment (TME) components: In prostate cancer research, ENO1 antibody (HuL227) has been shown to reduce monocyte recruitment and intratumoral angiogenesis. It also inhibits inflammation-enhanced osteoclast activity and the secretion of invasion-related cytokines such as CCL2 and TGFβ from osteoclasts .

  • Inhibition of VEGF-A-induced tube formation: ENO1 antibody reduces VEGF-A-induced tube formation of endothelial cells, which is crucial for angiogenesis and tumor growth .

These mechanisms collectively contribute to the anti-cancer effects of ENO1 antibodies by targeting multiple aspects of tumor progression and metastasis.

How can ENO1 antibodies be utilized to target multiple players in the tumor microenvironment?

ENO1 antibodies demonstrate remarkable versatility in targeting multiple components of the tumor microenvironment (TME), making them potentially powerful therapeutic agents:

  • Simultaneous targeting of multiple cell types: Research has shown that ENO1 is functionally expressed on the surface of various cell types within the TME, including cancer cells, monocytes, osteoclasts, and endothelial cells. ENO1 antibodies can simultaneously target all these cell types, providing a multifaceted approach to cancer therapy .

  • Inhibition of angiogenesis: ENO1 antibody (HuL227) has been demonstrated to significantly reduce VEGF-A–induced tube formation of endothelial cells in vitro and intratumoral angiogenesis in vivo. This anti-angiogenic effect contributes to reduced tumor growth and metastasis .

  • Suppression of inflammatory responses: ENO1 antibodies inhibit inflammation-enhanced osteoclast activity and reduce the secretion of pro-invasive cytokines. This immunomodulatory effect is particularly relevant for cancers with inflammatory components .

  • Disruption of metastatic processes: In prostate cancer models, ENO1 antibody has been shown to reduce tumor growth and osteoclast activation in bone, suggesting its potential in preventing bone metastasis. This is particularly significant as bone is a common site for prostate cancer metastasis .

These multifactorial effects make ENO1 antibodies promising candidates for targeting complex tumor microenvironments, particularly in advanced cancers where multiple cellular interactions drive disease progression.

What are the implications of maternal anti-ENO1 antibodies crossing the blood-brain barrier?

Research has revealed concerning implications regarding maternal anti-ENO1 antibodies crossing the blood-brain barrier and affecting fetal development:

  • Neurological development impairment: Studies using pregnant mouse models with high serum ENO1-specific antibody (ENO1Ab) have demonstrated that maternal ENO1Ab can pass through both the blood-placenta barrier and the compromised blood-brain barrier into the brain tissues of offspring. This transmission is associated with impaired learning and memory abilities in the offspring .

  • Complement-dependent cytotoxicity: Once in the brain tissue, ENO1Ab deposits colocalize with ENO1 protein and complement component 3 (C3). The membrane attack complex (MAC) becomes detectable in the brain tissues of offspring from dams with high serum ENO1Ab, suggesting that neurological damage occurs primarily through complement-dependent cytotoxicity .

  • Structural changes in the brain: Pups from mothers with high ENO1Ab showed obviously thinner tight junctions in the brain tissue, indicating structural damage to the blood-brain barrier. This could potentially lead to increased permeability and further neurological complications .

  • Long-term cognitive effects: The learning and memory impairments observed in the water maze test and decreased long-term potentiation suggest that maternal ENO1Ab exposure may have lasting effects on cognitive function .

These findings have significant implications for understanding the potential role of maternal autoantibodies in neurological disorders in offspring and suggest that monitoring ENO1Ab levels during pregnancy might be important in certain clinical contexts.

How do ENO1 antibodies interact with the glycolytic functions of ENO1 in cancer cells?

ENO1 antibodies demonstrate a dual mechanism of action by targeting both the surface receptor function and the enzymatic glycolytic activity of ENO1 in cancer cells:

  • Inhibition of glycolytic enzyme activity: When delivered intracellularly (such as via nanoparticle carriers), ENO1 antibodies can antagonize the enzymatic activity of ENO1, which is a critical enzyme in glycolysis. Research has shown that ENO1 monoclonal antibodies delivered via PLGA nanoparticles significantly decreased the levels of lactic acid and pyruvate in cervical cancer cells, indicating impaired glycolytic function .

  • Metabolic reprogramming effects: By inhibiting ENO1's glycolytic activity, these antibodies can potentially reverse the Warburg effect (preferential use of glycolysis even in the presence of oxygen) that is characteristic of many cancer cells. This metabolic reprogramming may contribute to the anti-proliferative effects observed in cancer cell lines .

