NAAA Antibody

Shipped with Ice Packs
In Stock

Description

Functional Role of NAAA

NAAA regulates lipid signaling by degrading anti-inflammatory mediators like PEA. Its inhibition has been shown to:

  • Reduce tumor growth in colorectal cancer (CRC) xenografts and azoxymethane-induced models .

  • Attenuate chronic pain by modulating spinal cord lipid levels .

  • Influence tumor microenvironments by altering immune cell infiltration (e.g., macrophages, T cells) .

Key Pathways Affected by NAAA Inhibition

Pathway/ProcessImpact of NAAA Inhibition
PPAR-α/TRPV1 signalingReduces CRC cell proliferation
Toll-like receptor (TLR) pathwaysModulates innate immunity
Cyclin A2/CDK2 expressionInduces S-phase cell cycle arrest

Clinical Significance in Disease

NAAA dysregulation is linked to multiple pathologies:

  • Cancer:

    • Overexpression correlates with poor prognosis in breast cancer (BRCA), glioblastoma (LGG), and uveal melanoma (UVM) .

    • Downregulation in CRC tissues increases NAE levels, altering tumor secretome composition (e.g., reduced EGF family members) .

  • Autoimmunity:

    • NAAA inhibitors mimic IVIG effects, neutralizing pathogenic IgG autoantibodies in disorders like myasthenia gravis .

In Cancer Studies

  • CRC Models: NAAA inhibitor AM9053 reduced tumor growth by 40% in xenografts and suppressed EGF-related secretome factors .

  • Immune Modulation: High NAAA expression correlates with increased tumor-associated macrophages (TAMs), suggesting a pro-tumorigenic role .

In Neurological Disorders

  • Chronic Pain: NAAA inhibition during a critical post-injury window (PFD3–PFD4) blocked pain chronification in mice .

Future Directions

NAAA antibodies are emerging as biomarkers and therapeutic targets. Current challenges include optimizing inhibitor specificity and understanding isoform-specific roles in diseases like lupus and prostate cancer . Collaborative efforts between academic and pharmaceutical entities are critical for advancing clinical applications.

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
We typically dispatch products within 1-3 business days of receiving your order. Delivery time may vary depending on the purchase method and location. Please consult your local distributors for specific delivery timeframes.
Synonyms
Acid ceramidase like protein antibody; Acid ceramidase-like protein antibody; ASAH like protein antibody; ASAH-like protein antibody; N acylethanolamine acid amidase antibody; N acylethanolamine hydrolyzing acid amidase antibody; N acylsphingosine amidohydrolase (acid ceramidase) like antibody; N acylsphingosine amidohydrolase like antibody; N acylsphingosine amidohydrolase like protein antibody; N-acylethanolamine-hydrolyzing acid amidase subunit beta antibody; N-acylsphingosine amidohydrolase-like antibody; Naaa antibody; NAAA_HUMAN antibody; PLT antibody
Target Names
NAAA
Uniprot No.

Target Background

Function
NAAA is an enzyme that degrades bioactive fatty acid amides to their corresponding acids. It exhibits a preference for the following substrates: N-palmitoylethanolamine > N-myristoylethanolamine > N-lauroylethanolamine = N-stearoylethanolamine > N-arachidonoylethanolamine > N-oleoylethanolamine. Additionally, NAAA displays weak hydrolytic activity against the ceramides N-lauroylsphingosine and N-palmitoylsphingosine.
Gene References Into Functions
  1. Variants a1 and a2 encode the same full-length NAAA protein. PMID: 27693242
  2. Research suggests that N-acylethanolamine acid amidase and protein tyrosine kinase 7 may serve as potential tissue biomarkers to avoid overtreatment of non-aggressive prostate cancer. PMID: 24741114
  3. MALDI-TOF MS analysis of the human NAAA zymogen (47.7 kDa) treated with peptide-N-glycosidase F (PNGase F) identified 4 glycosylation sites. Acid cleavage of the zymogen into alpha- and beta-subunits (14.6 and 33.3 kDa) activated the enzyme. PMID: 22040171
  4. The level and activity of acid ceramidase in Alzheimer disease (AD) brain may play a role in controlling neuronal apoptosis and may mediate signaling pathways involved in the molecular mechanism of AD. PMID: 15610181
  5. Asn-37, Asn-107, Asn-309, and Asn-333 are actual N-glycosylation sites. Glycosylation appears to play a significant role in stabilizing the enzyme protein. PMID: 17980170
  6. This research describes the overexpression and processing of recombinant human acid ceramidase in insect cells, its purification, and characterization. PMID: 18020949
  7. These results demonstrate that both N-acylethanolamine-hydrolysing acid amidase and fatty acid amide hydrolase are functionally active in human prostate cancer cells. PMID: 18806270

