nos Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to NOS Antibodies

NOS antibodies are specialized research tools designed to detect and study nitric oxide synthases (NOS), enzymes catalyzing the production of nitric oxide (NO) from L-arginine. NO plays critical roles in signaling, immune responses, and vascular regulation. Three isoforms of NOS exist: neuronal NOS (nNOS/NOS1), endothelial NOS (eNOS/NOS3), and inducible NOS (iNOS/NOS2). These isoforms differ in expression patterns, regulation, and cellular localization, necessitating isoform-specific antibodies for precise research applications .

nNOS Antibodies

  • Target: Neuronal NOS (NOS1), expressed in neurons, skeletal muscle, and certain epithelial cells.

  • Applications:

    • WB: Detects bands at 155–165 kDa in brain lysates .

    • IHC: Stains neurons in brain sections and smooth muscle in vessel walls .

  • Challenges: Cross-reactivity with non-neuronal tissues (e.g., smooth muscle) in some polyclonal antibodies .

eNOS Antibodies

  • Target: Endothelial NOS (NOS3), critical for vascular NO production.

  • Applications:

    • WB: Detects eNOS at 130–140 kDa in endothelial cell lysates .

    • IHC: Stains endothelial cells in blood vessels but may show nonspecific glial cell reactivity in brain sections .

iNOS Antibodies

  • Target: Inducible NOS (NOS2), upregulated in inflammation and cancer.

  • Applications:

    • WB: Detects iNOS at ~160 kDa in macrophage lysates .

    • IHC: Identifies iNOS in tumor-associated macrophages or epithelial cells .

Pan-NOS Antibodies

  • Target: Detects eNOS, nNOS, and iNOS.

  • Applications:

    • WB: Broad utility for total NOS quantification .

    • Limitations: Poor isoform discrimination, unsuitable for studies requiring specificity .

Challenges in Antibody Specificity

Commercial NOS antibodies often exhibit cross-reactivity, particularly in non-human models:

Antibody TypeReported Cross-ReactivityImpact on ResearchSources
PolyclonalSmooth muscle, glial cellsFalse-positive staining in IHC
MonoclonalLimited to species-specific epitopesRequires validation in target tissues
Pan-NOSOverlapping bands in WBComplicates isoform-specific analysis

Case Study:

  • A rabbit anti-human nNOS antibody (Santa Cruz) showed strong reactivity with smooth muscle cells in rodent tissues, masking neuronal signals .

  • Solution: Validate antibodies in positive/negative controls and use species-matched secondary antibodies .

Research Applications and Findings

NOS antibodies have enabled pivotal discoveries in cancer, neuroscience, and vascular biology:

Study FocusFindingsImplicationsSources
Metaplastic Breast CancerNOS inhibition + PI3K blockade reduced chemoresistance and EMT markersNovel therapeutic strategy for MpBC
Radiation TherapyNOS inhibition enhanced tumor radiosensitivity by reducing blood flowSynergistic treatment for solid tumors
NeuroinflammationnNOS antibodies localized neuronal NO production in neurodegenerative modelsAdvances in Parkinson’s/Alzheimer’s research

Key Insight:

  • NOS2 (iNOS) upregulation in tumors correlates with poor prognosis. Antibodies targeting iNOS enable tracking of its role in cancer progression .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Description

This nos polyclonal antibody is generated using a recombinant Drosophila melanogaster (Fruit fly) nos protein (amino acids 211-401) as the immunogen. The antibody is elicited in a rabbit and subsequently collected from its serum. The antibody undergoes rigorous affinity chromatography purification for enhanced specificity. Its functionality is validated through ELISA and WB assays, ensuring its ability to reliably detect the Drosophila melanogaster (Fruit fly) nos protein in various experimental settings.

The Nanos (nos) protein plays a crucial role in the developmental processes of Drosophila melanogaster. Classified as a maternal-effect gene, it contributes significantly to the specification of cell fates along the anterior-posterior axis of the embryo, with a particular focus on the posterior region. Furthermore, it participates in the formation and specification of primordial germ cells, which are essential for the development of reproductive cells in the adult organism. Its influence extends to posterior morphogenesis, where it actively defines the body plan and contributes to the proper formation of the embryo's posterior pole.

Form
Liquid
Lead Time
Typically, we can dispatch your orders within 1-3 working days of receipt. Delivery times may vary depending on the chosen purchasing method and location. For precise delivery timelines, kindly consult your local distributors.
Synonyms
Protein nanos nos CG5637
Target Names
nos
Uniprot No.

