NAGPA Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Biological Role of NAGPA

NAGPA, also termed "uncovering enzyme" (UCE), catalyzes the second step in forming the mannose 6-phosphate (M6P) targeting signal on lysosomal hydrolases. By removing N-acetylglucosamine residues, it ensures proper trafficking of enzymes to lysosomes via M6P receptors in the Golgi apparatus . Dysregulation of NAGPA is linked to lysosomal storage disorders and developmental stuttering .

NAGPA Antibody Types and Applications

NAGPA antibodies are available in polyclonal, monoclonal, and recombinant formats, tailored for techniques such as:

  • Immunohistochemistry (IHC)

  • Western Blot (WB)

  • Enzyme-Linked Immunosorbent Assay (ELISA)

  • Immunofluorescence (IF)

Table 1: Representative NAGPA Antibodies

Product IDHost SpeciesClonalityApplicationsConjugateReactivitySource
ABIN7160684RabbitPolyclonalELISA, WB, IHCUnconjugatedHumanAntibodies-Online
NBP3-05813RabbitPolyclonalIHC-ParaffinUnconjugatedHumanBio-Techne
DPABH-13159RabbitPolyclonalWBUnconjugatedHuman (aa 146-195)Creative-Diagnostics

Key Research Findings

  • Clinical Relevance: NAGPA mutations disrupt lysosomal enzyme sorting, contributing to disorders like mucopolysaccharidosis .

  • Technical Validation:

    • IHC: Antibody NBP3-05813 demonstrated robust staining in human kidney tissues, confirming NAGPA’s Golgi localization .

    • Neutralization: While not directly antiviral, NAGPA antibodies share design principles with broadly neutralizing antibodies (bNAbs) like HIV-targeting N6, which utilize high somatic hypermutation for potency .

Challenges and Innovations

  • Specificity: Early polyclonal antisera exhibited cross-reactivity with structurally similar molecules (e.g., N-acetyl-aspartate), necessitating monoclonal variants for precision .

  • Engineering: Recombinant alpaca-derived secondary antibodies (e.g., CABT-L4215) improve signal-to-noise ratios in NAGPA detection .

Future Directions

Research priorities include optimizing NAGPA antibodies for high-throughput screens and therapeutic applications, particularly in lysosomal disorder diagnostics. Advances in phage display and single-cell sequencing may yield next-generation reagents with enhanced affinity and reduced off-target binding .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship your order within 1-3 business days after receiving it. The delivery time may vary depending on the shipping method and destination. Please consult your local distributor for specific delivery details.
Synonyms
Mannose 6-phosphate-uncovering enzyme antibody; N-acetylglucosamine-1-phosphodiester alpha-N-acetylglucosaminidase antibody; NAGPA antibody; NAGPA_HUMAN antibody; Phosphodiester alpha-GlcNAcase antibody
Target Names
NAGPA
Uniprot No.

Target Background

Function
NAGPA antibody catalyzes the second step in the formation of the mannose 6-phosphate targeting signal on lysosomal enzyme oligosaccharides. It removes GlcNAc residues from GlcNAc-alpha-P-mannose moieties, which are generated in the first step. Additionally, it hydrolyzes UDP-GlcNAc, a sugar donor for Golgi N-acetylglucosaminyltransferases.
Gene References Into Functions
  1. 14 variations were discovered in GNPTAB, GNPTG and NAGPA genes. PMID: 29289611
  2. SNPs encompassing GNPTAB, GNPTG and NAGPA underwent genotyping, and association analysis was performed on all SNPs. Significant association of rs17031962 in GNPTAB and rs882294 in NAGPA with developmental dyslexia in a Chinese population was identified after false discovery rate correction for multiple comparisons. PMID: 25643770
  3. Mutational analysis of several residues within a highly conserved surface cavity of hUCE revealed their essentiality for function. PMID: 23572527
  4. To date, mutations in GNPTAB, GNPTG, and NAGPA have been linked to stuttering. These genes encode the lysosomal enzyme targeting pathway, which is defective in mucolipidosis. (Review) PMID: 22884963
  5. Analysis of mannose 6-phosphate uncovering enzyme mutations associated with persistent stuttering. PMID: 21956109
  6. Identified three mutations in the NAGPA gene associated with stuttering PMID: 20147709
  7. Synthesized as a proenzyme that is activated by furin [mannose 6-phosphate-uncovering enzyme] PMID: 12058031
  8. The mannose 6-phosphate uncovering enzyme participates in the uncovering of the mannose 6-phosphate recognition tag on lysosomal enzymes, a process that facilitates recognition of those enzymes by mannose 6-phosphate receptors for delivery to lysosomes. PMID: 15976452

