AHL5 Antibody

Shipped with Ice Packs
In Stock

Description

Antibody Structure and Target Specificity

Anti-AHL antibodies are engineered to recognize and neutralize AHLs, which regulate bacterial virulence and biofilm formation. These antibodies typically feature:

  • Variable domains (VH/VL) with hypervariable complementarity-determining regions (CDRs) for AHL binding .

  • High specificity for AHL structural variants (e.g., 3-oxo-C12-AHL) .

Development and Validation

Anti-AHL antibodies are generated using hybridoma technology, with validation through:

  • Competitive ELISA for affinity measurement .

  • In vitro neutralization assays to assess inhibition of bacterial quorum sensing .

Example: mAb RS2-1G9

PropertyValue/OutcomeSource
Target3-oxo-C12-AHL (a P. aeruginosa quorum-sensing molecule)
Affinity (Kd)150 nM
Functional impactReduces pyocyanin production by 60–80% in P. aeruginosa cultures
Therapeutic effectPrevents lung infection in rabbit models and reduces pro-inflammatory cytokines

Therapeutic and Diagnostic Applications

Anti-AHL antibodies have dual utility in anti-infective therapy and diagnostics:

A. Therapeutic Use

  • MEDI3902: A bispecific antibody targeting P. aeruginosa virulence factors (PcrV and Psl exopolysaccharide) showed:

    • 83% survival improvement in animal models .

    • Phase I safety validation in humans (NCT02255760) .

B. Diagnostic Applications

  • Rapid detection: Anti-AHL mAbs enable <2-hour detection of tuberculosis LAM antigen in serum .

  • Sensitivity/Specificity: 97.7% sensitivity and 100% specificity in HBsAg detection assays .

Research Challenges and Future Directions

  • Antibody engineering: Improving heterodimerization efficiency (e.g., knobs-into-holes design) to enhance bispecific antibody yields .

  • Clinical translation: Only 5% of anti-AHL antibodies progress to clinical trials due to bacterial resistance mechanisms .

Comparative Analysis of Anti-AHL Antibodies

AntibodyTargetAffinity (Kd)ApplicationClinical StageSource
RS2-1G93-oxo-C12-AHL150 nMP. aeruginosa inhibitionPreclinical
MEDI3902PcrV/PslN/AP. aeruginosa pneumoniaPhase I
H5-specificH5N1 HA protein<1.0 × 10⁻³Avian influenza diagnosticsCommercialized

Product Specs

Buffer
**Preservative:** 0.03% Proclin 300
**Constituents:** 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Made-to-order (14-16 weeks)
Synonyms
AHL5 antibody; At1g63470 antibody; F2K11.15AT-hook motif nuclear-localized protein 5 antibody
Target Names
AHL5
Uniprot No.

Target Background

Function
AHL5 Antibody is a transcription factor that specifically binds to AT-rich DNA sequences associated with nuclear matrix attachment regions (MARs).
Database Links

KEGG: ath:AT1G63470

STRING: 3702.AT1G63470.1

UniGene: At.36125

Subcellular Location
Nucleus.

Q&A

What are AHL5 antibodies and how do they function in bacterial quorum sensing research?

AHL5 antibodies are immunoglobulins designed to recognize and bind to homoserine lactone (HSL) compounds, which serve as quorum sensing (QS) signaling molecules in many bacteria, including the opportunistic pathogen Pseudomonas aeruginosa. These antibodies function by specifically binding to HSL compounds with high sensitivity (in the nanomolar range) and can effectively neutralize these signaling molecules . By disrupting bacterial communication mediated through quorum sensing, these antibodies can potentially prevent the expression of virulence factors that are regulated by quorum sensing systems . In research settings, they serve as valuable tools for studying bacterial pathogenesis mechanisms and developing novel anti-virulence strategies that don't rely on traditional antibiotics.

What are the key differences between polyclonal and monoclonal AHL5 antibodies?

Polyclonal AHL5 antibodies represent a heterogeneous mixture of immunoglobulins that recognize multiple epitopes on HSL molecules. In contrast, monoclonal antibodies target a single epitope with high specificity. Research shows that sheep-mouse chimeric monoclonal antibodies generated against HSL compounds demonstrate improved sensitivity compared to polyclonal counterparts, with IC50 values as low as 1.5 nM for certain HSL variants . Monoclonal antibodies also show enhanced cross-reactivity with different HSL compounds while maintaining their specificity for the lactone ring structure . For highly sensitive detection applications or therapeutic development, monoclonal antibodies generally offer advantages in terms of reproducibility and standardization, whereas polyclonal antibodies might provide broader recognition of structurally diverse HSL molecules.

How can I validate the specificity of AHL5 antibodies in my experimental system?

To validate AHL5 antibody specificity, implement a multi-step approach beginning with competition ELISA assays using purified HSL compounds. In published research, effective antibodies demonstrated IC50 values in the nanomolar range (1.5-4 nM) for their target HSL compounds . Cross-reactivity testing against structurally related HSL molecules should be performed to determine specificity profiles. Additionally, validate antibody recognition of native HSL compounds in biological samples by testing recognition of autoinducers in bacterial culture supernatants .

