HEXA Antibody

Hexosaminidase A, Mouse Anti Human
Shipped with Ice Packs
In Stock

Description

Definition and Biological Context

HEXA Antibody is a research tool targeting the hexosaminidase A (HEXA) enzyme, encoded by the HEXA gene located on chromosome 15. HEXA is the alpha subunit of beta-hexosaminidase A, a lysosomal enzyme critical for degrading GM2 gangliosides. Mutations in HEXA impair this degradation, leading to GM2 accumulation and Tay-Sachs disease (TSD) .

Enzyme Composition

  • Beta-hexosaminidase A is a heterodimer: α subunit (HEXA) + β subunit (HEXB) .

  • HEXA contains a unique loop structure (Gly-280, Ser-281, Glu-282, Pro-283) and Arg-424 residue, enabling binding to GM2 gangliosides and hydrolysis via GM2 activator protein .

Pathological Relevance

  • Tay-Sachs Disease: Over 210 HEXA mutations disrupt enzyme activity, causing lethal neurodegeneration. Residual enzyme activity ≥10% determines disease severity .

  • Cancer: Microglial HEXA/HEXB overexpression in glioblastoma (GBM) correlates with poor prognosis and promotes tumor progression .

Key Applications

ApplicationDetailsReferences
Western Blot (WB)Detects HEXA at ~54–60 kDa in HepG2, MCF-7, and PC-3 cell lines .
ImmunofluorescenceLocalizes HEXA in lysosomes/cytoplasm of neuronal cells (e.g., human brain hypothalamus) .
ImmunohistochemistryIdentifies HEXA in paraffin-embedded tissues (e.g., GBM microglia, rat testis) .
Therapeutic ResearchEvaluates HEXA-targeted gene therapies and HexaBody® technology for cancer immunotherapy .

Therapeutic Strategies

  • Gene Therapy: Herpes simplex vectors restored beta-hexosaminidase activity in TSD mouse models, reducing GM2 accumulation .

  • HexaBody® Technology:

    • Mechanism: E430G mutation in IgG1 enhances antibody hexamerization, boosting complement-dependent cytotoxicity (CDC) against CD20/CD38+ cancers .

    • Efficacy: HexaBody-CD38 induced complete tumor regression in B-cell lymphoma PDX models and inhibited CD38 enzymatic activity in multiple myeloma .

Biomarker Potential in Cancer

  • GBM: HEXA/HEXB overexpression in microglia promotes tumor proliferation and migration via paracrine signaling .

  • Survival Correlation: High HEXA/HEXB levels in GBM patients predict shorter survival (CGGA/TCGA data) .

Key Mutants and Disease Models

MutantPhenotypeTherapeutic Insights
G269SAdult-onset TSD (impaired folding) Chemical chaperones improved HexA activity in patient fibroblasts .
E482KInfantile TSD (severe activity loss) ER-associated degradation (ERAD) inhibitors rescued mutant α .

Experimental Protocols

  • WB: Dilution range 1:500–1:3000 .

  • IHC/IF: Antigen retrieval with Tris-EDTA (pH 9.0) enhances signal .

Product Specs

Introduction
HEXA is the alpha subunit of the lysosomal enzyme beta-hexosaminidase. This enzyme, along with the GM2 activator protein, breaks down the ganglioside GM2 and other molecules containing N-acetyl hexosamines terminus. Beta-hexosaminidase is composed of alpha and beta subunits, each encoded by separate genes. Mutations in the alpha subunit gene can lead to Tay-Sachs disease (GM2-gangliosidosis type I).
Physical Appearance
Clear, colorless solution that has been sterilized by filtration.
Formulation
The solution contains 1mg/ml of HEXA Antibody in a buffer of PBS at pH 7.4, with 10% Glycerol and 0.02% Sodium Azide.
Storage Procedures
Store at 4°C for up to 1 month. For longer-term storage, keep at -20°C. Avoid repeated freezing and thawing.
Stability / Shelf Life
The product is stable for 12 months when stored at -20°C and for 1 month at 4°C.
Applications
This antibody has been validated for ELISA and Western blot analysis to ensure its specificity and reactivity. However, optimal working dilutions should be determined experimentally for each application. A starting dilution of 1:3000 is recommended.
Synonyms

TSD, hexosaminidase A, Beta-hexosaminidase subunit alpha, Beta-N-acetylhexosaminidase subunit alpha, Hexosaminidase subunit A, N-acetyl-beta-glucosaminidase subunit alpha.