Show More

Hide All

Database Links

HGNC: 17378

OMIM: 607985

KEGG: hsa:51172

STRING: 9606.ENSP00000310998

UniGene: Hs.21334

Involvement In Disease
Defects in NAGPA have been suggested to play a role in susceptibility to persistent stuttering. Stuttering is a common speech disorder characterized by repetitions, prolongations, and interruptions in the flow of speech.
Subcellular Location
Golgi apparatus, Golgi stack membrane; Single-pass type I membrane protein. Golgi apparatus, trans-Golgi network. Note=Cis/medial Golgi.
Tissue Specificity
Isoform 2 may be brain-specific.

Q&A

What is NAGPA and why is it important in research?

NAGPA (N-acetylglucosamine-1-phosphodiester alpha-N-acetylglucosaminidase) is a key enzyme that catalyzes the second step in forming the mannose 6-phosphate targeting signal on lysosomal enzyme oligosaccharides by removing GlcNAc residues from GlcNAc-alpha-P-mannose moieties . This process is critical for proper trafficking of lysosomal enzymes. Research on NAGPA contributes to understanding fundamental cellular sorting mechanisms, lysosomal storage disorders, and related pathologies. The enzyme's wide expression across multiple tissue types makes it relevant for various physiological studies beyond just lysosomal biology .

What are the common applications for anti-NAGPA antibodies?

Anti-NAGPA antibodies are primarily utilized in several immunodetection techniques. Immunohistochemistry represents the most common application, allowing researchers to visualize NAGPA distribution in tissue sections . Immunocytochemistry and immunofluorescence are also frequently employed to characterize subcellular localization, particularly in the Golgi apparatus where NAGPA is predominantly found . Less commonly, these antibodies may be used in Western blotting, ELISA, and co-immunoprecipitation studies to measure protein expression levels or investigate protein-protein interactions with NAGPA.

What molecular characteristics should researchers know about human NAGPA?

Human NAGPA has the following key molecular characteristics:

FeatureSpecification
Amino acid length515 residues (canonical form)
Molecular weight56.1 kDa
Subcellular localizationGolgi apparatus
Number of isoformsUp to 3 different reported isoforms
Expression patternWidely expressed across multiple tissue types
Known orthologsMouse, rat, bovine, frog, zebrafish, chimpanzee, chicken

These molecular characteristics are important considerations when designing experiments, as they inform antibody selection, expected band sizes in Western blots, and cross-reactivity potential across species .

How should NAGPA antibodies be stored and handled to maintain activity?

Most commercial anti-NAGPA antibodies should be stored at -20°C in their original buffered aqueous glycerol solution . For optimal antibody performance and longevity, avoid repeated freeze-thaw cycles by aliquoting the antibody upon receipt. When preparing working dilutions, use sterile, protein-free buffers and perform dilutions immediately before use. For short-term storage (1-2 weeks), diluted antibodies can be kept at 4°C with appropriate preservatives. Antibody degradation can be monitored through decreased signal intensity in control samples over time. Following these handling protocols is essential for experimental reproducibility and reliability of results.

What optimization strategies should be employed for NAGPA immunohistochemistry?

Optimizing NAGPA immunohistochemistry requires systematic adjustment of several parameters. Begin with antigen retrieval optimization, testing both heat-induced epitope retrieval methods (citrate buffer pH 6.0 and EDTA buffer pH 9.0) alongside enzymatic retrieval to determine which best exposes the NAGPA epitope . A dilution series (typically 1:50 to 1:1000, with commercial recommendations suggesting 1:200-1:500) should be performed to identify the optimal antibody concentration that maximizes specific signal while minimizing background .

Incorporate appropriate positive controls (tissues with known NAGPA expression) and negative controls (primary antibody omission and isotype controls) to validate specificity. When troubleshooting weak signals, consider extended incubation times (overnight at 4°C rather than 1 hour at room temperature) and signal amplification systems. For consistent results, standardize fixation protocols, as overfixation can mask epitopes while underfixation may compromise tissue morphology.

How can researchers distinguish between NAGPA isoforms in experimental settings?

Distinguishing between the three reported NAGPA isoforms requires specialized approaches:

  • Isoform-specific antibodies: Source or develop antibodies targeting unique epitopes in each isoform. This typically requires selecting regions with amino acid sequence variations between isoforms.

  • Western blot differentiation: Employ high-resolution SDS-PAGE (10-12%) with extended run times to separate closely related molecular weights of different isoforms. Thermal shift assays can also help identify differences in protein stability between isoforms.

