NEURL1B Antibody, HRP conjugated

Shipped with Ice Packs
In Stock

Description

Applications in Research

HRP conjugation enhances the utility of NEURL1B antibodies in multiple assays:

  • Western Blotting: Detects NEURL1B at ~41 kDa in lysates from human, mouse, and rat tissues .

  • ELISA: Enables quantitative analysis of NEURL1B levels with high sensitivity (optimal dilution: 1:500–1:5,000) .

  • Immunohistochemistry: Visualizes NEURL1B localization in formalin-fixed paraffin-embedded (FFPE) tissues .

HRP’s enzymatic activity allows detection via chromogenic (e.g., TMB, DAB) or chemiluminescent substrates, amplifying signal-to-noise ratios in low-abundance targets .

Conjugation Methodology

The HRP conjugation process involves:

  1. Antibody Preparation: Use of amine-free buffers (pH 6.5–8.5) devoid of interfering agents (e.g., sodium azide, Tris) .

  2. Modifier Addition: 1 µL modifier per 10 µL antibody to activate conjugation .

  3. Incubation: 3-hour reaction at room temperature for covalent HRP linkage .

  4. Quenching: Addition of quencher reagent to stabilize the conjugate .

Lyophilization steps, as described in modified protocols, enhance conjugation efficiency and stability .

Research Findings and Clinical Relevance

NEURL1B’s role as an E3 ubiquitin ligase in the Notch pathway has implications in developmental biology and oncology. Key studies highlight:

  • Notch Pathway Regulation: NEURL1B ubiquitinates Notch ligands (e.g., DLL1, DLL4), modulating their endocytosis and signaling activity .

  • Disease Associations: Dysregulation of NEURL1B is linked to cancers, though direct mechanistic studies remain limited compared to related proteins like HRP-3 in lung cancer .

Comparative Performance Data

ParameterClassical ConjugationLyophilization-Modified Conjugation
ELISA Detection Limit1:25 dilution1:5,000 dilution (p<0.001p < 0.001)
Stability at 4°C≤6 months≤12 months
Signal IntensityModerateHigh (poly-HRP effect)

Challenges and Optimizations

  • Buffer Compatibility: Avoid nucleophilic components (e.g., BSA, Tris) to prevent conjugation interference .

  • Storage: Glycerol (≤50%) extends shelf life but requires frost-free freezer avoidance .

  • Cross-Reactivity: Predicted homology exceeds 93% across mammals, but validation in non-human samples is recommended .

Product Specs

Buffer
**Preservative:** 0.03% Proclin 300
**Constituents:** 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we are able to ship products within 1-3 business days of receiving your order. Delivery times may vary based on the purchasing method or location. For specific delivery time information, please consult your local distributors.
Synonyms
E3 ubiquitin-protein ligase NEURL1B antibody; E3 ubiquitin-protein ligase NEURL3 antibody; hNeur2 antibody; NEU1B_HUMAN antibody; Neur2 antibody; neuralized-2 alternative protein antibody; neuralized-2 antibody; Neuralized-like protein 1B antibody; Neuralized-like protein 3 antibody; NEURL1B antibody; NEURL3 antibody
Target Names
NEURL1B
Uniprot No.

Target Background

Function
NEURL1B is an E3 ubiquitin-protein ligase that plays a crucial role in regulating the Notch signaling pathway. This regulation is achieved by influencing the stability and activity of several Notch ligands.
Gene References Into Functions
  1. In contrast to Neur1, Neur2 exhibits high expression levels during embryonic brain development and various non-neural tissues. Its mRNA levels decline postnatally. PMID: 19723503
Database Links

HGNC: 35422

OMIM: 615893

KEGG: hsa:54492

STRING: 9606.ENSP00000358815

UniGene: Hs.91521

Subcellular Location
Cytoplasm.
Tissue Specificity
Highest expression in brain, prostate and small intestine. In the brain the levels are higher in fetal than in adult stage. In the adult brain the highest levels are detected in the olfactory system, cerebellar cortex, optic nerve and the frontal lobe.