Purification Method
HEXA antibody was purified by protein-A affinity chromatography.
Type
Mouse Anti Human Monoclonal.
Clone
PAT20F1A.
Immunogen
Anti-human HEXA mAb, clone PAT20F1A, is derived from hybridization of mouse F0 myeloma cells with spleen cells from BALB/c mice immunized with a recombinant human HEXA protein 89-529 amino acids purified from E. coli.
Ig Subclass
Mouse IgG2a heavy chain and Lambda light chain.

Q&A

What is Hexosaminidase A (HEXA) and why is it significant in research?

Hexosaminidase A (HEXA) is a critical lysosomal enzyme composed of alpha and beta subunits that catalyzes the degradation of GM2 gangliosides and other glycoconjugates containing terminal N-acetylhexosamine residues. The protein plays an essential role in the normal turnover of gangliosides within the central nervous system. Mutations in the HEXA gene, which encodes the alpha subunit, lead to Tay-Sachs disease, a devastating neurodegenerative disorder characterized by the accumulation of GM2 gangliosides in neurons. HEXA research is significant in understanding lysosomal storage disorders, neurodegenerative diseases, and more recently, in investigating potential roles in cancer progression. The protein has been detected in various human tissues, with particularly notable expression in neuronal cell bodies, specifically in the cytoplasm and lysosomes . Research utilizing HEXA antibodies enables scientists to explore both normal cellular physiology and pathological conditions associated with HEXA dysfunction.

What detection methods are most effective for HEXA using available antibodies?

Multiple detection methods have demonstrated effectiveness for HEXA visualization and quantification, each with specific advantages depending on research objectives. Western blotting effectively detects HEXA in cell and tissue lysates, typically revealing a specific band at approximately 60 kDa when using appropriate antibodies under reducing conditions . Immunohistochemistry (IHC) provides valuable spatial information, with notable results in fixed paraffin-embedded sections of human brain tissue, particularly in the hypothalamus, where specific staining localizes to cytoplasm and lysosomes in neuronal cell bodies . Immunocytochemistry/immunofluorescence offers cellular-level localization, while ELISA provides quantitative analysis for HEXA levels . For optimal results, researchers should employ heat-induced epitope retrieval with appropriate antigen retrieval reagents prior to antibody incubation in IHC applications. Secondary detection systems like HRP-DAB provide robust visualization, while counterstaining with hematoxylin enables clear cellular context identification . Method selection should be guided by specific experimental questions, with consideration for tissue type, fixation method, and required sensitivity.

How should researchers validate the specificity of HEXA antibodies?

Rigorous validation of HEXA antibody specificity requires a multi-pronged approach to ensure experimental reliability. First, Western blot analysis should confirm detection of a single predominant band at the expected molecular weight (~60 kDa) in appropriate positive control samples such as HepG2 cells or human liver tissue . Cross-reactivity testing against related hexosaminidase family members, particularly HEXB, is essential to confirm isoform specificity. Researchers should perform knockout/knockdown validation by comparing staining patterns in HEXA-knockout or siRNA-treated samples versus wild-type controls. Peptide competition assays, where pre-incubating the antibody with purified HEXA protein or immunizing peptide should abolish specific signals, provide further confirmation of specificity. Additionally, comparative analysis using multiple antibody clones targeting different epitopes of HEXA can strengthen validation. For functional specificity, neutralization assays measuring the antibody's ability to inhibit HEXA enzymatic activity (such as the cleavage of 4-Methylumbelliferyl-N-Acetyl-beta-D-glucosaminide) can provide functional validation, with effective antibodies demonstrating a dose-dependent inhibition curve . Comprehensive validation through these complementary approaches ensures reliable research outcomes and reproducibility.