  • Dual targeting approach: The ability to simultaneously target both the surface receptor function (inhibiting migration and invasion) and the intracellular enzymatic function (inhibiting glycolysis) provides ENO1 antibodies with a powerful dual mechanism for cancer therapy .

  • Delivery systems for intracellular targeting: Research has demonstrated that folic acid-modified PLGA nanoparticles can effectively deliver ENO1 antibodies into tumor cells, allowing them to target the intracellular glycolytic functions of ENO1. This approach significantly inhibited proliferation, migration, and clone formation of cervical cancer cells compared to control groups .

This dual targeting approach represents an advanced application of ENO1 antibodies that could potentially overcome resistance mechanisms and enhance therapeutic efficacy in cancer treatment.

What are the optimal methods for producing and purifying ENO1 antibodies?

Based on the research literature, several methodological approaches have been validated for producing and purifying high-quality ENO1 antibodies:

  • Recombinant ENO1 protein production: The ENO1 gene can be cloned into expression vectors such as pFastBac1 and expressed in insect cell systems (e.g., Sf9 cells). This approach allows for the production of properly folded human ENO1 protein that can be used for immunization .

  • Hybridoma technology for monoclonal antibody production:

    • Immunization protocol: BALB/c mice can be repeatedly immunized with purified ENO1 protein to develop a robust immune response .

    • Cell fusion: Spleen cells from immunized mice with high antibody titers are fused with Sp2/0 myeloma cells to create hybridoma cell lines .

    • Screening: ELISA can be used to determine the levels of IgG antibody against ENO1 protein in the culture supernatant of hybridoma cells to identify high-titer clones .

  • Antibody purification methods:

    • Protein A/G affinity chromatography is commonly used for purification of IgG antibodies.

    • Size exclusion chromatography can be employed as a polishing step to achieve high purity.

  • Functional screening approaches: Beyond titer-based screening, functional assays can be incorporated to select antibodies with desired biological activities. For example, evaluating the inhibitory effect on migration and invasion of cancer cells can help identify ENO1 antibodies that effectively block cell surface ENO1 .

These methodological considerations are critical for ensuring the production of high-quality ENO1 antibodies with the desired specificity and functional properties for research applications.

What delivery systems can be used to target ENO1 antibodies to intracellular locations?

Effective intracellular delivery is crucial for targeting the glycolytic function of ENO1. The research literature highlights several promising approaches:

  • Folic acid-conjugated PLGA nanoparticles: Research has demonstrated the effectiveness of folic acid (FA)-modified poly(lactic-co-glycolic acid) (PLGA) nanoparticles for delivering ENO1 antibodies into cancer cells. These nanoparticles leverage the overexpression of folate receptors on many cancer cells to achieve targeted delivery .

  • Disulfide bond-based release systems: FA-SS-PLGA nanoparticles containing a disulfide bond enable selective release of the antibody cargo in the reducing environment of the cytoplasm. This system has been successfully used to deliver ENO1 monoclonal antibodies into cervical cancer cells .

  • Optimization of nanoparticle properties:

    • Size: Nanoparticles in the range of 50-200 nm typically show optimal cellular uptake.

    • Surface charge: Slightly positive charge can enhance interaction with the negatively charged cell membrane.

    • Antibody loading capacity: Optimizing the antibody-to-polymer ratio is critical for efficient delivery.

  • Validation of intracellular delivery:

    • Functional assays measuring glycolytic parameters (such as lactic acid and pyruvate levels) can confirm successful delivery and activity of the intracellular antibody .

    • Fluorescently labeled antibodies can be used to track intracellular localization.

These delivery approaches enable researchers to target both the surface receptor function and the intracellular enzymatic function of ENO1, representing an advancement in antibody-based therapeutic strategies for cancer.

What are the key experimental models for evaluating ENO1 antibody efficacy in cancer research?

The research literature describes several experimental models that have proven valuable for evaluating the efficacy of ENO1 antibodies:

  • In vitro cellular models:

    • Migration and invasion assays: These assess the ability of ENO1 antibodies to inhibit cancer cell migration and invasion, which are key processes in metastasis .

    • Tube formation assays: These evaluate the effect of ENO1 antibodies on VEGF-A-induced angiogenesis by measuring tube formation in endothelial cells .

    • Glycolytic function assays: Measuring lactic acid and pyruvate levels can assess the impact of ENO1 antibodies on cancer cell metabolism .

    • Clone formation assays: These evaluate the long-term proliferative capacity of cancer cells following ENO1 antibody treatment .

  • In vivo xenograft models:

    • Subcutaneous xenograft models: These have been used to demonstrate that ENO1 antibodies (e.g., HuL227) can reduce tumor growth, monocyte recruitment, and intratumoral angiogenesis .