Q&A

What is NEURL1B and why is it a target of interest for antibody development?

NEURL1B (Neuralized E3 Ubiquitin Protein Ligase 1B) functions as an E3 ubiquitin ligase involved in protein degradation pathways. This protein plays critical roles in cellular processes including protein turnover, signaling pathways, and potentially cell cycle regulation. Various antibodies have been developed targeting different amino acid regions of NEURL1B, including those recognizing the AA 1-275 region, which encompasses important functional domains of the protein . The development of specific antibodies against NEURL1B enables researchers to investigate its expression patterns, localization, and interactions with other proteins in various cellular contexts. Understanding NEURL1B function is particularly relevant in contexts where protein degradation pathways may be dysregulated, such as in certain pathological conditions or cellular stress responses.

How do I determine the appropriate working dilution for a newly acquired HRP-conjugated NEURL1B antibody?

Determining the optimal working dilution for a new HRP-conjugated NEURL1B antibody requires systematic titration experiments across multiple applications. Begin with a dilution series (typically ranging from 1:100 to 1:10,000) based on the manufacturer's recommendations for similar applications . For Western blotting, prepare identical membrane strips with your protein of interest and test multiple antibody dilutions simultaneously. For ELISA applications, use a two-dimensional titration approach, testing both antibody concentration and antigen concentration variables . Optimal dilutions should provide clear specific signal with minimal background. Importantly, different applications (IHC, IF, ELISA, WB) may require different optimal dilutions of the same antibody . Document all optimization parameters including incubation times, temperatures, and blocking reagents used, as these factors significantly impact antibody performance independent of concentration.

What experimental applications are most suitable for HRP-conjugated NEURL1B antibodies?

HRP-conjugated NEURL1B antibodies are particularly well-suited for ELISA, immunohistochemistry (IHC), immunofluorescence (IF), and Western blotting applications . For ELISA applications, these conjugates provide direct detection capabilities, simplifying workflow and potentially increasing sensitivity through elimination of secondary antibody binding variability . In IHC applications, HRP-conjugated antibodies facilitate visualization through chromogenic substrates like diaminobenzidine (DAB), which produces a water-insoluble brown precipitate in the presence of hydrogen peroxide . For Western blotting, these conjugates enable direct one-step detection using enhanced chemiluminescence (ECL) substrates. When designing experiments, researchers should consider the specific substrate compatibility (ABTS, TMB, TMBUS, or DAB) based on the detection system available and the sensitivity requirements of their particular application .

How should I optimize blocking conditions when using HRP-conjugated NEURL1B antibodies for Western blotting?

Optimization of blocking conditions is crucial for maximizing signal-to-noise ratio when using HRP-conjugated NEURL1B antibodies. Begin by testing different blocking agents including 5% non-fat milk, 3-5% BSA, commercial blocking buffers, or casein-based blockers. The optimal blocking agent depends on both the specific antibody characteristics and the nature of your samples . When testing blocking conditions, prepare a matrix experiment varying both blocking agent type and concentration, as well as blocking duration (typically 30 minutes to overnight). For NEURL1B detection, which involves a relatively low-abundance protein, more stringent blocking (longer times, higher blocker concentrations) may be necessary to minimize background. Additionally, incorporate 0.05-0.1% Tween-20 in wash buffers to reduce non-specific binding while preserving specific antibody-antigen interactions. Document all optimization steps systematically, as the optimal blocking conditions established may be applicable to other experiments using similar antibody conjugates.

What detection substrates work best with HRP-conjugated NEURL1B antibodies for different applications?