How can researchers optimize immunohistochemistry protocols for HEXA detection in different neural tissues?

Optimizing immunohistochemistry protocols for HEXA detection in neural tissues requires careful consideration of tissue-specific variables and methodological refinements. For human brain tissues, particularly hypothalamus sections, heat-induced epitope retrieval using basic antigen retrieval reagents (pH ≥7.5) significantly enhances epitope accessibility for HEXA antibodies . Optimal antibody concentrations for paraffin-embedded neural tissues typically range between 10-15 μg/mL, with overnight incubation at 4°C maximizing sensitivity while maintaining specificity . For cerebellar tissues with high lipid content, extending the permeabilization step with 0.3% Triton X-100 improves antibody penetration. When examining tissues with potential autofluorescence (such as aged brain samples), researchers should utilize chromogenic detection systems like HRP-DAB rather than fluorescent methods, or implement additional quenching steps. To minimize background in neural tissues with high endogenous peroxidase activity, dual blocking with both hydrogen peroxide and avidin-biotin blocking systems is recommended prior to primary antibody application. For multilabel studies examining HEXA alongside neuronal or glial markers, tyramide signal amplification systems can enhance detection of low-abundance HEXA while enabling multiplexing. Importantly, researchers should always include both positive controls (known HEXA-expressing regions like hypothalamus) and negative controls (primary antibody omission and isotype controls) to validate staining specificity in each neural tissue type.

What are the critical considerations for using HEXA antibodies in studying lysosomal storage disorders?

When employing HEXA antibodies to investigate lysosomal storage disorders, researchers must address several critical methodological considerations to ensure meaningful results. First, sample preparation must be carefully controlled—tissues from Tay-Sachs patients or model systems may demonstrate altered cellular architecture due to accumulated substrates, potentially affecting epitope accessibility and requiring customized antigen retrieval protocols. Comparative quantification between affected and normal tissues demands standardized image acquisition parameters and calibrated analysis methods to accurately measure potential differences in HEXA expression, localization, or processing. When examining potential enzyme replacement therapies, researchers should employ neutralization assays with recombinant HEXA to determine if therapeutic antibodies inhibit rather than restore enzymatic function . The ND50 (neutralization dose) provides a critical metric, with effective therapeutic candidates typically showing minimal neutralization at physiological concentrations. For studies involving HEXA trafficking in disease states, co-localization with lysosomal markers (LAMP1/2) using dual immunolabeling provides essential context for understanding mislocalization patterns. Additionally, researchers must consider species differences when using animal models—mouse Hexa may exhibit different epitope accessibility than human HEXA, requiring antibody validation across species. Finally, enzymatic activity assays using fluorogenic substrates should complement immunodetection methods to correlate protein levels with functional activity, especially important when evaluating therapeutic interventions that may restore protein folding but not necessarily enzymatic function.

How should researchers design experiments to investigate HEXA interactions with other lysosomal proteins?