    • Orthotopic implantation models: These can provide insights into the effects of ENO1 antibodies in the context of the native tumor microenvironment.

  • Metastasis models:

    • Intratibial implantation models: In prostate cancer research, this model has been used to demonstrate that ENO1 antibody reduces tumor growth and osteoclast activation in bone, highlighting its potential in preventing bone metastasis .

  • Combination therapy models:

    • Evaluating ENO1 antibodies in combination with standard-of-care treatments can provide valuable insights into potential synergistic effects.

These experimental models provide a comprehensive framework for evaluating the multifaceted effects of ENO1 antibodies on cancer cells, the tumor microenvironment, and metastatic processes.

What are common challenges in ENO1 antibody specificity and how can they be addressed?

Researchers working with ENO1 antibodies may encounter several specificity challenges that can affect experimental outcomes:

  • Cross-reactivity with other enolase isoforms: ENO1 shares sequence homology with other enolase isoforms (ENO2 and ENO3), which may lead to cross-reactivity. To address this:

    • Perform western blot analysis against all three enolase isoforms to confirm specificity.

    • Use knockout/knockdown cell lines as negative controls to validate antibody specificity.

    • Consider epitope mapping to identify antibodies targeting unique regions of ENO1.

  • Distinguishing between surface and intracellular ENO1: Since ENO1 functions both as a cytoplasmic enzyme and a cell surface receptor, it is critical to distinguish between these pools:

    • For surface ENO1 detection, use non-permeabilized cells in flow cytometry or immunofluorescence.

    • For total ENO1 detection, use permeabilized cells or cell lysates.

    • Consider using subcellular fractionation to separate membrane and cytoplasmic fractions before antibody application.

  • Validation in relevant biological contexts:

    • Confirm antibody recognition of native ENO1 through immunoprecipitation experiments.

    • Validate antibody function through inhibition assays (e.g., migration, invasion, or enzymatic activity).

    • Use multiple antibody clones targeting different epitopes to confirm results.

  • Optimizing antibody concentration:

    • Perform titration experiments to determine the optimal antibody concentration that provides specific signal with minimal background.

    • Include appropriate isotype controls at the same concentration as the primary antibody.

Addressing these specificity challenges is crucial for generating reliable and reproducible results in ENO1 antibody research.

How can researchers overcome challenges in intracellular delivery of ENO1 antibodies?

Successful intracellular delivery of ENO1 antibodies presents several technical challenges that researchers can address through the following approaches:

  • Optimizing nanoparticle formulation parameters:

    • Particle size: Adjust polymer concentration and sonication parameters to achieve optimal size (typically 50-200 nm) for cellular uptake.

    • Surface charge: Modify the zeta potential to enhance cell membrane interaction without causing cytotoxicity.

    • Antibody loading: Optimize the antibody-to-polymer ratio and encapsulation techniques to maximize loading efficiency while preserving antibody activity .

  • Enhancing endosomal escape:

    • Incorporate endosomal escape enhancers (e.g., pH-sensitive polymers or fusogenic peptides) into nanoparticle formulations to prevent lysosomal degradation of antibodies.

    • Consider using disulfide-based linkages that can be cleaved in the reducing environment of the cytoplasm, as demonstrated with FA-SS-PLGA nanoparticles .

  • Confirming successful intracellular delivery:

    • Use confocal microscopy with fluorescently labeled antibodies to visualize intracellular localization.

    • Perform subcellular fractionation followed by western blotting to confirm cytoplasmic localization.

    • Conduct functional assays (e.g., measuring glycolytic parameters) to confirm that delivered antibodies maintain their activity .

  • Addressing cell type-specific challenges:

    • Adjust targeting ligands based on cell surface receptor expression (e.g., folate receptor for cancer cells).

    • Optimize transfection conditions for different cell types based on their endocytic capacity.

    • Consider cell-specific barriers (e.g., mucus layers for epithelial cells) when designing delivery systems.

By systematically addressing these challenges, researchers can improve the efficiency and reproducibility of intracellular ENO1 antibody delivery for both research and potential therapeutic applications.

What controls should be included in ENO1 antibody experiments for reliable interpretation of results?

Proper experimental controls are essential for generating reliable and interpretable results when working with ENO1 antibodies:

  • Antibody specificity controls:

    • Isotype control antibodies: Match the isotype, concentration, and conjugation of the ENO1 antibody to control for non-specific binding.

    • ENO1 knockdown/knockout cells: These serve as negative controls to validate antibody specificity.

    • Pre-absorption controls: Pre-incubating the antibody with recombinant ENO1 protein should abolish specific staining.

