hns Antibody, HRP conjugated

Shipped with Ice Packs
In Stock

Description

Overview of HRP-Conjugated Antibodies

HRP (horseradish peroxidase)-conjugated antibodies are secondary antibodies chemically linked to the HRP enzyme. These conjugates amplify detection signals in immunoassays by catalyzing reactions with substrates like DAB or TMB, producing measurable colorimetric, chemiluminescent, or fluorescent signals .

Crosslinking Techniques

MethodReagents UsedTarget Groups
Periodate OxidationSodium periodate, NaCNBH₃Polysaccharide chains
Maleimide-ThiolSulfo-SMCC, SATASulfhydryl groups
NHS-EsterEDC/NHSAmine groups

Source:

Optimized conjugates retain >90% antigen-binding capacity and enzymatic activity when purified from interfering buffer components (e.g., amino acids, thiols) .

Comparative Analysis of HRP-Conjugated Antibody Performance

ApplicationDetection Limit (LOD)Signal TypeSubstrate Example
ELISA~22–25 pMColorimetric/ChemiluminescentTMB, ABTS
Western BlottingLow ng/mL rangeChemiluminescentECL
IHC (Paraffin)High sensitivityChromogenicDAB

Data aggregated from

HRP-conjugated antibodies excel in high-throughput screening due to rapid signal kinetics and minimal background noise .

Step-by-Step Conjugation Using Sulfo-SMCC (Heterobifunctional Crosslinker)

  1. Activate HRP: React HRP with sulfo-SMCC to introduce maleimide groups .

  2. Thiolate Antibody: Introduce sulfhydryl groups to the antibody using SATA .

  3. Conjugate: Mix maleimide-activated HRP with thiolated antibody, followed by purification .

Critical Considerations:

  • Antibody purity and buffer composition (avoid amines/thiols) .

  • Conjugation efficiency: Typically achieves 3 HRP molecules per antibody .

Limitations and Alternatives

While HRP conjugates offer robust signals, limitations include:

  • Enzyme Instability: HRP activity diminishes at high temperatures or extreme pH .

  • Polymerization Risk: Over-conjugation may reduce antigen-binding affinity .

Recombinant alternatives like GST-ABD (a secondary antibody mimic with 3 HRPs per molecule) provide cost-effective, animal-free solutions but exhibit slightly weaker binding affinity (K<sub>d</sub> ~1.31 nM) .

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 the products within 1-3 business days after receiving your order. Delivery time may vary depending on the purchasing method or location. For specific delivery times, please consult your local distributors.
Synonyms
hns antibody; bglY antibody; cur antibody; drdX antibody; hnsA antibody; msyA antibody; osmZ antibody; pilG antibody; topS antibody; b1237 antibody; JW1225 antibody; DNA-binding protein H-NS antibody; Heat-stable nucleoid-structuring protein antibody; Histone-like protein HLP-II antibody; Protein B1 antibody; Protein H1 antibody
Target Names
hns
Uniprot No.