Show More

Hide All

Database Links

HGNC: 736

OMIM: 607469

KEGG: hsa:27163

STRING: 9606.ENSP00000286733

UniGene: Hs.437365

Protein Families
Acid ceramidase family
Subcellular Location
Lysosome. Membrane; Peripheral membrane protein.
Tissue Specificity
Expressed in numerous tissues, with highest levels in liver and kidney, followed by pancreas.

Q&A

What is NAAA and why are antibodies against it important in research?

NAAA (N-acylethanolamine acid amidase) is a cysteine hydrolase that plays a key role in promoting inflammatory responses through the deactivation of palmitoylethanolamide (PEA), an endogenous bioactive lipid mediator. The protein is approximately 40.1 kilodaltons in mass and may also be known as PLT, ASAHL, N-acylethanolamine-hydrolyzing acid amidase, and ASAH-like protein . NAAA antibodies are crucial research tools for detecting and quantifying NAAA protein in various experimental systems, allowing investigators to study its role in inflammation, cancer, and other pathological conditions. These antibodies enable researchers to track both the intact and cleaved (activated) forms of NAAA through various detection methods including Western blotting, immunohistochemistry, and ELISA .

How do NAAA antibodies distinguish between intact and cleaved forms of the enzyme?

Anti-NAAA antibodies can detect both the intact (precursor) and cleaved (activated) forms of NAAA in experimental systems. When applied to blot membranes, these antibodies reveal both forms, typically showing a prevalence of the activated enzyme form . The ability to distinguish between these forms is critical for understanding NAAA activation dynamics in different physiological and pathological conditions. Researchers should select antibodies raised against epitopes that can detect both forms or specifically target regions unique to either the intact or cleaved enzyme, depending on experimental objectives. Western blot protocols optimized with appropriate molecular weight markers can help distinguish between the intact form (~40 kDa) and the cleaved active form.

What are the typical experimental applications for NAAA antibodies?

NAAA antibodies serve multiple experimental applications in research settings, with effectiveness varying by antibody type and experimental design:

ApplicationDescriptionTechnical Considerations
Western Blot (WB)Detection of NAAA protein levels in cell/tissue lysatesRequires optimization of protein extraction conditions and careful selection of reducing/non-reducing conditions
Immunohistochemistry (IHC)Visualization of NAAA distribution in tissue sectionsMay require antigen retrieval and careful validation of specificity
ELISAQuantitative measurement of NAAA levelsVarious formats available; sandwich ELISA often preferred for complex samples
ImmunoprecipitationIsolation of NAAA protein complexesSelection of antibodies with high affinity for native protein structure
ImmunofluorescenceSubcellular localization of NAAARequires validation with appropriate controls for specificity

Each application requires careful validation and optimization depending on the species and sample type being studied .

How should researchers validate NAAA antibody specificity?

Validation of NAAA antibody specificity is essential for generating reliable research data. A comprehensive validation approach should include:

  • Genetic controls: Testing antibody reactivity in NAAA knockout or knockdown models compared to wild-type samples. The absence of signal in knockout samples confirms specificity .

  • Peptide competition assays: Pre-incubation of the antibody with purified NAAA peptide should abolish specific signals in Western blot, IHC, or other detection methods.

  • Multiple antibody verification: Comparing results using antibodies from different sources or raised against different epitopes of NAAA.

  • Recombinant protein standards: Using purified NAAA protein as a positive control in Western blots or other applications.

  • Mass spectrometry verification: After immunoprecipitation with anti-NAAA antibody, mass spectrometry can confirm the identity of the precipitated protein.