Designing robust experiments to investigate HEXA interactions with other lysosomal proteins requires a strategic combination of complementary techniques. For co-immunoprecipitation studies, researchers should carefully select lysis conditions that preserve protein interactions while efficiently extracting lysosomal membrane proteins—typically using mild detergents like CHAPS or digitonin rather than stronger detergents like SDS. When performing pull-down assays, the use of recombinant HEXA proteins with GST or His tags facilitates efficient capture of binding partners from cellular lysates . Proximity ligation assays (PLA) offer superior spatial resolution for detecting in situ protein-protein interactions, with positive signals indicating proteins are within 40nm of each other—making this method excellent for confirming lysosomal compartment interactions. For dynamic interaction studies, fluorescence resonance energy transfer (FRET) or bimolecular fluorescence complementation (BiFC) enables visualization of real-time interactions in living cells. When investigating HEXA interactions with potential binding partners, researchers should design comprehensive controls including reverse immunoprecipitation, IgG controls, and validation in multiple cell types relevant to lysosomal storage disorders. For unbiased discovery of novel HEXA-interacting proteins, proximity-dependent biotin identification (BioID) or ascorbate peroxidase (APEX) proximity labeling followed by mass spectrometry analysis provides a powerful approach. Additionally, comparative interaction analysis between wild-type HEXA and disease-associated mutants can reveal how pathogenic mutations disrupt normal protein interactions, potentially explaining disease mechanisms beyond simple loss of enzymatic function.

What are common causes of non-specific binding with HEXA antibodies and how can they be minimized?

Non-specific binding issues with HEXA antibodies typically stem from several identifiable factors that can be systematically addressed. Cross-reactivity with structurally similar lysosomal proteins, particularly HEXB, represents a common challenge since these proteins share sequence homology and similar subcellular localization. To mitigate this, researchers should select antibodies specifically validated against recombinant HEXA protein (Met1-Thr529) rather than shorter peptide sequences that may not fully distinguish between family members . Insufficient blocking often leads to high background, particularly in tissues with high endogenous biotin or protein content like liver samples. This can be addressed by extending blocking time (60-90 minutes) and implementing dual blocking protocols with both protein-based blockers (3-5% BSA or normal serum) and commercial background-reducing agents. In Western blot applications, non-specific bands frequently result from inappropriate buffer selection—researchers should specifically use Immunoblot Buffer Group 1 for HEXA detection under reducing conditions . For immunohistochemistry, extended washing steps (5 washes, 5 minutes each) with 0.1% Tween-20 in PBS significantly reduces background without compromising specific staining. Overfixation of tissues can create artifactual cross-linking that increases non-specific binding; this can be addressed by optimizing fixation time or implementing more rigorous antigen retrieval procedures. Finally, when working with brain tissues that have high lipid content, pre-extraction with detergents or lipid-removing solvents prior to antibody incubation can significantly improve signal-to-noise ratios when detecting HEXA in neuronal populations.

How can researchers resolve contradictory results between different HEXA antibody clones?

Resolving contradictory results between different HEXA antibody clones requires systematic investigation of key experimental variables and antibody characteristics. First, researchers should examine epitope differences—antibodies targeting distinct regions of HEXA may yield different results due to epitope masking in certain cellular contexts or protein conformations. The clone specificity documentation should be consulted to determine if antibodies recognize different domains of the HEXA protein that might be differentially accessible in various experimental conditions . Antibody validation using knockout/knockdown controls provides definitive evidence of specificity for each clone—researchers should prioritize data from clones validated against HEXA-deficient samples. Cross-reactivity testing against HEXB is particularly important as some clones may detect both isoforms, leading to different staining patterns in tissues with variable HEXA/HEXB ratios. Technical factors often contribute to discrepancies, including differences in optimal antibody concentration, incubation time, and antigen retrieval methods—systematic optimization of these parameters for each clone is essential. Researchers should also compare antibody performance across multiple detection methods, as some clones perform better in Western blotting while others excel in immunohistochemistry. When antibodies yield different subcellular localization patterns, co-localization studies with established compartment markers can determine which clone accurately reflects the expected lysosomal distribution of HEXA. Finally, contradictory results should be evaluated in the context of biological variables, including potential detection of different post-translational modifications, splice variants, or proteolytically processed forms of HEXA that may have genuine biological significance rather than representing technical artifacts.

What quality control metrics should be implemented when using HEXA antibodies for quantitative analysis?