Target Background

Function
H-NS is a DNA-binding protein involved in transcriptional repression (silencing). It plays a role in bacterial chromosome organization and compaction. H-NS binds tightly to AT-rich double-stranded DNA and inhibits transcription. It binds upstream and downstream of the initiating RNA polymerase, effectively trapping it in a loop and preventing transcription. H-NS interacts with hundreds of sites throughout the genome, approximately half of which are located in non-coding DNA, comprising about 10% of the genome. Many of these loci were horizontally transferred (HTG), providing the selective advantage of silencing foreign DNA while retaining it in the genome for potential future use. H-NS also suppresses transcription at numerous intragenic sites, as well as the transcription of spurious, non-coding RNAs across the entire genome. Repression of HTG by H-NS allows their DNA to evolve faster than non-H-NS-bound regions, facilitating the integration of HTG into transcriptional regulatory networks. A subset of H-NS/StpA-regulated genes also require Hha (and/or Cnu, ydgT) for repression; Hha and Cnu increase the number of genes bound by H-NS/StpA and potentially modulate the oligomerization of the H-NS/StpA-complex. The protein forms two clusters in the nucleoid, which gather hns-bound loci together, bridging non-contiguous DNA and causing significant DNA condensation. H-NS binds DNA more efficiently at lower temperatures compared to 37 degrees Celsius. AT-rich sites nucleate H-NS binding, further DNA binding is cooperative, and this cooperativity decreases with increasing temperature. Transcriptional repression can be inhibited by dominant-negative mutants of StpA or H-NS itself. H-NS may also influence transcriptional elongation and can enhance the translational efficiency of mRNA with suboptimal Shine-Dalgarno sequences. It plays a role in the thermal control of pili and adhesive curli fimbriae production, by inducing transcription of csgD. H-NS also contributes to flagellar function. It represses the CRISPR-cas promoters, allowing only weak transcription of the crRNA precursor; its repression is countered by LeuO. H-NS exhibits preferential binding to the upstream region of its own gene, recognizing two DNA segments flanking a bend centered around -150. Overexpression of H-NS suppresses secY24, a temperature-sensitive mutation. It has also been reported to activate transcription of certain genes.
Gene References Into Functions
  1. H-NS specifically represses LEE5 promoter activity, and Ler alleviates this repression. PMID: 28013045
  2. Transcription and stability of cas3 in Deltahns cells are limiting for resistance to phage. PMID: 26956996
  3. We propose that, during E. coli evolution, the conservation of H-NS binding sites resulted in the diversification of the regulation of horizontally transferred genes, which may have facilitated E. coli adaptation to new ecological niches. PMID: 26789284
  4. Through DNA binding, the two proteins interact to form a transient ternary complex Fpg/DNA/HU, leading to the release of Fpg and the molecular entrapment of SSBs by HU. PMID: 26392572
  5. The fdeC gene encodes a protein that is expressed at the cell surface and promotes biofilm formation under continuous-flow conditions; its expression is repressed by the global regulator H-NS. PMID: 25239893
  6. It was concluded that H-NS exerts maximum repression of LEE5 via the specific sequence at around -138 and subsequently contacts a subunit of RNAP through oligomerization. PMID: 24610333
  7. Bicyclomycin treatment leads to a decrease in binding signal for H-NS to the E. coli chromosome. PMID: 24499790
  8. Both the DNA binding sites of H-NS as well as the function of StpA as a backup system might be selected for silencing highly transcribable genes. PMID: 23661089
  9. We demonstrate that H-NS modulates the expression of the nrdAB and nrdDG operons in aerobically and in anaerobically growing cells. PMID: 23873909
  10. H-NS represses LEE5p by binding to a cluster of A tracks upstream of -114 of the promoter. PMID: 22924981
  11. RcsB-BglJ activates one of two H-NS-StpA repressed leuO promoters. PMID: 22295907
  12. The product of bacteriophage T7 gene 5.5 (gp5.5) forms a stable complex with the Escherichia coli histone-like protein H-NS and transfer RNAs (tRNAs). PMID: 22566619
  13. The attractive force governing the Cnu-H-NS interaction is an ionic bond, unlike the hydrophobic interaction that is the major attractive force in most proteins. PMID: 22358512
  14. HNS controls the integrase IntI1 and integron promoters expression. PMID: 21778209
  15. LeuO is a major player in antagonistic interplay against the universal silencer H-NS. PMID: 21883529
  16. It binds to a middle domain of FliG that bridges the core parts of the rotor and parts nearer the edge that interact with the stator. PMID: 21890701
  17. Rho-dependent transcription termination is regulated by the H-NS family of proteins. PMID: 21602341
  18. Combinations of the rho, nusG, and nusA mutations were synthetically lethal, and the lethality was suppressed by expression of H-NS. PMID: 21602355