Target Background

Function
Nanos protein acts as a maternal RNA-binding protein, essential for germ cell proliferation and self-renewal. It functions by forming a complex with pum and brat proteins, which regulate translation and mRNA stability. This complex specifically binds to the Nanos Response Element (NRE), a 16 bp sequence located within the hb mRNA 3'-UTR, preventing its translation. Notably, Nanos protein plays a critical role in posterior development, serving as a rescuing factor for the abdominal defects observed in posterior group mutants. It is important to emphasize that other posterior group genes are not essential for Nanos function but rather contribute to its localization and distribution within the organism.
Gene References Into Functions
  1. Nos protein exhibits dual functions in translational repression and stabilization of specific RNAs, crucial for proper germline development. PMID: 29278271
  2. The germ stem cell-rescuing activity of Wolbachia TomO protein demonstrates a strong connection to its ability to enhance Nos protein expression. PMID: 29701280
  3. TomO facilitates the maintenance of germ stem cells (GSCs) by increasing Nanos (Nos) expression through its interaction with nos mRNA. This ultimately leads to the restoration of germ cell production in Sxl mutant females, which otherwise lack GSCs. [ PMID: 27498563
  4. Translational repression of the Drosophila nanos mRNA involves the RNA helicase Belle and RNA coating by Me31B and Trailer hitch. Bel is crucial for nos mRNA translational repression. PMID: 28701521
  5. Research has indicated that nos mRNA, identified as one of the target mRNAs of Hrp38 in this study, is regulated by Hrp38 for translational control during ovary development. PMID: 27402862
  6. Experimental findings demonstrate that dynein motor machinery components mediate the transport of nos mRNA in proximal dendrites. PMID: 24027279
  7. Mei-P26 interacts with Bam, Bgcn and Sxl, as well as nanos mRNA, during early germline stem cell development, suggesting its role in repressing the translation of nanos mRNA. PMID: 23526974
  8. nanos stands out as a critical regulatory target of polar granule components in primordial germ cells. PMID: 23173091
  9. Research findings suggest that Sxl enables the transition from germline stem cell to committed daughter cell by posttranscriptional down-regulation of nanos expression. PMID: 22645327
  10. Evidence indicates that during late oogenesis, nos translation is also repressed post-initiation, with Glo playing a key role in this event. PMID: 21460235
  11. The minimal promoter (from -108 to +20) of Nanos proves sufficient in germline stem cells for the green fluorescent protein expression in transgenic Drosophila. PMID: 20026207
  12. Ectopic expression of the Translational Control Element of nos suggests that a common translational control mechanism is at work in axial patterning and neuroendocrine signaling in Drosophila. PMID: 12091303
  13. Removal of Nanos from either germline stem cells (GSCs) or their precursors, PGCs, causes both cell types to differentiate into germline cysts. Nanos is crucial for both establishing and maintaining GSCs by preventing their premature entry into oogenesis. PMID: 14976263
  14. Nos plays a critical role in establishing the germ-line/soma dichotomy and is also essential for maintaining germ-line fate. PMID: 15240884
  15. In-depth analysis of a translational control element in the nanos mRNA provides valuable insights. PMID: 15525666
  16. Nanos downregulates transcription and modulates CTD phosphorylation during Drosophila development. PMID: 15817222
  17. The nanos translational control element represses translation in somatic cells through a Bearded box-like motif. PMID: 15936341
  18. Nanos is essential for regulating the formation of the germ cell-specific spectrosome, further supporting its role in promoting germ cell identity. PMID: 16028275
  19. Smaug response elements found in the nanos 3'-untranslated region are necessary and sufficient to induce deadenylation. PMID: 16793774
  20. Rumpelstiltskin regulates anterior-posterior axis patterning by directly acting as a nanos mRNA localization factor. PMID: 18234721
  21. Localization and translational control of nos mRNA are crucial for dendritic arborization neuron morphogenesis. PMID: 18472422
  22. These observations suggest that Bam-Bgcn collaborate to antagonize Nos expression, thereby derepressing cystoblast-promoting factors. These findings highlight the importance of translational repression in balancing stem cell self-renewal and differentiation. PMID: 19470484

Show More

Hide All

Database Links

KEGG: dme:Dmel_CG5637

STRING: 7227.FBpp0083146

UniGene: Dm.1485

Protein Families
Nanos family
Subcellular Location
Cytoplasm, Cytoplasmic ribonucleoprotein granule.
Tissue Specificity
Posterior part of the embryo. While the transcript is present throughout the embryo, nos translation is controlled by smg, and the protein is found in pole plasm and pole cells.