Implementing rigorous quality control metrics is essential when using HEXA antibodies for quantitative analyses to ensure reproducibility and reliability. Standard curve validation using recombinant HEXA protein at concentrations spanning the expected physiological range (typically 0.1-100 ng/mL) should demonstrate linearity with R² > 0.98 and consistent slope across multiple experiments. Researchers must determine the limit of detection (LOD) and limit of quantification (LOQ) for each antibody batch by analyzing serial dilutions of positive control samples, with values typically 3× and 10× standard deviation of the blank, respectively. Inter-assay and intra-assay coefficients of variation should be calculated from multiple replicate measurements, with acceptable values being <15% for intra-assay and <20% for inter-assay variability. When analyzing tissue samples, normalization strategies must be carefully selected—for Western blot quantification, housekeeping proteins like GAPDH may be unreliable in certain disease states, making total protein normalization (using stain-free gels or Ponceau staining) more appropriate. For immunohistochemical quantification, standardized acquisition parameters, including exposure time, gain settings, and threshold values, must be established and maintained across all samples. Reference standards should be included in each experimental run to account for day-to-day variations in staining intensity or detection sensitivity. Finally, researchers should implement blinded analysis protocols where the investigator performing quantification is unaware of sample identity, particularly in comparative studies between normal and disease tissues, to eliminate unconscious bias in region selection or intensity measurement.

How can HEXA antibodies be utilized to study beta-hexosaminidase trafficking in neurodegenerative disorders?

HEXA antibodies provide powerful tools for investigating beta-hexosaminidase trafficking disruptions in neurodegenerative disorders, particularly when employed within sophisticated experimental paradigms. For studying real-time trafficking dynamics, researchers can conjugate purified HEXA antibodies directly to pH-sensitive fluorophores (pHrodo™), enabling visualization of enzyme movement from neutral pH environments (endoplasmic reticulum, Golgi) to acidic compartments (lysosomes) in living neurons. When investigating mislocalization patterns in disease states, triple immunolabeling combining HEXA antibodies with both ER markers (calnexin) and lysosomal markers (LAMP1) in the same sample allows quantification of retention ratios between compartments. For analyzing disease-specific trafficking defects, patient-derived induced pluripotent stem cells (iPSCs) differentiated into neurons provide an ideal model system, with quantitative immunofluorescence using HEXA antibodies revealing mutation-specific patterns of mislocalization . In pulse-chase experiments, biotinylated HEXA antibodies applied to permeabilized cells at defined time points can track cohorts of newly synthesized enzyme through the secretory pathway. Advanced microscopy techniques including super-resolution microscopy (STED, STORM) with HEXA antibodies enable visualization of nanoscale trafficking intermediates not resolvable with conventional microscopy. For in vivo tracking in animal models, intracerebroventricular injection of fluorescently-labeled HEXA antibody fragments (Fab) allows monitoring of enzyme distribution across brain regions, particularly valuable when evaluating enzyme replacement therapies. Correlative light and electron microscopy (CLEM) combining immunofluorescence with ultrastructural analysis provides unprecedented detail regarding HEXA trafficking through specialized endolysosomal compartments in neurons, revealing structural abnormalities in vesicular transport machinery associated with specific neurodegenerative conditions.

What approaches can researchers use to study the functional relationship between HEXA antibodies and complement-dependent cytotoxicity?