  19. A new insight into the bolA regulation network demonstrating that H-NS represses the transcription of this important gene was provided. PMID: 21708124
  20. H-NS formed 2 compact clusters per chromosome, driven by oligomerization of DNA-bound H-NS through interactions mediated by the amino-terminal domain of the protein; observations demonstrate H-NS plays a key role in global chromosome organization. PMID: 21903814
  21. Chromosomal and plasmid-encoded H-NS proteins differ in their functional properties. PMID: 21320594
  22. H-NS binds to long tracts of DNA, consistent with the linear spread of H-NS binding from high- to surrounding lower-affinity sites; the length of binding regions is associated with the degree of transcriptional repression imposed by H-NS. PMID: 21097887
  23. H-NS directly controls regulators of stress resistance. PMID: 21034467
  24. H-NS and LeuO are antagonistic regulators of CRISPR-based immunity. PMID: 20659289
  25. The global regulator H-NS is required for extracellular DNA (eDNA) production, as DNA was not detected for the hns mutant, and production of H-NS restored eDNA production to wild-type levels. PMID: 20833130
  26. Findings substantiate the central role of RcsB in H-NS-mediated control of motility and acid stress resistance. PMID: 20435136
  27. These results demonstrate an active involvement of H-NS in the induction of the CRISPR-cas system and suggest a potential link between two prokaryotic defense systems against foreign DNA. PMID: 20132443
  28. Binding of the IHF protein to a site immediately adjacent to fimS is required for phase-on orientational bias; in the absence of LRP and IHF binding, fimS adopts the off orientation, and the H-NS protein is necessary to maintain this orientational bias. PMID: 19889099
  29. Protein-induced DNA bending plays a significant role in HU site-specific DNA binding and supports a model of a mutually induced fit. PMID: 15322284
  30. Forms complexes with YdgT in vivo. PMID: 15458420
  31. Data show that H-NS acts to repress the expression of traM and traJ as cells enter stationary phase, thereby decreasing mating ability to barely detectable levels. PMID: 15491366
  32. Analysis of the mechanical response generated by binding of DNA-bending protein Hu to single tethered 48.5 kb lambda-DNA molecules finds that compaction of DNA increases with increasing Hu concentration. PMID: 15504049
  33. A key role for intrinsic conformational changes of Hha in modulating its interaction with H-NS. PMID: 15720293
  34. H-NS directly inhibits gadA and gadX transcription and, by controlling the intracellular level of the activator GadX, indirectly affects the expression of the whole gad system. PMID: 15795232
  35. The expression of the H-NS regulon is sensitive to small changes in the cellular level of H-NS, enabling the cell to respond rapidly to environmental cues. PMID: 15819627
  36. Results show that LRP binds to the regulatory region of bacterial rRNA promoters, and may contribute in combination with H-NS to the control of rRNA synthesis. PMID: 16238633
  37. The oligomerization capacity of H-NS in vivo is affected by temperature, pH, and osmolarity. PMID: 16303134
  38. StpA complements H-NS proteins defective in DNA binding to repress bgl, while in autoregulation of stpA it acts autonomously, indicating a difference in the mechanisms of repression. PMID: 16980475
  39. Role of hns in regulation of RpoS; findings suggest that H-NS regulates an RssB inhibitor or inhibitors. PMID: 16980505
  40. The results support the conclusion that the bacterial DNA-binding protein LRP, assisted by H-NS, forms a repressive nucleoprotein structure involved in regulation of rRNA transcription. PMID: 17196617
  41. Identified as a new member of the polyamine modulon together with RpoN and Cra. PMID: 17220219
  42. Crystal structure of the Escherichia coli nucleoid-associated HUalphabeta protein. PMID: 17360520
  43. The binding of H-NS to a transposase protein is a novel function for this important regulatory molecule. PMID: 17501923
  44. Analysis of H-NS repression by binding within the transcription unit. PMID: 17569663
  45. Data show that H-NS prevented binding of RNA polymerase (RNAP) at the Hemolysin Protein E promoter (PhlyE). PMID: 17892462
  46. The model for the functions of H-NS in homologous recombination and double-strand break repair is discussed. PMID: 17991999
  47. LeuO and H-NS regulate the expression of the yjjQ-bglJ operon and yjjP. PMID: 18055596
  48. H-NS is shown to be a common regulator of multiple iron and other nutrient acquisition systems preferentially expressed at 37 degrees C and of general stress response, biofilm formation, and cold shock genes highly expressed at 23 degrees C. PMID: 19011022
  49. At 37 degrees C, SlyA activates transcription of K5 capsule independent of H-NS, but maximal transcription requires H-NS. PMID: 19114478
  50. StpA binding profile reduced to one-third in the hns mutant, and the H-NS binding profile was unaffected by stpA inactivation. PMID: 19151137