Q&A

What are the different NOS isoforms and how do I select the appropriate antibody?

Nitric Oxide Synthase exists in three major isoforms: neuronal (nNOS), endothelial (eNOS), and inducible (iNOS). When selecting an appropriate antibody, researchers should consider both the target epitope and species reactivity. Commercially available antibodies target different regions of these isoforms, including N-terminus, mid-region, and C-terminus epitopes .

The choice depends on your experimental needs:

  • For cross-species applications, select antibodies targeting conserved epitopes

  • For isoform specificity, choose antibodies targeting unique regions

  • For specific applications (like Western Blotting vs. IHC), select antibodies validated for that technique

NOS isoformSpeciesAntigenAmino acidCommon Applications
nNOSRatSynthetic peptide; mid-region724-739WB, IHC, ICC
nNOSHumanSynthetic peptide; N-terminus37-56WB, IHC, ICC
eNOSBovineSynthetic peptide; mid-region599-613WB, IHC, IF
iNOSMouseSynthetic peptide; C-terminus1131-1144WB, ELISA, FCM
iNOSHumanSynthetic peptide; C-terminus1135-1153WB, IHC, IF

How is the specificity of NOS antibodies determined experimentally?

Determining NOS antibody specificity requires a multi-faceted approach. Researchers typically employ:

  • Cross-reactivity testing across species: Testing antibodies on tissues from different species (human, guinea pig, rat, mouse) to determine conservation of epitope recognition

  • Isoform testing: Validation across all three major NOS isoforms (nNOS, eNOS, iNOS) to confirm isoform-specific binding

  • Complementary techniques: Using both immunohistological and immunoblotting techniques in parallel to confirm specificity

  • Control experiments: Including pre-absorption with immunizing peptides, testing on knockout tissues, and comparing staining patterns with established antibodies

For definitive specificity analysis, researchers employ double-staining techniques with markers like CD68 (for macrophages) when examining NOS reactivity in human lung and brain tissues .

What are the common pitfalls in NOS antibody-based experimental design?

When designing experiments using NOS antibodies, researchers commonly encounter several challenges:

  • Non-specific binding: Particularly problematic in highly vascularized tissues where endogenous peroxidase activity can create false positives

  • Species cross-reactivity issues: An antibody's reactivity profile can vary significantly across species, requiring careful validation for each target species

  • Fixation artifacts: Over-fixation can mask epitopes, while under-fixation can compromise tissue morphology

  • Isoform cross-reactivity: Some antibodies may react with multiple NOS isoforms despite manufacturer claims of specificity

Best methodological practices include:

  • Incorporating proper negative controls (omitting primary antibody)

  • Using tissue from knockout animals when available

  • Pre-absorbing antibodies with immunizing peptides

  • Including 1% normal serum from the tissue species to minimize non-specific binding

How should I optimize immunohistochemical protocols for NOS antibody detection?

Optimizing immunohistochemical protocols for NOS detection requires attention to several methodological details:

  • Tissue fixation: For most NOS isoforms, 4% paraformaldehyde for 24 hours provides optimal epitope preservation while maintaining tissue architecture.

  • Antigen retrieval: Heat-induced epitope retrieval using citrate buffer (pH 6.0) often improves NOS antibody binding, particularly for formalin-fixed tissues.

  • Blocking procedures: Optimal blocking requires 1% (v/v) normal serum from the species from which the tissue was tested, which minimizes non-specific binding .

  • Detection systems: For NOS antibodies:

    • Use peroxidase-conjugated secondary antibodies for single staining

    • For double staining protocols, combine peroxidase and alkaline phosphatase conjugated secondaries

    • Develop peroxidase reactivity using 3-amino-9-ethylcarbazole and H₂O₂

  • Counterstaining: Light hematoxylin counterstaining provides optimal nuclear detail without obscuring NOS-positive signals.

What approaches ensure reliable quantification of NOS expression using antibodies?

Reliable quantification of NOS expression requires methodological rigor:

  • Standard curve calibration: Use recombinant NOS proteins at known concentrations to establish standard curves for quantitative analyses.