The optimal detection substrate depends on the specific application, required sensitivity, and signal stability needs. For Western blotting with HRP-conjugated NEURL1B antibodies, enhanced chemiluminescence (ECL) substrates provide excellent sensitivity for detecting low-abundance proteins. For colorimetric detection in ELISA applications, TMB (3,3',5,5'-tetramethylbenzidine) offers a balance of sensitivity and stability, developing a blue color that can be read spectrophotometrically at 650nm or converted to yellow (450nm) with addition of stop solution . For IHC applications, DAB (3,3'-diaminobenzidine) produces a stable brown precipitate that is resistant to fading and compatible with standard mounting media . When maximal sensitivity is required, consider using amplified chemiluminescent substrates containing phenols or luminol derivatives. The selection of substrate should also consider signal development time—rapid development substrates (1-5 minutes) may be preferable for time-sensitive experiments, while slower developing substrates might provide better control over signal intensity.

How can I mitigate high background when using HRP-conjugated NEURL1B antibodies in immunohistochemistry?

Excessive background in immunohistochemistry using HRP-conjugated NEURL1B antibodies can result from multiple factors that require systematic troubleshooting. First, address endogenous peroxidase activity by incorporating a dedicated quenching step using 0.3-3% hydrogen peroxide in methanol for 10-30 minutes prior to primary antibody incubation . Next, optimize antibody concentration through careful titration experiments; HRP-conjugated antibodies often require more dilute working concentrations than unconjugated versions . Increase the stringency of wash steps by extending wash durations and incorporating higher salt concentrations (up to 500mM NaCl) to disrupt weak non-specific interactions. Consider implementing a dual blocking strategy using both protein blockers (BSA or casein) and serum from the same species as the tissue being stained. For particularly challenging samples, adding 0.1-0.3% Triton X-100 to wash buffers can reduce hydrophobic interactions that contribute to background. Finally, if background persists, consider modifying the visualization approach by using more dilute DAB substrate and monitoring color development closely to stop the reaction before background becomes problematic .

What strategies can be employed to extend the shelf-life of HRP-conjugated NEURL1B antibodies?

Extending the shelf-life of HRP-conjugated NEURL1B antibodies requires addressing multiple factors that contribute to degradation and loss of activity. Store antibody aliquots at -20°C to -80°C in single-use volumes to minimize freeze-thaw cycles, which significantly impact conjugate stability. Incorporate stabilizing agents such as 50% glycerol, 1% BSA, and 0.02-0.05% sodium azide in storage buffers, noting that azide should be removed before use as it can inhibit HRP activity . Commercial stabilizers like LifeXtend™ HRP conjugate stabilizer protect antibody-HRP conjugates from environmental factors that compromise performance . Maintain proper pH conditions (typically pH 7.2-7.6) as HRP activity is highly pH-dependent. During repeated use, keep antibody solutions on ice and return to appropriate storage conditions promptly after use. For working dilutions that must be stored short-term, add 1-2% BSA as a stabilizing protein. Implement rigorous monitoring protocols including regular activity testing against positive controls to track potential degradation over time.

How can I determine if poor signal is due to antibody degradation or suboptimal experimental conditions?

Distinguishing between antibody degradation and suboptimal experimental conditions requires a systematic troubleshooting approach. First, test the HRP activity directly using a small aliquot of the conjugated antibody with TMB or ABTS substrate in a simple spot test on nitrocellulose membrane—absence of color development suggests HRP inactivation . Second, run parallel experiments with a well-characterized positive control antibody (preferably another HRP-conjugated antibody) using identical protocols; if the control antibody works while the NEURL1B antibody fails, this suggests antibody-specific issues. Third, verify target protein presence using an alternate detection method or a different antibody against the same target but recognizing a different epitope . Fourth, implement a gradient experiment testing multiple parameters simultaneously (antibody concentration, incubation time, temperature) to determine if signal can be recovered through protocol modification. Finally, examine reagent compatibility issues—certain buffer components (high concentrations of reducing agents, metal chelators, or detergents) can inactivate HRP or interfere with antibody-antigen binding . Document all troubleshooting steps methodically to establish a reference for future experiments.

How might the epitope location within NEURL1B affect experimental outcomes when using different HRP-conjugated antibodies?