  • Cross-reactivity testing: Evaluating potential cross-reactivity with related enzymes, particularly acid ceramidase (AC), which shares 33-34% identity and 70% similarity with NAAA .

Thorough validation ensures that experimental observations genuinely reflect NAAA biology rather than artifacts or off-target effects.

What are the optimal protocols for detecting NAAA using antibody-based techniques?

The optimal protocol for NAAA detection depends on the experimental context and objective. For Western blot analysis:

  • Sample preparation: Tissues or cells should be lysed in appropriate buffers containing protease inhibitors. For NAAA, which can exist in different activation states, care must be taken to preserve the protein's native state if studying the intact versus cleaved forms.

  • Electrophoresis conditions: Use reducing conditions (with β-mercaptoethanol or DTT) to fully denature the protein, allowing accurate molecular weight assessment.

  • Transfer parameters: For a 40.1 kDa protein like NAAA, semi-dry transfer at 15-20V for 30-45 minutes or wet transfer at 100V for 1 hour is typically effective.

  • Blocking: 5% non-fat dry milk or BSA in TBST for 1 hour at room temperature.

  • Primary antibody incubation: Anti-NAAA antibody diluted according to manufacturer's recommendations (typically 1:1000 to 1:5000) in blocking buffer, incubated overnight at 4°C.

  • Secondary antibody: HRP-conjugated or fluorescently labeled secondary antibody specific to the host species of the primary antibody, diluted 1:5000 to 1:10000, incubated for 1 hour at room temperature.

  • Detection: Enhanced chemiluminescence for HRP-conjugated antibodies or fluorescence imaging for fluorescently labeled antibodies.

For each new experimental system, these parameters should be optimized and validated.

How can researchers effectively use NAAA antibodies in studying inflammatory responses?

NAAA plays a significant role in inflammatory processes by deactivating PEA, which has anti-inflammatory properties. Researchers can effectively use NAAA antibodies to study inflammation through several approaches:

  • Correlation studies: Analyze NAAA expression levels in relation to inflammatory markers such as IL-6, IL-8, and NF-κB pathway activation. Studies have shown that inhibition of NAAA activity reduces the secretion of these inflammatory mediators in triple-negative breast cancer cells .

  • Cell-specific expression analysis: Use immunohistochemistry with anti-NAAA antibodies to identify cell types expressing NAAA in inflammatory lesions. This approach can be complemented with cell-specific genetic blockade of NAAA to understand its role in different cell populations .

  • Activation state monitoring: Track the ratio of intact to cleaved NAAA during inflammatory progression using Western blotting with anti-NAAA antibodies that recognize both forms .

  • Intervention studies: Combine anti-NAAA antibody detection with NAAA inhibitors (e.g., AM11095) to correlate changes in enzyme levels/activation with inflammation markers .

  • Co-localization with inflammatory mediators: Use dual immunofluorescence with anti-NAAA antibodies and antibodies against inflammatory mediators to study their spatial relationship in tissues.

These approaches can provide valuable insights into the role of NAAA in various inflammatory conditions and potential therapeutic strategies.

How can activity-based protein profiling be combined with NAAA antibodies to enhance functional studies?

Combining activity-based protein profiling (ABPP) with NAAA antibodies creates a powerful approach for studying NAAA function. A strategic protocol involves:

  • ABPP probe application: Use β-lactam ABPP probes like Compound 1 (from search result 2) with a terminal alkyne functionality to selectively label catalytically active NAAA . These probes covalently bind to the active site cysteine of NAAA.

  • Click chemistry reaction: Perform 1,3-dipolar Huisgen cycloaddition to attach a reporter tag (biotin or fluorophore) to the probe-labeled NAAA.

  • Enrichment or detection: For biotin tags, use streptavidin pull-down to enrich for active NAAA; for fluorophores, direct visualization can be performed.

  • Antibody-based verification: Use anti-NAAA antibodies in Western blot to confirm the identity of the captured proteins.

  • Competitive ABPP: Pre-treatment with NAAA inhibitors (e.g., ARN726 or ARN077) blocks probe binding, allowing quantification of inhibitor effectiveness .