  • Image analysis parameters:

    • Set consistent threshold values for positive staining

    • Use automated particle counting with size exclusion parameters

    • Normalize to cell count or tissue area

  • Technical considerations:

    • Maintain identical acquisition parameters across samples

    • Process experimental and control samples simultaneously

    • Include internal reference standards in each experimental batch

  • Statistical validation:

    • Perform replicate analysis (minimum n=3)

    • Validate with alternative techniques (e.g., qPCR for mRNA expression)

    • Apply appropriate statistical tests for the specific experimental design

How do I conduct multi-species validation of NOS antibodies?

Comprehensive validation across species requires methodical testing protocols:

  • Sequential validation approach:

    • Begin with Western blotting to confirm molecular weight

    • Proceed to immunohistochemistry on known positive control tissues

    • Compare staining patterns across species for known NOS-rich regions

  • Species panel testing: Test antibodies systematically across human, guinea pig, rat and mouse tissues, focusing on cerebrum, cerebellum, lung, spleen, liver, and kidney samples .

  • Cross-reactivity documentation: Document all observed cross-reactivity patterns, including:

    • Different staining intensities

    • Non-specific binding patterns

    • Species-specific background issues

  • Epitope conservation analysis: Compare the amino acid sequence of the immunizing peptide across species to predict potential cross-reactivity .

What computational approaches can improve NOS antibody specificity prediction?

Advanced computational modeling has revolutionized antibody specificity prediction:

  • Biophysics-informed modeling: Combines physicochemical principles with experimental data to predict antibody binding profiles across targets .

  • Selection experiment integration: Phage display selection data can be incorporated into computational models to improve prediction accuracy for NOS antibody binding .

  • Energy function optimization: Mathematical models using energy functions can be developed to predict and optimize antibody sequences with desired binding profiles:

    • For cross-specific sequences, jointly minimize the energy functions associated with desired ligands

    • For specific sequences, minimize energy functions for desired ligands while maximizing those for undesired ligands

These computational methods allow researchers to design custom antibodies with predetermined specificity profiles, either targeting specific NOS isoforms or creating cross-reactive antibodies as needed for particular research applications .

How can I design experiments to generate NOS antibodies with custom specificity profiles?

Creating NOS antibodies with tailored specificity requires sophisticated experimental design:

  • Phage display selection strategy:

    • Design selection experiments against multiple ligands or combination of ligands

    • Perform sequential selections with appropriate amplification steps

    • Include pre-selection steps to deplete non-specific binders

  • Cross-specific vs. specific binding design:

    • For cross-specific antibodies: select against mixtures of similar antigens

    • For highly specific antibodies: implement negative selection strategies

    • Monitor library composition at each selection step

  • Validation requirements:

    • Test predicted sequences not present in training sets

    • Evaluate binding using multiple orthogonal techniques

    • Confirm specificity across physiologically relevant conditions

  • Computational integration:

    • Use experimental data to train computational models

    • Apply models to design novel sequences with desired properties

    • Iterate between computational prediction and experimental validation

How does post-translational modification affect NOS antibody binding and experimental design?

Post-translational modifications (PTMs) significantly impact NOS antibody recognition:

  • Common NOS modifications affecting antibody binding:

    • Phosphorylation: Particularly at serine, threonine, and tyrosine residues

    • S-nitrosylation: Can alter epitope accessibility

    • Proteolytic processing: May eliminate epitopes or create new ones

  • Experimental approaches for PTM-aware research:

    • Use phospho-specific antibodies when studying NOS regulation

    • Perform parallel analyses with antibodies targeting different epitopes

    • Treat samples with phosphatases to determine phosphorylation effects

  • PTM-sensitive application considerations:

    • Western blotting: PTMs may alter migration patterns

    • Immunoprecipitation: PTMs can affect antibody-antigen complexation

    • Immunohistochemistry: Fixation methods may preserve or destroy PTMs

  • Controls for PTM-dependent binding:

    • Include positive controls with known modification states

    • Compare native and denaturing conditions to assess conformational dependencies

    • Validate with mass spectrometry to confirm modification states

How can NOS antibodies be utilized in the context of neurodegenerative disease research?