Investigating the functional relationship between HEXA antibodies and complement-dependent cytotoxicity (CDC) requires specialized approaches that bridge basic antibody characterization with advanced immunological mechanisms. The HexaBody technology represents a biology-driven Fc-engineering strategy that enhances the natural hexamerization of antibodies on cell surfaces, significantly improving C1q binding and CDC activity . To evaluate CDC potency, researchers can conduct complement-mediated cell lysis assays using target cells expressing HEXA with human serum as a complement source, measuring cell death through propidium iodide uptake or lactate dehydrogenase release. Flow cytometry-based complement deposition assays quantify C1q, C4b, and C3b binding to antibody-coated cells, providing mechanistic insight into the CDC activation cascade. For structure-function analysis, researchers should compare wild-type IgG1 antibodies with Fc-engineered variants containing specific point mutations (E345R and E430G) that enhance non-covalent Fc interactions and facilitate hexamerization . Real-time confocal microscopy with fluorescently-labeled complement components allows visualization of membrane attack complex formation following HEXA antibody binding. Importantly, researchers must distinguish between antigen-dependent and antigen-independent complement activation by comparing activation in the presence of HEXA-expressing cells versus in solution alone—effective HexaBodies demonstrate enhanced CDC without target-independent complement activation . For therapeutic applications, neutralization assays measuring the antibody's ability to inhibit enzymatic activity of HEXA provide critical functional data, with the neutralization dose (ND50) serving as a key metric of potency . Comprehensive characterization requires evaluation across multiple cellular models with varying HEXA expression levels to determine how antigen density affects the relationship between antibody binding and complement activation.

How can HEXA antibodies contribute to developing novel therapeutic strategies for lysosomal storage disorders?

HEXA antibodies play multifaceted roles in developing innovative therapeutic strategies for lysosomal storage disorders, particularly Tay-Sachs and related conditions. For enzyme replacement therapy (ERT) development, neutralization assays using monoclonal antibodies help identify therapeutic antibody candidates that preserve rather than inhibit HEXA enzymatic activity, with effective candidates demonstrating minimal neutralization of substrate cleavage . Internalization studies tracking fluorescently-labeled HEXA antibodies in neuronal cultures reveal receptor-mediated uptake mechanisms that can be exploited for enhanced delivery of replacement enzymes across the blood-brain barrier. In gene therapy approaches, HEXA antibodies provide essential tools for validating transgene expression in preclinical models, with immunohistochemical analysis confirming successful transduction patterns across treated brain regions . For pharmacological chaperone development, researchers can employ conformation-specific HEXA antibodies that distinguish between properly folded and misfolded variants, enabling high-throughput screening of compounds that promote correct protein folding. Biodistribution studies using radiolabeled HEXA antibodies in animal models provide crucial pharmacokinetic data regarding enzyme distribution following various administration routes. When developing immunomodulatory approaches, HEXA-specific antibodies can be engineered with modified Fc regions (HexaBody technology) that enhance complement-dependent cytotoxicity against infiltrating inflammatory cells in the CNS while sparing neurons . For substrate reduction therapy evaluation, quantitative immunoassays using HEXA antibodies help monitor changes in enzyme levels following treatment with inhibitors of ganglioside synthesis. Finally, HEXA antibodies enable development of companion diagnostics for patient stratification, with immunoassays detecting specific mutant forms of HEXA that might respond differentially to personalized therapeutic interventions.

What emerging technologies are enhancing the research applications of HEXA antibodies?

Cutting-edge technologies are dramatically expanding the research capabilities of HEXA antibodies beyond traditional applications. Single-cell proteomics using mass cytometry (CyTOF) with metal-conjugated HEXA antibodies enables simultaneous quantification of HEXA expression alongside dozens of other proteins at single-cell resolution, revealing previously unrecognized cellular heterogeneity in disease states. Proximity extension assays (PEA) utilizing paired HEXA antibodies conjugated to complementary oligonucleotides achieve femtomolar detection sensitivity, allowing quantification of HEXA in limited biological samples like cerebrospinal fluid. For spatial transcriptomics integration, in situ sequential immunofluorescence with HEXA antibodies combined with RNA sequencing provides unprecedented correlation between protein localization and gene expression patterns in complex neural tissues. The HexaBody technology represents a revolutionary approach that enhances the natural hexamerization of antibodies through specific point mutations (E345R, E430G) in the Fc domain, significantly improving complement-dependent cytotoxicity—particularly valuable for developing therapeutic antibodies . Antibody-drug conjugates (ADCs) utilizing HEXA-targeting antibodies enable selective delivery of therapeutic payloads to cells with abnormal HEXA expression or localization. Intrabodies generated from HEXA antibody sequences and expressed within cells allow real-time tracking of endogenous HEXA trafficking in living neurons. For clinical applications, automated digital pathology platforms incorporating machine learning algorithms trained on HEXA immunohistochemistry patterns can identify subtle alterations in enzyme distribution that correlate with disease progression. Finally, programmable biosensors using HEXA antibody fragments coupled to CRISPR-Cas systems enable molecular recording of enzyme activity fluctuations over time, providing dynamic information not accessible through traditional static immunodetection methods.