This combined approach provides information not only about NAAA expression levels but also about its enzymatic activity, which is particularly valuable when studying disease mechanisms or evaluating potential therapeutics. The method has successfully been used to detect catalytically active NAAA in both HEK293 cells overexpressing human NAAA and in rat lung tissue .

How do NAAA expression patterns differ across tissue types and disease states?

NAAA expression exhibits significant variability across tissues and disease states, which can be systematically analyzed using antibody-based detection methods:

Tissue/ConditionNAAA Expression PatternMethodologySignificance
Healthy lungModerate expression in macrophages and epithelial cellsIHC, Western blotBaseline for comparison with inflammatory conditions
Inflamed lungUpregulated, particularly in infiltrating immune cellsIHC, Western blotIndicates role in inflammatory response regulation
Triple-negative breast cancerElevated levels of full-length and splice mRNA variantsRT-PCR, Western blotPotential therapeutic target for cancer management
Normal breast tissueLow expressionIHC, Western blotBaseline for comparison with cancer
Immune cellsCell-type specific expressionFlow cytometry with anti-NAAA antibodiesImportant for understanding role in immune regulation

Researchers investigating NAAA expression should employ multiple detection methods, including quantitative Western blotting and immunohistochemistry with validated anti-NAAA antibodies, to generate comprehensive expression profiles. This information is crucial for identifying potential therapeutic opportunities, as exemplified by the finding that NAAA inhibition may constitute a potential therapeutic approach in managing triple-negative breast cancer-associated inflammation and tumor growth .

What are the challenges in designing highly specific antibodies against NAAA?

Developing highly specific antibodies against NAAA presents several technical challenges:

  • Structural homology with related enzymes: NAAA shares 33-34% identity and 70% similarity with acid ceramidase (AC) , which can lead to cross-reactivity. Careful epitope selection is required to avoid regions of high homology.

  • Multiple isoforms: NAAA exists in both intact and cleaved forms, and potential splice variants have been identified in triple-negative breast cancer cells . Antibodies must be designed to either distinguish between these forms or recognize common epitopes, depending on the research question.

  • Post-translational modifications: These can alter epitope accessibility or antibody binding. Understanding the native structure and modifications of NAAA is essential for effective antibody design.

  • Species cross-reactivity: Creating antibodies that recognize NAAA across species (human, mouse, rat) requires targeting conserved epitopes, which may overlap with conserved regions in related enzymes.

  • Confirmation of specificity: Innovative approaches are needed for specificity validation, including the biophysics-informed modeling described in search result , which can help design antibodies with custom specificity profiles.

Addressing these challenges requires a combination of careful epitope selection, extensive validation using knockout controls, and possibly the application of newer technologies like biophysics-informed modeling that can associate distinct binding modes with potential ligands .

How should researchers address discrepancies between NAAA protein levels detected by antibodies and mRNA expression?

Discrepancies between NAAA protein and mRNA levels are common and should be systematically addressed:

  • Validate both detection methods: Ensure NAAA antibodies are specific using appropriate controls, and confirm mRNA primers are correctly designed and specific (e.g., using the primers described in search result : 5'-TGCCCTTACAATCCTCCTGC-3' (forward), 5'-TGCCATCTAGATCCCTGACA-3' (reverse)) .

  • Consider post-transcriptional regulation: NAAA may be subject to microRNA regulation, RNA stability differences, or translational control that affects the relationship between mRNA and protein levels.

  • Evaluate protein stability and turnover: Differences in protein half-life can lead to accumulation of protein despite lower mRNA levels or vice versa. Pulse-chase experiments with protein synthesis inhibitors can help understand NAAA turnover rates.

  • Analyze activation state: Since NAAA exists in both inactive precursor and cleaved active forms, antibodies detecting only one form might not correlate with total mRNA levels. Use antibodies capable of detecting both forms for comprehensive analysis .

  • Examine spatial and temporal dynamics: Sample collection timing and specific cell populations analyzed can influence the correlation between mRNA and protein levels. Single-cell analysis techniques may provide more detailed insights.