  • Mass spectrometry: Use targeted proteomic approaches to identify unique peptide signatures for each isoform.

  • RT-PCR and qPCR: Design primers spanning unique exon junctions to quantify isoform-specific mRNA expression levels.

  • Recombinant expression studies: Express each isoform individually to serve as controls for antibody validation and binding specificity tests .

The resulting data should be analyzed through comparative approaches, with careful attention to pattern differences across tissue types, as expression ratios between isoforms may vary by cellular context.

What are the considerations for developing new anti-NAGPA antibodies for specific research needs?

Developing customized anti-NAGPA antibodies involves multiple strategic considerations. The immunogen selection is critical - researchers must choose between using the full protein, specific domains, or synthetic peptides based on the intended application . For peptide-based approaches, prioritize regions with high antigenicity, surface exposure, and low sequence conservation with related proteins to enhance specificity.

Computational approaches like JAM can accelerate antibody design by generating candidates with precise epitope targeting and strong developability profiles . If pursuing traditional methods, consider host species selection based on phylogenetic distance from the target species to enhance immunogenicity. For polyclonal development, implementation of affinity purification against the immunogen is essential to reduce cross-reactivity.

Post-development validation should include comprehensive cross-reactivity testing against related proteins, knockout/knockdown controls, and application-specific assessments (Western blot, IHC, IF) to confirm the antibody performs as intended across multiple experimental contexts .

How can researchers validate the specificity of anti-NAGPA antibodies?

Validating anti-NAGPA antibody specificity requires a multi-tiered approach:

  • Genetic controls: Test antibody against NAGPA knockout/knockdown systems; true anti-NAGPA antibodies should show diminished signal proportional to protein reduction.

  • Peptide competition assays: Pre-incubation of the antibody with excess immunizing peptide should abolish specific staining in subsequent experiments.

  • Orthogonal detection methods: Confirm antibody results with alternative detection methods such as in situ hybridization for mRNA or mass spectrometry for protein detection.

  • Cross-reactivity profiling: Test against recombinant NAGPA alongside structurally similar proteins, particularly other phosphodiesterases, to ensure signal specificity.

  • Multi-antibody confirmation: Validate findings using two or more antibodies against different NAGPA epitopes; overlapping staining patterns increase confidence in specificity .

Comprehensive validation should include both positive controls (tissues with known high NAGPA expression) and negative controls (tissues with minimal expression) to establish a dynamic range for detection.

What factors influence the selection of detection methods for NAGPA antibodies?

Selection of appropriate detection methods depends on research objectives, sample types, and required sensitivity. For chromogenic detection in IHC, consider enzyme systems (HRP vs. AP) based on endogenous enzyme activity in the tissue and potential for signal amplification requirements . Fluorescent detection offers multiplexing capabilities for co-localization studies but requires consideration of autofluorescence, photobleaching, and signal-to-noise optimization.

For quantitative applications, enzymatic immunoassays like ELISA provide better quantification than Western blots but typically require higher antibody amounts . When selecting secondary detection reagents, match them to the host species of your primary antibody, and evaluate options for direct conjugation versus avidin-biotin systems based on sensitivity needs.

The experimental design should also consider background mitigation strategies specific to each detection method, including appropriate blocking reagents (BSA, normal serum, commercial blockers) and washing optimization to enhance signal-to-noise ratios.

How do fixation and antigen retrieval methods affect NAGPA antibody performance?

Fixation and antigen retrieval significantly impact NAGPA antibody performance due to their effects on epitope accessibility. Formalin fixation creates protein cross-links that can mask NAGPA epitopes, particularly affecting the phosphodiester regions that are critical for enzyme function. Optimal fixation protocols typically involve 10% neutral buffered formalin for 24-48 hours for tissue sections, while cell preparations may require shorter fixation (10-15 minutes with 4% paraformaldehyde).

Heat-induced epitope retrieval (HIER) with citrate buffer (pH 6.0) frequently provides better results for NAGPA detection than alkaline buffers, though this should be empirically determined for each antibody . For challenging samples, a combination approach using both HIER and proteolytic digestion (proteinase K or trypsin) may enhance epitope exposure. The optimal retrieval method varies based on specific antibody characteristics, with some antibodies working best with higher pH retrieval buffers (Tris-EDTA, pH 9.0).

Systematic comparison of multiple retrieval methods is recommended when establishing protocols for new antibodies or tissues. Document changes in staining intensity, pattern specificity, and background levels under each condition to establish optimal parameters.

How should researchers quantify and interpret NAGPA immunostaining results?