  • Functional assay controls:

    • Positive control antibodies: Include antibodies with known inhibitory effects on the processes being studied.

    • Concentration gradients: Test multiple concentrations of ENO1 antibodies to establish dose-response relationships.

    • Time course experiments: Evaluate temporal effects to distinguish between direct and indirect antibody effects.

  • Delivery system controls:

    • Empty nanoparticles: Control for potential effects of the delivery system itself.

    • Non-targeting antibodies in the same delivery system: Control for non-specific effects of antibody delivery.

    • Free antibody (non-encapsulated): Compare with nanoparticle-delivered antibody to confirm delivery advantages .

  • In vivo experimental controls:

    • Vehicle control groups: Animals receiving only the vehicle without antibody.

    • Non-specific antibody groups: Animals receiving isotype-matched control antibodies.

    • Untreated control groups: Animals receiving no treatment to establish baseline parameters.

    • Positive control groups: Animals receiving standard treatments with known effects .

  • Validation across multiple experimental systems:

    • Confirm key findings using different cell lines or animal models.

    • Use complementary techniques to validate observations (e.g., both in vitro and in vivo approaches).

Incorporating these controls systematically will enhance the rigor and reproducibility of ENO1 antibody research and facilitate more reliable interpretation of experimental results.

What are emerging applications of ENO1 antibodies beyond cancer research?

While much of the current research focuses on cancer applications, ENO1 antibodies show promise in several emerging areas:

  • Autoimmune disease research and diagnostics: The discovery that ENO1 autoantibodies play a role in autoimmune diseases opens opportunities for diagnostic applications and mechanistic studies. Research has shown that anti-ENO1 autoantibodies can cause neurological damage, suggesting potential applications in autoimmune neurological disorders .

  • Neurodevelopmental disorder investigations: The finding that maternal ENO1 antibodies can cross the placental barrier and affect fetal brain development suggests potential applications in studying mechanisms of neurodevelopmental disorders. This could lead to diagnostic tools for identifying at-risk pregnancies or therapeutic approaches to prevent antibody-mediated neurological damage .

  • Inflammatory disease modulation: ENO1 antibodies have shown anti-inflammatory effects by reducing monocyte recruitment and inhibiting inflammation-enhanced osteoclast activity. These properties could be exploited for therapeutic applications in inflammatory diseases beyond cancer .

  • Metabolic disorder applications: Given ENO1's role in glycolysis, antibodies targeting its enzymatic function could have applications in metabolic disorders characterized by dysregulated glycolysis.

  • Biomarker development: ENO1 antibody levels could serve as biomarkers for disease progression or treatment response in various conditions, including autoimmune diseases and cancer.

These emerging applications highlight the versatility of ENO1 antibodies beyond their current focus in cancer research and suggest promising new research directions.

What are the prospects for developing ENO1 antibodies as therapeutic agents?

The development of ENO1 antibodies as therapeutic agents shows considerable promise, with several key considerations for future advancement:

  • Multifunctional therapeutic potential:

    • The ability of ENO1 antibodies to simultaneously target multiple aspects of disease pathology (cell migration, invasion, angiogenesis, glycolysis) provides unique therapeutic advantages .

    • HuL227, a humanized ENO1 monoclonal antibody, has shown promising results in preclinical models of prostate cancer, demonstrating effects on both the primary tumor and metastatic processes .

  • Delivery system innovations:

    • The development of targeted nanoparticle delivery systems, such as FA-SS-PLGA nanoparticles, enhances the potential for intracellular delivery of ENO1 antibodies .

    • Future advancements in delivery technologies may further improve the efficacy and specificity of ENO1 antibody therapeutics.

  • Combination therapy approaches:

    • ENO1 antibodies could potentially be combined with standard-of-care treatments to enhance efficacy or overcome resistance mechanisms.

    • The multiple mechanisms of action of ENO1 antibodies suggest potential synergies with other targeted therapies.

  • Translational considerations:

    • Humanization of ENO1 antibodies (as demonstrated with HuL227) will be crucial for reducing immunogenicity in clinical applications .

    • Optimization of dosing regimens, administration routes, and formulation stability will be important for clinical translation.

    • Safety profiling, particularly regarding potential autoimmune effects, will be critical given ENO1's physiological roles.

  • Expanding therapeutic indications:

    • Beyond the currently studied prostate and cervical cancers, ENO1 antibodies may have applications in other cancer types with elevated ENO1 expression.

    • The potential role in autoimmune diseases and neurological disorders suggests additional therapeutic applications beyond oncology .

These prospects highlight the significant potential of ENO1 antibodies as therapeutic agents, while also acknowledging the challenges that must be addressed in their development for clinical applications.

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.