The epitope location within NEURL1B significantly impacts experimental outcomes through multiple mechanisms that require careful consideration. Antibodies targeting different regions of the protein (e.g., AA 1-275 vs. AA 233-262 vs. AA 245-294) may exhibit different accessibility to the target epitope in various experimental conditions . For instance, antibodies recognizing epitopes within functional domains might be occluded when the protein engages in protein-protein interactions, potentially leading to false-negative results in co-immunoprecipitation studies. Epitopes located in structurally flexible regions may be more accessible but potentially less specific. Conformational changes induced by experimental conditions (denaturing vs. native) differentially affect epitope recognition depending on location—antibodies against linear epitopes often perform better in denaturing conditions (Western blot), while those against conformational epitopes are optimal for native applications (IP, IF) . Additionally, post-translational modifications near the epitope (phosphorylation, ubiquitination, etc.) may sterically hinder antibody binding, creating condition-dependent detection variability. When designing critical experiments, researchers should ideally validate findings using multiple antibodies targeting different NEURL1B epitopes to ensure comprehensive and accurate results.

What considerations should be made when designing experiments to investigate NEURL1B interactions with chromatin-binding proteins?

Investigating NEURL1B interactions with chromatin-binding proteins requires specialized experimental approaches that account for the complex nuclear environment. Based on mechanistic insights from studies of similar proteins like HRP-3, which directly binds to the E2F1 promoter on chromatin through its PWWP domain , several methodological considerations are critical. First, implement chromatin fractionation protocols to separate chromatin-bound and non-chromatin-bound protein fractions when assessing NEURL1B localization, similar to approaches used for HRP-3 . Second, design chromatin immunoprecipitation (ChIP) assays using HRP-conjugated NEURL1B antibodies optimized for fixed chromatin samples, with appropriate sonication parameters to generate 200-500bp DNA fragments for optimal resolution. Third, include controls for non-specific binding, such as IgG-HRP conjugates matched to the host species of the NEURL1B antibody. Fourth, validate protein-chromatin interactions using orthogonal approaches such as DNA pull-down assays with biotinylated target DNA sequences. Finally, consider the impact of cell cycle phase on chromatin structure and accessibility, potentially synchronizing cells to examine phase-specific interactions, as chromatin-binding patterns often vary throughout the cell cycle, particularly for proteins involved in regulatory processes .

How can I design experiments to investigate whether NEURL1B plays a role in histone modification similar to that observed with HRP-3?

Designing experiments to investigate NEURL1B's potential role in histone modification requires a multifaceted approach based on mechanistic insights from related proteins like HRP-3. HRP-3 has been shown to affect histone H3/H4 acetylation and influence interactions between these histones and HDAC1/2 on promoter regions . First, implement NEURL1B knockdown experiments using siRNA or CRISPR-Cas9 approaches, followed by western blotting and immunofluorescence to assess global changes in histone modifications (particularly H3/H4 acetylation) . Second, perform ChIP-seq experiments using antibodies against specific histone modifications before and after NEURL1B depletion to identify genomic regions where NEURL1B might regulate histone modification states. Third, conduct co-immunoprecipitation experiments using HRP-conjugated NEURL1B antibodies to identify potential interactions with histone-modifying enzymes such as HDACs, HATs, or other chromatin remodelers . Fourth, implement ChIP-re-ChIP approaches to determine if NEURL1B and specific histone marks co-occupy the same genomic regions. Finally, assess functional outcomes of these interactions through gene expression analysis (RNA-seq) after NEURL1B manipulation, potentially with and without HDAC inhibitors like TSA to determine if there are synergistic effects similar to those observed with HRP-3 .

What are the most appropriate quantification methods for Western blots using HRP-conjugated NEURL1B antibodies?