Quantification of NAGPA immunostaining requires standardized approaches to ensure reproducibility and meaningful interpretation. For immunohistochemistry, implement a scoring system that accounts for both staining intensity (0-3+ scale) and percentage of positive cells to calculate H-scores or quick scores . Digital image analysis using software like ImageJ or QuPath provides more objective quantification through color deconvolution and automated intensity measurement.

For immunofluorescence, measure mean fluorescence intensity within regions of interest, normalized to appropriate controls. When analyzing subcellular localization, calculate colocalization coefficients (Pearson's or Mander's) with Golgi markers to confirm expected localization patterns .

Interpretation should consider normal NAGPA expression patterns and potential artifacts. Changes in staining pattern (e.g., from Golgi-specific to diffuse cytoplasmic) may indicate pathological alterations in protein trafficking rather than simple expression changes. Report results with appropriate statistical analyses and clear documentation of scoring methods to facilitate cross-study comparisons.

What are common pitfalls in NAGPA antibody experiments and how can they be avoided?

Several common pitfalls can compromise NAGPA antibody experiments:

  • Nonspecific binding: Manifests as diffuse background staining or unexpected subcellular localization patterns. Mitigate by optimizing blocking protocols with species-matched normal serum (5-10%) and including detergents (0.1-0.3% Triton X-100) to reduce hydrophobic interactions .

  • Edge artifacts: Particularly common in immunohistochemistry, appearing as intense staining at tissue edges. Minimize by ensuring adequate hydration of sections and implementing extended blocking steps.

  • Batch variation: Different lots of the same antibody may show performance variations. Maintain reference samples for inter-batch comparisons and document lot numbers in research records.

  • Cross-reactivity: Can produce misleading signals from related proteins. Address through comprehensive validation using multiple controls, including genetic knockdowns and competing antigens .

  • Hook effect: At very high antibody concentrations, paradoxical signal reduction can occur. Perform proper dilution series to identify optimal antibody concentrations.

Rigorous experimental design with appropriate positive and negative controls remains the best strategy for detecting and mitigating these potential problems.

How can conflicting results between different anti-NAGPA antibodies be reconciled?

Reconciling conflicting results between different anti-NAGPA antibodies requires systematic investigation of several factors:

  • Epitope mapping: Determine which specific regions of NAGPA each antibody targets. Antibodies recognizing different epitopes may produce divergent results, especially if certain epitopes are masked in specific contexts or isoforms .

  • Validation status assessment: Evaluate the extent of validation for each antibody. Prioritize results from antibodies with more comprehensive validation evidence, including knockout controls and peptide competition assays.

  • Application-specific optimization: An antibody performing well in Western blots may fail in IHC due to differences in protein conformation. Optimize each antibody for the specific application rather than assuming cross-application consistency.

  • Independent verification: Employ non-antibody-based methods (mRNA analysis, mass spectrometry) to resolve discrepancies in protein detection.

  • Methodology triangulation: When feasible, use three or more antibodies against different epitopes to establish consensus patterns. Consistent findings across multiple antibodies increase result confidence .

Document all experimental conditions thoroughly to facilitate troubleshooting and share detailed protocols in publications to enable better cross-laboratory comparisons.

How can NAGPA antibodies be employed in studies of lysosomal storage disorders?

NAGPA antibodies serve as powerful tools in investigating lysosomal storage disorders (LSDs) due to NAGPA's critical role in lysosomal enzyme trafficking. In research applications, these antibodies can be used to:

  • Evaluate enzyme trafficking defects: Immunofluorescence co-localization studies using NAGPA antibodies alongside markers for lysosomal, Golgi, and endosomal compartments can reveal trafficking abnormalities in LSD models.

  • Analyze phosphomannosyl signal formation: Since NAGPA catalyzes a critical step in mannose 6-phosphate signal generation, antibodies can help assess whether this pathway is compromised in specific LSDs .

  • Monitor therapeutic interventions: In experimental treatments aimed at correcting trafficking defects, NAGPA antibodies provide a readout of pathway restoration.

  • Detect compensatory mechanisms: Quantitative analyses using anti-NAGPA antibodies may reveal upregulation of the enzyme as a compensatory response in certain disorders.

Designing these studies requires careful consideration of sample preparation to preserve the native localization of NAGPA, typically requiring mild fixation protocols and careful membrane permeabilization to maintain Golgi structure integrity.

What are the considerations for using anti-NAGPA antibodies in multi-species comparative studies?