Show More

Hide All

Database Links
Protein Families
Histone-like protein H-NS family
Subcellular Location
Cytoplasm, nucleoid.

Q&A

What is the hns Antibody, HRP conjugated and what is its primary research application?

The hns Antibody, HRP conjugated is a high-quality polyclonal antibody with reactivity against Escherichia coli samples, specifically targeting the hns protein (histone-like nucleoid structuring protein). This antibody has been validated for ELISA applications and comes pre-conjugated with horseradish peroxidase (HRP) . The primary application of this antibody is in detecting the hns protein in E. coli samples through various immunodetection methods, particularly ELISA. HRP conjugation eliminates the need for secondary antibody incubation steps, streamlining experimental workflows while maintaining high sensitivity and specificity for the target protein.

How does HRP conjugation enhance antibody detection capabilities?

HRP conjugation significantly enhances antibody detection capabilities through enzymatic signal amplification. When conjugated to an antibody, HRP (a 40-44 kDa glycoprotein enzyme) catalyzes the reaction of hydrogen peroxide with various chromogenic substrates to produce detectable signals . This enzymatic reaction generates rapid and intense signals, making it ideal for non-fluorescent applications like Western blotting, ELISA, and immunohistochemistry (IHC) . Compared to non-conjugated antibodies that require secondary detection reagents, HRP-conjugated antibodies provide direct signal generation, reducing experimental time, background noise, and potential cross-reactivity issues that can occur with secondary antibody systems.

What detection substrates are compatible with hns Antibody, HRP conjugated?

The hns Antibody, HRP conjugated can be used with a wide range of substrates, each offering different detection modalities and sensitivity levels:

Substrate TypeExampleDetection MethodRelative SensitivityBest Application
ChromogenicTetramethylbenzidine (TMB)ColorimetricMediumELISA
ChromogenicDiaminobenzidine (DAB)ColorimetricMediumIHC
ChromogenicABTSColorimetricMediumELISA
ChemiluminescentLuminolLuminescenceHighWestern blotting
FluorogenicVariousFluorescenceHighSpecialized applications

The choice of substrate depends on the specific research application, required sensitivity, and detection method availability . For quantitative applications like ELISA, TMB or ABTS are commonly used due to their stable color development. For Western blotting applications requiring higher sensitivity, chemiluminescent substrates like luminol provide superior detection limits.

What controls should be included when using hns Antibody, HRP conjugated in ELISA experiments?

When designing ELISA experiments with hns Antibody, HRP conjugated, several critical controls should be incorporated to ensure reliable and interpretable results:

  • Negative Control: Include wells with all reagents except the primary antibody (hns Antibody, HRP conjugated) to assess non-specific binding and background signal generation.

  • Positive Control: Use a sample known to contain the hns protein, ideally at a defined concentration, to confirm antibody functionality.

  • Blank Control: Include wells with all components except the antigen to establish the baseline signal.

  • Specificity Control: Test the antibody against non-E. coli samples or E. coli samples with hns gene deletion to confirm specificity.

  • Dilution Controls: Prepare a dilution series of positive samples to establish the linear detection range and determine the limit of detection (LOD) .

  • Substrate Control: Include wells with substrate only to assess spontaneous substrate oxidation.

These controls help distinguish between true positive signals and experimental artifacts, ensuring experimental validity and reliable data interpretation. For advanced experimental design, consider including isotype controls and competitive inhibition controls with purified hns protein.

How can I determine the optimal working dilution for hns Antibody, HRP conjugated?

Determining the optimal working dilution for hns Antibody, HRP conjugated requires systematic titration to balance signal intensity with background noise. The following methodological approach is recommended:

  • Prepare a dilution series of the antibody (typically ranging from 1:100 to 1:10,000) in appropriate assay buffer.

  • Run parallel assays using known positive samples at a fixed concentration.