NOS antibodies have become valuable tools in neurodegenerative research:

  • nNOS-specific applications:

    • Tracking neuronal stress responses in Alzheimer's and Parkinson's disease

    • Monitoring excitotoxicity-related nNOS upregulation

    • Evaluating nNOS subcellular redistribution during disease progression

  • iNOS applications in neuroinflammation:

    • Quantifying microglial activation states

    • Correlating inflammatory responses with disease severity

    • Evaluating therapeutic efficacy of anti-inflammatory interventions

  • Methodological considerations:

    • Use double-labeling with neuronal and glial markers

    • Implement cell type-specific analysis of NOS expression

    • Compare expression patterns between affected and unaffected brain regions

  • Technical advances:

    • Super-resolution microscopy for subcellular NOS localization

    • Multiplexed imaging to correlate NOS expression with other disease markers

    • Live-cell imaging using tagged NOS antibodies for dynamic studies

What role do NOS antibodies play in understanding viral infection mechanisms?

NOS antibodies provide insights into host-pathogen interactions:

  • iNOS antibody applications in viral research:

    • Tracking macrophage activation during infection

    • Correlating NO production with viral clearance

    • Monitoring tissue-specific responses to viral challenge

  • Relevance to broadly neutralizing antibody development:

    • NOS expression correlates with neutralizing antibody development in some viral infections

    • Broadly neutralizing antibody responses typically develop 2-4 years after HIV infection, in 10-20% of individuals

    • High viral load and low CD4+ T cell counts are associated with neutralizing antibody breadth

  • Methodological integration:

    • Combining NOS expression analysis with viral load quantification

    • Correlating immune cell activation with antibody development kinetics

    • Tracking temporal changes in NOS expression during infection progression

  • Technical considerations:

    • Use tightly controlled time points for accurate kinetic analysis

    • Implement tissue-specific extraction protocols to preserve NOS activity

    • Combine protein and mRNA analysis for comprehensive expression profiling

How should I interpret contradictory results when using different NOS antibodies?

Resolving contradictory results requires systematic investigation:

  • Epitope mapping comparison:

    • Compare the exact epitopes recognized by different antibodies

    • Determine if epitopes are in regions susceptible to conformational changes

    • Check for potential post-translational modification sites within epitopes

  • Methodological reconciliation approach:

    • Standardize fixation and permeabilization protocols

    • Implement identical blocking and washing procedures

    • Use consistent detection systems across antibodies

  • Validation strategy:

    • Confirm results with alternative detection methods (qPCR, enzyme activity)

    • Test antibodies on knockout or siRNA-treated samples

    • Perform pre-absorption studies with immunizing peptides

  • Documentation requirements:

    • Record complete antibody information (supplier, catalog number, lot)

    • Document exact experimental conditions for each antibody

    • Report all optimization steps and protocol variations

How might single-cell analysis techniques be integrated with NOS antibody research?

Single-cell technologies offer new frontiers for NOS research:

  • Mass cytometry (CyTOF) applications:

    • Simultaneous detection of multiple NOS isoforms in heterogeneous cell populations

    • Correlation of NOS expression with dozens of other cellular markers

    • Identification of novel NOS-expressing cell subtypes

  • Single-cell RNA-seq integration:

    • Correlation of protein-level NOS detection with transcriptomic profiles

    • Discovery of novel regulatory networks controlling NOS expression

    • Identification of cell state-specific NOS regulation

  • Spatial transcriptomics approaches:

    • Mapping NOS expression in tissue microenvironments

    • Correlating NOS expression with local signaling gradients

    • Studying NOS regulation in specific tissue niches

  • Technological considerations:

    • Antibody conjugation strategies for multiplexed detection

    • Fixation compatibility with single-cell isolation procedures

    • Data integration methods for multi-omics analysis

What considerations are important when designing experiments to study NOS antibody-mediated signaling?

Investigating NOS antibody-mediated signaling requires specialized approaches:

  • Functional activation/inhibition studies:

    • Test antibodies for agonist/antagonist activity on NOS enzymatic function

    • Evaluate effects on NOS dimerization and complex formation

    • Assess impact on subcellular localization and trafficking

  • Signal pathway analysis:

    • Monitor downstream NO-dependent signaling cascades

    • Measure cGMP production as a functional readout

    • Assess nitrosylation of target proteins following antibody treatment

  • Real-time monitoring approaches:

    • Use NO-sensitive fluorescent probes in conjunction with antibody treatment

    • Implement calcium imaging to correlate with NOS activation

    • Apply FRET-based sensors to detect NOS conformational changes

  • Controls and validation:

    • Include small molecule NOS inhibitors as reference standards

    • Compare effects of Fab fragments versus complete antibodies

    • Validate with genetic approaches (overexpression, knockdown)

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.