Using anti-NAGPA antibodies across species requires careful consideration of evolutionary conservation and epitope specificity. NAGPA has documented orthologs in mouse, rat, bovine, frog, zebrafish, chimpanzee, and chicken species, though sequence homology varies . When designing comparative studies:

  • Epitope conservation analysis: Perform sequence alignment of the antibody's target epitope across species of interest. Higher conservation predicts better cross-reactivity.

  • Validation gradient: Establish a validation hierarchy, with most rigorous validation in primary species, followed by decreasing stringency for more distant species.

  • Species-specific controls: Include positive controls from each species alongside experimental samples to confirm signal specificity.

  • Optimization by species: Modify protocols for each species, particularly antigen retrieval and antibody concentration, as fixation effects vary by tissue composition.

  • Cross-reactivity prediction: Use computational tools to predict cross-reactivity based on epitope conservation, then empirically confirm predictions before proceeding to full experimental designs.

This methodical approach minimizes false positives and negatives when extending NAGPA studies across evolutionary boundaries.

How can advanced computational approaches enhance NAGPA antibody development and application?

Computational approaches are revolutionizing antibody research, with specific applications for NAGPA studies:

  • Epitope prediction and optimization: Advanced algorithms can identify optimal epitopes within NAGPA for antibody development, prioritizing regions with high antigenicity, surface accessibility, and minimal cross-reactivity potential with related proteins .

  • De novo antibody design: Emerging systems like JAM enable computational design of antibodies with therapeutic-grade properties, including precise epitope targeting and strong developability profiles without experimental optimization . These approaches can generate NAGPA-specific antibodies with predefined characteristics.

  • Affinity and specificity prediction: Machine learning models can predict binding affinity and potential cross-reactivity issues before physical antibody production, streamlining development.

  • Structural analysis: Molecular dynamics simulations can model antibody-NAGPA interactions, informing optimization strategies for enhanced binding or specific recognition of NAGPA isoforms.

  • Immunogenicity assessment: Computational tools can evaluate potential immunogenicity of therapeutic anti-NAGPA antibodies, guiding humanization strategies when needed .

These computational approaches significantly reduce development timelines (to <6 weeks) and enable parallel pursuit of multiple design strategies, ultimately yielding higher-quality antibodies for NAGPA research .

What emerging delivery methods might improve anti-NAGPA antibody applications in neurological research?

Innovative delivery methods are expanding the potential applications of antibodies in neurological research, with implications for NAGPA studies:

  • Intranasal delivery: Recent studies demonstrate that intranasally applied therapeutic monoclonal antibodies can reach the central nervous system in detectable concentrations, with highest levels in olfactory bulbs . This non-invasive approach could facilitate studies of NAGPA in neurological disorders without requiring invasive procedures.

  • Enhanced BBB penetration: Engineering strategies that facilitate blood-brain barrier crossing, such as receptor-mediated transcytosis (using transferrin receptor or insulin receptor targeting), could improve NAGPA antibody delivery to brain tissues.

  • Nanoparticle encapsulation: Encapsulating anti-NAGPA antibodies in lipid nanoparticles or polymeric nanocarriers may enhance stability and tissue penetration while providing controlled release properties.

  • In situ expression: Viral vector-mediated expression of anti-NAGPA antibody fragments in specific brain regions offers highly localized, sustained antibody production for longitudinal studies.

These approaches demonstrate particular promise for investigating NAGPA's potential roles in neurodegenerative conditions where lysosomal dysfunction is implicated, such as Parkinson's and Alzheimer's diseases.

How might NAGPA antibodies contribute to understanding disease mechanisms beyond lysosomal disorders?

NAGPA antibodies have untapped potential for investigating diverse disease mechanisms:

  • Cancer biology: As protein trafficking pathways are frequently dysregulated in cancer, NAGPA antibodies can help assess alterations in lysosomal enzyme targeting that might contribute to tumor cell survival or metastatic potential.

  • Neurodegenerative diseases: Beyond classical lysosomal storage disorders, NAGPA dysfunction might contribute to protein aggregation in conditions like Alzheimer's and Parkinson's through impaired protein degradation pathways.

  • Immune system regulation: Given the importance of lysosomes in antigen processing and presentation, NAGPA antibodies could help investigate potential links between enzyme trafficking and immunological disorders.

  • Developmental biology: NAGPA's wide expression across tissues suggests developmental roles that could be explored using temporal and spatial expression mapping with specific antibodies.

  • Metabolic disorders: Disruptions in lysosomal enzyme trafficking could impact cellular metabolism through altered degradation and recycling pathways, potentially contributing to metabolic disease pathogenesis.

These applications require careful experimental design and integration of NAGPA studies with broader pathway analyses to establish mechanistic connections.

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.