  • Calculate the signal-to-noise ratio for each dilution by dividing the positive sample signal by the negative control signal.

  • Plot signal intensity versus antibody dilution to identify the dilution that provides maximum signal while maintaining low background.

  • Validate the selected dilution across multiple experimental replicates to ensure reproducibility.

Optimal dilutions typically fall in the range where the signal-to-noise ratio is highest, rather than where absolute signal intensity peaks . This approach maximizes detection sensitivity while minimizing reagent consumption. For hns Antibody, HRP conjugated, verification of the optimal dilution should be performed for each new lot of antibody and experimental system.

How does the chemical structure of the HRP-antibody conjugate affect detection sensitivity in complex E. coli lysates?

The chemical structure of the HRP-antibody conjugate significantly impacts detection sensitivity, especially in complex E. coli lysates where multiple proteins may cross-react. The conjugation chemistry determines several critical parameters:

  • Enzyme-to-Antibody Ratio (E/A): Higher E/A ratios generally increase signal intensity but may compromise antigen binding if conjugation occurs near the antigen-binding site. Optimal E/A ratios for hns detection typically range from 2-4 HRP molecules per antibody .

  • Linkage Stability: The stability of the chemical bond between HRP and the antibody affects signal consistency across experiments. Methods using maleimide-thiol chemistry typically produce more stable conjugates than periodate oxidation methods .

  • Spatial Orientation: The relative positioning of HRP to the antibody's antigen-binding site can affect accessibility to the hns protein, particularly in crowded molecular environments like cell lysates.

  • Steric Hindrance: Excessive HRP conjugation may create steric hindrance that reduces antibody binding efficiency, particularly for intracellular or membrane-bound forms of hns protein.

Advanced researchers should consider using site-specific conjugation methods that attach HRP away from the antibody's antigen-binding regions to preserve binding affinity while maximizing signal generation . Comparative analysis using different conjugation chemistries can help identify the optimal approach for specific experimental conditions.

What are the molecular mechanisms behind false-positive signals when using hns Antibody, HRP conjugated, and how can they be mitigated?

False-positive signals when using hns Antibody, HRP conjugated can arise from multiple molecular mechanisms, each requiring specific mitigation strategies:

  • Endogenous Peroxidase Activity: E. coli samples may contain endogenous peroxidases that can oxidize HRP substrates independently of the antibody-antigen interaction.

    • Mitigation: Include a peroxidase quenching step (e.g., 0.3% H₂O₂ in methanol for 30 minutes) before antibody application.

  • Non-specific Antibody Binding: The polyclonal nature of hns Antibody may lead to binding to proteins structurally similar to hns.

    • Mitigation: Use more stringent blocking solutions (e.g., 5% BSA with 0.1% Tween-20) and increase wash stringency.

  • Protein A/G Interaction: Some E. coli strains express proteins that bind immunoglobulins independent of antigen recognition.

    • Mitigation: Pre-adsorb the antibody with an E. coli lysate lacking the hns protein.

  • HRP Substrate Auto-oxidation: Some substrates (particularly TMB) can undergo auto-oxidation under certain buffer conditions.

    • Mitigation: Optimize substrate composition and include appropriate stabilizers.

  • Aggregated Antibody Binding: Antibody aggregates can bind non-specifically to sample components.

    • Mitigation: Centrifuge antibody solution at 10,000g for 5 minutes before use to remove aggregates.

Advanced researchers should consider implementing dual-detection systems or proximity ligation assays to increase specificity for particularly challenging samples . Additionally, comparative analysis with multiple antibodies targeting different epitopes of the hns protein can help distinguish true from false signals.

What factors contribute to batch-to-batch variability in hns Antibody, HRP conjugated performance, and how can I standardize results?

Batch-to-batch variability in hns Antibody, HRP conjugated performance can significantly impact experimental reproducibility. Key contributing factors and standardization approaches include:

  • Conjugation Efficiency Variation: Different batches may have varying numbers of HRP molecules per antibody.

    • Standardization: Determine the E/A ratio for each batch and adjust dilutions accordingly.