How can researchers reconcile contradictory findings in HEXA expression across different experimental models?

Reconciling contradictory findings regarding HEXA expression across different experimental models requires systematic evaluation of biological, technical, and analytical variables. Species-specific differences represent a primary consideration—human and mouse HEXA share only 73% sequence identity, potentially affecting antibody cross-reactivity and leading to apparent discrepancies when using the same antibody across species . Cell-type specificity significantly impacts HEXA expression patterns; neuronal populations typically show distinct subcellular localization compared to glial or peripheral cells, necessitating careful consideration of cellular composition when comparing tissues or primary cultures. Developmental regulation of HEXA expression creates temporal variables—findings in embryonic models may contradict adult expression patterns, requiring age-matched comparisons. At the technical level, methodological differences in sample preparation significantly impact detection—aldehyde fixation for immunohistochemistry may mask epitopes that remain accessible in frozen sections or Western blot samples prepared with detergent extraction . Different antibody clones recognizing distinct epitopes can yield contradictory results if certain epitopes are differentially accessible across experimental systems. Quantification approaches also contribute to discrepancies—pixel intensity-based measurements in immunofluorescence versus band densitometry in Western blots may not correlate directly. Researchers should implement standardized protocols including matched antibody concentrations, identical incubation conditions, and consistent image acquisition parameters to minimize technical variability. When discrepancies persist, orthogonal validation using antibody-independent methods such as RNA-seq for transcript levels, mass spectrometry for protein quantification, or enzymatic activity assays provide critical corroborating evidence. Finally, researchers should consider disease-specific alterations—pathological conditions may fundamentally alter HEXA processing, localization, or antibody accessibility, creating genuine biological differences rather than technical artifacts.

Product Science Overview

Introduction

Hexosaminidase A (HEXA) is an enzyme that plays a crucial role in the degradation of glycosphingolipids, specifically GM2 gangliosides, within lysosomes. This enzyme is composed of two subunits, alpha and beta, which are encoded by the HEXA and HEXB genes, respectively . The enzyme’s primary function is to hydrolyze terminal N-acetyl-D-hexosamine residues in GM2 gangliosides and globo-sphingolipids .

Importance in Human Health

Deficiencies in HEXA activity lead to the accumulation of GM2 gangliosides, resulting in severe neurodegenerative disorders such as Tay-Sachs disease and Sandhoff disease . These lysosomal storage diseases are characterized by progressive neurological deterioration, leading to early mortality in affected individuals .

Mouse Anti-Human Hexosaminidase A Antibody

The mouse anti-human Hexosaminidase A antibody is a monoclonal antibody that specifically detects human HEXA in various applications, including Western blotting and immunohistochemistry . This antibody is produced by immunizing mice with recombinant human HEXA protein, followed by hybridoma technology to generate a specific monoclonal antibody .

Applications
  1. Western Blotting: The mouse anti-human HEXA antibody is used to detect HEXA in cell lysates and tissue samples. It is particularly useful in identifying the presence and quantity of HEXA in different biological samples .
  2. Immunohistochemistry: This antibody is employed to visualize HEXA expression in tissue sections, providing insights into the localization and distribution of HEXA within cells and tissues .
Research Significance

Research involving HEXA and its antibodies has significant implications for understanding and treating lysosomal storage diseases. By studying the expression and activity of HEXA, scientists can develop therapeutic strategies to enhance HEXA function and mitigate the effects of GM2 ganglioside accumulation .

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.