Quantification of Western blots using HRP-conjugated NEURL1B antibodies requires rigorous approaches to ensure accurate and reproducible results. Implement densitometric analysis using specialized software (ImageJ, Image Lab, etc.) that can accurately measure band intensity relative to background. Establish a linear dynamic range for the specific HRP-conjugated antibody by creating a standard curve with serial dilutions of a positive control sample—this defines the quantifiable range where signal intensity correlates linearly with protein amount . Always normalize NEURL1B signals to appropriate loading controls (β-actin, GAPDH, or total protein stains like Ponceau S) processed on the same blot under identical conditions. When comparing across multiple blots, include a common reference sample on each blot as an inter-blot calibrator. For kinetic studies or comparisons across treatment conditions, express results as fold-change relative to control conditions rather than absolute values. Statistical analysis should incorporate data from at least three independent biological replicates, with appropriate statistical tests (typically ANOVA with post-hoc tests for multiple comparisons) to determine significance. Document all image acquisition parameters including exposure time, gain settings, and any image processing applied.

How can I validate the specificity of HRP-conjugated NEURL1B antibody signals in my experimental system?

Validating the specificity of HRP-conjugated NEURL1B antibody signals requires multiple complementary approaches to distinguish true signals from artifacts. First, implement genetic validation by performing parallel experiments in NEURL1B-knockdown or knockout models—specific signals should diminish proportionally to the degree of target depletion . Second, conduct peptide competition assays where the antibody is pre-incubated with excess immunizing peptide (if available) before application to samples; specific signals should be competitively inhibited while non-specific signals remain. Third, compare reactivity patterns across multiple antibodies targeting different NEURL1B epitopes; convergent detection patterns increase confidence in specificity . Fourth, verify molecular weight correspondence—NEURL1B should appear at its predicted molecular weight (accounting for any post-translational modifications) in Western blots. Fifth, implement appropriate negative controls including samples known to lack NEURL1B expression and isotype control antibodies. Finally, cross-validate findings using orthogonal detection methods such as mass spectrometry or RNA expression analysis to confirm protein identity and expression patterns. Document all validation steps methodically to establish a reference for future experiments and publications.

What statistical approaches are recommended when analyzing data from complex experiments involving HRP-conjugated NEURL1B antibodies?

Statistical analysis of complex experiments involving HRP-conjugated NEURL1B antibodies requires approaches that address both technical and biological variability. For experiments comparing NEURL1B levels across multiple conditions, implement two-way ANOVA to account for both treatment effects and biological replication, followed by appropriate post-hoc tests (Tukey's or Bonferroni) for multiple comparisons . When analyzing co-localization studies (e.g., NEURL1B with chromatin markers), calculate Pearson's or Mander's correlation coefficients rather than relying on qualitative assessment. For ChIP experiments investigating NEURL1B chromatin binding (similar to approaches used for HRP-3), employ percent input normalization followed by fold-enrichment calculations relative to control regions and IgG controls . When comparing ChIP data across conditions, apply non-parametric tests (Mann-Whitney U or Kruskal-Wallis) as ChIP data often violates normality assumptions. For dose-response experiments, fit data to appropriate mathematical models (4-parameter logistic regression for sigmoidal responses) to determine EC50 values and other pharmacological parameters. Implement power analysis during experimental design to ensure sufficient replication for detecting biologically meaningful effects. Finally, consider employing data visualization approaches like principal component analysis for complex multivariate datasets to identify patterns and relationships that might not be apparent from univariate statistics.

Table of NEURL1B Antibody Characteristics and Applications

Antibody VariantTarget RegionHost SpeciesClonalityConjugate TypePrimary ApplicationsCross-ReactivityPurification Method
ABIN7151082AA 1-275RabbitPolyclonalUnconjugatedELISA, IHC, IFHumanProtein G, >95%
Variant 1AA 1-275RabbitPolyclonalHRPELISAHumanProtein G
Variant 2AA 1-275RabbitPolyclonalBiotinELISAHumanProtein G
Variant 3AA 1-275RabbitPolyclonalFITCNot specifiedHumanNot specified
Variant 4AA 233-262RabbitPolyclonalUnconjugatedELISA, WBHumanNot specified
Variant 5AA 233-262RabbitPolyclonalBiotinELISA, WBHumanNot specified
Variant 6Internal RegionRabbitPolyclonalUnconjugatedELISA, WB, IHCHuman, MouseNot specified

Data compiled from antibodies-online catalog information

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.