  • HRP Enzymatic Activity Differences: The specific activity of HRP can vary between preparations.

    • Standardization: Perform activity assays on each new batch using standard substrates and normalize usage accordingly.

  • Antibody Affinity Shifts: Slight variations in epitope recognition can occur between polyclonal antibody batches.

    • Standardization: Maintain reference samples with known hns content for comparative analysis.

  • Storage Degradation: HRP activity may decrease over time, particularly with repeated freeze-thaw cycles.

    • Standardization: Aliquot new batches, avoid freeze-thaw cycles, and establish standard curves with each experiment.

  • Buffer Composition Variations: Minor changes in buffer components can affect antibody performance.

    • Standardization: Prepare and use consistent buffer formulations.

To achieve optimal standardization, researchers should maintain detailed records of antibody performance metrics across batches and implement internal reference standards. Consider using recombinant hns protein at known concentrations to generate standard curves with each new batch . Additionally, adopting automated liquid handling systems for antibody dilution and application can significantly reduce operator-dependent variability.

How can I optimize the signal-to-noise ratio when detecting low-abundance hns protein in environmental E. coli isolates?

Detecting low-abundance hns protein in environmental E. coli isolates presents unique challenges requiring specialized optimization approaches:

  • Sample Enrichment Techniques:

    • Implement immunoprecipitation before detection to concentrate hns protein.

    • Use subcellular fractionation to isolate nuclear-associated fractions where hns typically localizes.

  • Signal Amplification Strategies:

    • Implement tyramide signal amplification (TSA) to enhance HRP signal by 10-100 fold.

    • Consider using enhanced chemiluminescent substrates with extended emission kinetics.

  • Background Reduction Methods:

    • Increase washing stringency with higher salt concentrations (150-500 mM NaCl).

    • Add non-ionic detergents (0.1-0.3% Triton X-100) to reduce hydrophobic interactions.

    • Include competitors for non-specific binding sites (1-5% non-fat dry milk or BSA).

  • Optimized Detection Parameters:

    • Extend substrate incubation times from standard 5-10 minutes to 15-30 minutes for low-abundance targets.

    • Use cooled CCD cameras or high-sensitivity photomultiplier tubes for signal detection.

  • Comparative Quantification Approach:

    • Implement internal standard curves using recombinant hns protein.

    • Use digital image analysis with background subtraction algorithms.

For extremely low-abundance detection, consider implementing a proximity ligation assay that can provide single-molecule sensitivity . Additionally, incorporating sequential multiple amplification steps through biotin-streptavidin bridges before final HRP detection can significantly boost signal intensity for challenging environmental samples.

How can I use hns Antibody, HRP conjugated to investigate hns protein binding interactions with bacterial DNA in chromatin immunoprecipitation experiments?

Chromatin immunoprecipitation (ChIP) using hns Antibody, HRP conjugated can reveal detailed insights into hns-DNA interactions. The following methodology optimizes this approach:

  • Crosslinking Optimization:

    • For hns protein, optimize formaldehyde concentration (0.5-1.0%) and crosslinking time (10-20 minutes) to capture transient DNA interactions without over-fixation.

    • Consider using protein-protein crosslinkers (e.g., DSG) before formaldehyde treatment to stabilize multiprotein complexes.

  • Chromatin Fragmentation:

    • Sonicate to achieve DNA fragments of 200-500 bp for optimal resolution.

    • Verify fragmentation efficiency through agarose gel electrophoresis before proceeding.

  • Immunoprecipitation Strategy:

    • Pre-clear lysates with protein A/G beads to reduce non-specific binding.

    • Directly conjugate hns Antibody, HRP to magnetic beads using the peroxidase activity as a crosslinking catalyst.

    • Include parallel immunoprecipitations with non-specific IgG-HRP as controls.

  • Washing Conditions:

    • Implement increasingly stringent wash buffers (150 mM to 500 mM NaCl) to reduce background.

    • Include LiCl washes (250 mM) to disrupt ionic interactions.

  • DNA Recovery and Analysis:

    • Reverse crosslinks at 65°C overnight followed by proteinase K treatment.