  • Consider technical limitations: Different detection methods have varying sensitivities and dynamic ranges. Western blotting may be less quantitative than ELISA or mass spectrometry for protein quantification.

When discrepancies persist, they may reflect genuine biological phenomena rather than technical issues, potentially revealing important regulatory mechanisms controlling NAAA expression.

What controls should be included when using NAAA antibodies in various experimental platforms?

Rigorous experimental design for NAAA antibody applications should include comprehensive controls:

Control TypeDescriptionRationaleApplication
Positive ControlRecombinant NAAA protein or lysate from cells known to express high NAAA levelsConfirms antibody functionalityAll platforms
Negative ControlSamples from NAAA knockout/knockdown models or tissues known not to express NAAAVerifies specificityAll platforms
Isotype ControlAntibody of same isotype but irrelevant specificityDetects non-specific binding due to Fc receptor interactionsIHC, flow cytometry
Secondary Antibody ControlSample incubated with secondary antibody only (no primary)Identifies background from secondary antibodyIHC, WB, IF
Peptide CompetitionPre-incubation of antibody with immunizing peptideConfirms epitope-specific bindingAll platforms
Multiple Antibody ValidationTesting with antibodies targeting different NAAA epitopesVerifies target identityWB, IHC
Dilution SeriesTesting antibody at multiple concentrationsOptimizes signal-to-noise ratioAll platforms
Cross-reactivity AssessmentTesting against acid ceramidase or other related proteinsConfirms NAAA-specific detectionWB, IHC

Implementing these controls systematically ensures that experimental observations accurately reflect NAAA biology rather than technical artifacts. This is particularly important given the potential methodological variability in antibody-based testing, similar to the issues observed with antinuclear antibody testing described in search result .

How can researchers distinguish between active and inactive forms of NAAA in experimental systems?

Distinguishing between active and inactive NAAA forms requires specialized techniques:

  • Activity-based protein profiling: β-lactam ABPP probes like Compound 1 selectively bind to catalytically active NAAA . This approach provides direct evidence of enzymatic activity rather than just protein presence.

  • Western blotting with form-specific antibodies: Use antibodies that can distinguish between the intact precursor (~40.1 kDa) and the cleaved active form. The cleaved form is typically more prevalent when NAAA is activated .

  • Enzymatic activity assays: Measure NAAA activity directly by quantifying the hydrolysis of substrate molecules like PEA. Correlation between detected protein levels and enzymatic activity provides insight into the active fraction.

  • Subcellular fractionation: Active NAAA is typically found in specific cellular compartments. Fractionation followed by Western blotting can help determine the activation state based on localization.

  • Conformation-specific antibodies: Though challenging to develop, antibodies that specifically recognize the active conformation of NAAA would provide direct evidence of activation state.

  • Correlation with regulatory factors: Monitor known activators or inhibitors of NAAA alongside protein levels to infer activation state.

  • Mass spectrometry: Peptide mapping can identify post-translational modifications associated with activation or inactivation of NAAA.

These approaches, particularly when used in combination, allow researchers to develop a comprehensive understanding of NAAA activation dynamics in different experimental contexts.

How can NAAA antibodies contribute to understanding the role of NAAA in cancer progression?

NAAA antibodies are instrumental in elucidating NAAA's role in cancer progression through several methodological approaches:

  • Expression profiling across cancer types: Systematic immunohistochemical analysis using validated NAAA antibodies can reveal differential expression patterns between normal tissues and various cancer types, stages, and grades. Recent findings indicate elevated NAAA expression in triple-negative breast cancer cells, including both full-length and splice variants .

  • Correlation with prognostic markers: Combined immunohistochemistry for NAAA and established cancer progression markers can identify associations between NAAA expression and tumor aggressiveness. Studies have suggested that NAAA may be involved in tumor aggressiveness .

  • Functional inhibition studies: Using NAAA antibodies alongside NAAA inhibitors like AM11095 (IC₅₀ = 20 nM) to correlate changes in enzyme levels/activation with cancer phenotypes. Research has demonstrated that AM11095 inhibits the secretion of IL-6 and IL-8, reduces activation of the NF-κB pathway, decreases expression of VEGF and Placental growth factor (PLGF) in triple-negative breast cancer cells, and inhibits tumor cell migration in vitro .