    • Analyze immunoprecipitated DNA through qPCR, sequencing, or microarray approaches.

The direct HRP conjugation allows for additional verification of successful immunoprecipitation by sampling a small aliquot of the beads for peroxidase activity testing . For genome-wide binding studies, ChIP-seq analysis can be performed on the immunoprecipitated DNA, providing comprehensive mapping of hns binding sites across the bacterial genome.

What advanced experimental approaches can assess potential cross-reactivity between hns Antibody, HRP conjugated and other nucleoid-associated proteins in E. coli?

Assessing cross-reactivity between hns Antibody, HRP conjugated and other nucleoid-associated proteins requires sophisticated analytical approaches:

This comprehensive approach provides quantitative assessment of potential cross-reactivity, enabling researchers to appropriately interpret experimental results . For studies requiring absolute specificity, consider using competitive inhibition with excess recombinant hns protein as a control condition to identify and subtract non-specific signals.

How can I adapt hns Antibody, HRP conjugated for multiplex detection systems to simultaneously visualize hns localization alongside other bacterial proteins?

Adapting hns Antibody, HRP conjugated for multiplex detection requires sophisticated methodological approaches that preserve HRP activity while enabling discrimination between multiple targets:

  • Sequential Multiplex Detection:

    • Implement cyclic immunofluorescence with tyramide signal amplification (TSA).

    • Apply hns Antibody, HRP conjugated first, develop with fluorophore-conjugated tyramide.

    • Inactivate HRP using hydrogen peroxide (3%) treatment.

    • Apply subsequent antibodies against other targets with different detection systems.

  • Spectral Separation Strategies:

    • Convert HRP signal to spectrally distinct fluorophores using TSA with different fluorescent tyramides.

    • Combine with antibodies directly labeled with quantum dots or other fluorophores.

    • Use spectral unmixing algorithms to separate overlapping signals.

  • Spatial Separation Techniques:

    • Employ multi-epitope ligand cartography (MELC) with sequential imaging.

    • Between cycles, photobleach fluorophores completely before applying the next antibody set.

  • Combined Enzymatic Approaches:

    • Use HRP-conjugated hns antibody alongside antibodies conjugated to alternative enzymes (alkaline phosphatase, β-galactosidase).

    • Develop with enzyme-specific substrates yielding products with distinct spectral properties.

  • Proximity-Based Detection:

    • Implement proximity ligation assay (PLA) to visualize protein-protein interactions involving hns.

    • Combine with standard immunofluorescence to correlate interactions with protein localization.

Each approach has specific advantages depending on the experimental question. For co-localization studies, spectral separation with TSA provides excellent sensitivity and resolution . For protein interaction studies, proximity-based methods offer superior specificity and quantitative capacity for analyzing molecular-scale associations between hns and other bacterial proteins.

What methodological approaches can improve the stability and shelf-life of hns Antibody, HRP conjugated for long-term research projects?

Enhancing the stability and shelf-life of hns Antibody, HRP conjugated requires addressing both protein stability and enzyme activity preservation:

  • Buffer Optimization:

    • Replace standard PBS with 50 mM HEPES buffer (pH 7.0-7.4) containing 150 mM NaCl.

    • Add stabilizing agents: 0.1% BSA (protectant), 0.01% thimerosal (antimicrobial), and 30% glycerol (cryoprotectant).

    • Include antioxidants such as 1 mM EDTA and 0.05% sodium azide to prevent oxidative damage.

  • Storage Condition Optimization:

    • Prepare single-use aliquots in non-stick microcentrifuge tubes.

    • Store at 2-8°C for short-term use (1-2 weeks).

    • For long-term storage, maintain at -20°C in a non-frost-free freezer .

    • Avoid repeated freeze-thaw cycles by preparing appropriate working aliquots.

  • Advanced Stabilization Techniques:

    • Consider lyophilization with trehalose (5-10%) as a stabilizing agent.

    • Implement controlled atmosphere packaging with nitrogen or argon.

    • Use oxygen-absorbing sachets for storage containers.

  • Quality Control Monitoring:

    • Establish baseline activity measurements using standardized substrates.

    • Implement periodic testing of aliquots to track activity degradation over time.

    • Maintain reference standards for comparative analysis.

  • Chemical Stabilization Approaches:

    • Add 4-hydroxyphenylacetic acid (100 μM) to specifically stabilize HRP activity.

    • Consider polymeric encapsulation techniques for extreme long-term storage.

Stability testing has shown that properly optimized hns Antibody, HRP conjugated preparations can maintain >90% activity for up to 12 months when stored according to these recommendations . For critical long-term projects, consider preparing larger batches with comprehensive quality control testing to ensure consistency throughout the project duration.

How can recent advances in antibody engineering and conjugation chemistry improve the performance of next-generation hns Antibody, HRP conjugated products?

Recent advances in antibody engineering and conjugation chemistry offer significant potential for enhancing hns Antibody, HRP conjugated performance:

  • Site-Specific Conjugation Technologies:

    • CRISPR-Cas9 engineering of antibody genes to introduce specific conjugation sites away from antigen-binding regions.

    • Incorporation of non-canonical amino acids with bioorthogonal reactive groups for precise HRP attachment.

    • These approaches can increase conjugation consistency while preserving antigen binding affinity.

  • Recombinant Antibody Fragments:

    • Development of single-chain variable fragments (scFvs) or antigen-binding fragments (Fabs) against hns.

    • Smaller fragments improve tissue penetration and reduce non-specific binding.

    • Site-specific conjugation of HRP to these fragments provides consistent E/A ratios.

  • Enhanced HRP Variants:

    • Engineered HRP enzymes with improved stability and catalytic efficiency.

    • Directed evolution approaches yielding HRP variants with 2-5 fold higher activity.

    • HRP mutants resistant to inhibition by common buffer components.

  • Novel Linker Technologies:

    • Development of cleavable linkers for controlled release applications.

    • pH-responsive linkers that enhance antibody performance in acidic bacterial microenvironments.

    • Hydrophilic PEG-based linkers that improve conjugate solubility and stability.

  • Antibody Mimetics:

    • Recombinant secondary antibody mimics that can bind to the Fc regions of primary antibodies and carry multiple HRP molecules.

    • These mimetics can deliver approximately 3 HRP molecules per bound antibody, enhancing signal generation .

These technological advances point toward a future where hns Antibody, HRP conjugated products will offer greater consistency, enhanced sensitivity, and improved experimental reproducibility . As these technologies mature and become more accessible, researchers can anticipate significantly improved detection capabilities for challenging hns protein applications.

What are the potential applications of hns Antibody, HRP conjugated in investigating bacterial stress responses and antibiotic resistance mechanisms?

The application of hns Antibody, HRP conjugated in bacterial stress response and antibiotic resistance research presents significant opportunities:

  • Monitoring Stress-Induced hns Regulation:

    • Quantitative assessment of hns protein levels under various antibiotic treatments.

    • Correlation of hns expression with activation of stress response pathways.

    • Time-course analyses revealing dynamics of hns involvement in adaptive responses.

  • Chromatin Reorganization Studies:

    • Investigation of hns-mediated DNA compaction during stress conditions.

    • Analysis of competitive binding between hns and stress-specific nucleoid proteins.

    • Correlation of nucleoid structural changes with transcriptional reprogramming.

  • Biofilm Formation Analysis:

    • Examination of hns distribution in biofilm structures at different developmental stages.

    • Correlation of hns levels with antibiotic penetration and efficacy in biofilms.

    • Investigation of hns-mediated gene silencing in persister cell formation.

  • Horizontal Gene Transfer Regulation:

    • Study of hns binding to foreign DNA elements during antibiotic exposure.

    • Analysis of hns-mediated silencing of resistance gene expression.

    • Investigation of competitive interactions between hns and antibiotic-responsive transcription factors.

  • Genome-Wide Binding Studies:

    • ChIP-seq analysis using hns Antibody, HRP conjugated to map genome-wide binding profiles under antibiotic stress.

    • Identification of antibiotic-specific binding patterns correlating with resistance mechanisms.

    • Integration with transcriptomic data to establish functional consequences of binding.

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.