  • In vivo tumor growth monitoring: Cellular magnetic resonance imaging of mice administered with human cancer cells treated with NAAA inhibitors has shown significant decreases in tumor numbers and volumes, with increased survival rates .

  • Mechanistic pathway analysis: Combining NAAA antibody detection with assessment of inflammatory mediators, angiogenic factors, and cell migration markers to elucidate the mechanisms through which NAAA influences cancer progression.

These approaches collectively demonstrate that NAAA inhibition may constitute a potential therapeutic approach in managing cancer-associated inflammation and tumor growth, particularly in aggressive cancers like triple-negative breast cancer .

What are the methodological considerations for studying NAAA in different cellular compartments?

Studying NAAA across cellular compartments requires specialized methodological approaches:

  • Subcellular fractionation optimization: Standard protocols should be modified to preserve NAAA activity during isolation of lysosomes, endosomes, and other relevant compartments. Use of protease inhibitors and gentle lysis conditions is crucial.

  • Immunofluorescence confocal microscopy: Co-staining with anti-NAAA antibodies and organelle markers (e.g., LAMP1 for lysosomes, EEA1 for early endosomes) can reveal the spatial distribution of NAAA. Z-stack acquisition and deconvolution improve resolution for accurate localization.

  • Live-cell imaging: Where possible, use of fluorescently tagged NAAA constructs validated against antibody staining can reveal dynamic trafficking between compartments. This approach requires careful validation to ensure the tag doesn't interfere with localization or function.

  • Proximity ligation assays: These can identify interactions between NAAA and compartment-specific proteins with greater sensitivity than conventional co-localization studies.

  • Super-resolution microscopy: Techniques such as STORM or STED, using high-quality anti-NAAA antibodies, can provide nanoscale resolution of NAAA localization beyond the diffraction limit of conventional microscopy.

  • Electron microscopy with immunogold labeling: Using anti-NAAA antibodies conjugated to gold particles allows ultrastructural localization of NAAA with the highest possible spatial resolution.

  • Compartment-specific activity assays: Combining subcellular fractionation with activity-based protein profiling using probes like Compound 1 can reveal the distribution of enzymatically active NAAA across cellular compartments.

These complementary approaches can provide a comprehensive understanding of NAAA's subcellular distribution and how this relates to its function in different physiological and pathological contexts.

How can antibody-based approaches help elucidate the interaction between NAAA and its endogenous inhibitors?

Antibody-based methodologies offer unique insights into NAAA interaction with endogenous inhibitors:

  • Co-immunoprecipitation (Co-IP): Using anti-NAAA antibodies to pull down NAAA complexes, followed by proteomic analysis to identify interacting partners, including potential endogenous inhibitors. This approach can be enhanced with crosslinking to capture transient interactions.

  • Biolayer interferometry or surface plasmon resonance: Immobilizing purified NAAA using capture antibodies, then measuring binding kinetics of potential endogenous inhibitors in real-time.

  • Proximity-based labeling: Techniques like BioID or APEX2, where NAAA is fused to a promiscuous biotin ligase, can identify proteins in proximity to NAAA under various conditions. Comparing results in the presence or absence of known endogenous inhibitors can reveal regulatory complexes.

  • Fluorescence resonance energy transfer (FRET): Using fluorescently labeled anti-NAAA antibodies and antibodies against potential endogenous inhibitors to detect molecular proximity in intact cells.

  • Competitive binding assays: Developing assays where labeled synthetic inhibitors like AM11095 compete with endogenous inhibitors for NAAA binding, with anti-NAAA antibodies used for detection or capture.

  • Conformational antibodies: Developing antibodies that specifically recognize NAAA conformations induced by inhibitor binding can provide insights into structural changes associated with inhibition.

  • In situ proximity ligation assay: This technique can visualize and quantify interactions between NAAA and its endogenous inhibitors in fixed cells or tissues with high sensitivity.

These approaches can reveal not only the identity of endogenous NAAA inhibitors but also the mechanisms and contexts of their regulation, potentially informing the development of therapeutic strategies targeting NAAA activity.

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.