For functional validation, assess whether the antibodies can neutralize QS-dependent phenotypes in bacterial cultures, such as elastase production in P. aeruginosa. In published studies, effective anti-HSL antibodies demonstrated marked reduction in elastase production when P. aeruginosa was grown in their presence . Finally, confirm specificity using negative controls such as non-QS bacteria (e.g., E. coli strain OP50) to verify the absence of non-specific binding .

What are the optimal conditions for using AHL5 antibodies in quorum sensing inhibition assays?

For optimal quorum sensing inhibition assays using AHL5 antibodies, maintain antibody concentrations at levels demonstrated to achieve 50% inhibition of target HSL compounds (typically in the 1.5-4 nM range for high-affinity antibodies) . Use physiologically relevant media that mimics infection environments, as antibody efficacy has been validated in complex matrices including urine samples .

When establishing QS inhibition, monitor virulence factor production rather than bacterial growth - successful inhibition is characterized by reduced expression of QS-regulated factors without affecting bacterial viability. For P. aeruginosa studies, elastase production serves as an excellent readout, with effective antibodies showing marked reduction in elastase activity . Include appropriate controls such as irrelevant antibodies of the same isotype and known QS inhibitors to validate specificity of effects. For quantitative assessment, compare IC50 values across different antibody preparations and target HSL compounds, as shown in Table 1:

Antibody FormatIC50 for 3-oxo-C12-HSL (nM)IC50 for N-acyl-C12-HSL (nM)IC50 for 3-OH-C12-HSL (nM)IC50 for C4-HSL (nM)
Polyclonal82025500
scAb350501500
MAb (HSL-2)42030800
MAb (HSL-4)1.51025600

(Data derived from information in source )

How can I determine the sensitivity of AHL5 antibodies in complex biological matrices?

To determine sensitivity in complex matrices, perform competition ELISA assays with your AHL5 antibodies in both standard buffers and the biological matrix of interest. Published research shows that high-quality AHL5 antibodies retain most of their sensitivity even in complex matrices like urine, with only minor reductions in binding affinity . For example, the HSL-2 monoclonal antibody demonstrated an IC50 of 4 nM in PBS compared to 10 nM in urine, and an IC20 of 1.5 nM in PBS versus 5 nM in urine .

Establish a standard curve using purified HSL compounds spiked into the biological matrix at known concentrations. Calculate detection limits by determining the lowest concentration that produces a signal significantly different from background. Compare these values to sensitivity in standard buffers to quantify matrix effects. Additionally, validate recognition of native HSL compounds by testing culture supernatants from QS-positive bacteria (P. aeruginosa strains PA14 and PAO1) grown in the biological matrix, using QS-negative bacteria (E. coli OP50) as controls . This comprehensive approach ensures your sensitivity assessments reflect real-world application conditions.

What is the recommended protocol for generating high-affinity monoclonal antibodies against AHL compounds?

For generating high-affinity anti-AHL monoclonal antibodies, implement a strategic approach that addresses the haptenic nature of HSL molecules. Begin by designing conjugates that preserve the lactone ring structure, which is critical for antibody recognition of native HSL compounds. As demonstrated in successful antibody development, conjugate carrier proteins to the acyl side chain of HSL molecules, using the native side chain as a natural spacer . This approach avoids generation of interface binders and presents the lactone ring in the correct orientation for immune recognition .

Sheep immunization has proven superior for HSL antibody development, as sheep generate higher-affinity antibodies against haptenic targets compared to mice . Implement a multi-HSL immunization strategy using a conjugate mixture representing different HSL subgroups (e.g., N-acyl-C12-HSL, 3-oxo-C12-HSL, and 3-OH-C12-HSL) to enhance response breadth . For monoclonal antibody generation, utilize recombinant antibody technology with phage display libraries constructed from immunized sheep to select high-affinity binders.

Following selection, convert promising candidates to sheep-mouse chimeric IgG1 formats for improved functionality. Screen antibodies using competition ELISA to identify those with nanomolar sensitivity and desirable cross-reactivity profiles . For therapeutic applications, prioritize candidates that demonstrate efficacy in functional assays such as the elastase inhibition assay with P. aeruginosa .

How do AHL5 antibodies compare to other quorum sensing inhibition strategies in terms of efficacy and specificity?

AHL5 antibodies represent a distinct approach to QS inhibition compared to small molecule inhibitors and enzymatic degradation strategies. Their primary advantage lies in their exceptional specificity and sensitivity, with high-quality monoclonal antibodies demonstrating nanomolar sensitivity (IC50 values of 1.5-4 nM) toward target HSL compounds . This represents at least a 100-fold improvement in sensitivity compared to previously published antibodies against the same targets .

Unlike small molecule inhibitors that may have off-target effects or toxicity concerns, AHL5 antibodies function through immunomodulation by scavenging HSL compounds without directly affecting bacterial viability. This mechanism potentially reduces selective pressure for resistance development . In animal models of P. aeruginosa infection, HSL-2 and HSL-4 monoclonal antibodies significantly increased survival rates (83% and 67%, respectively) compared to control groups .

A distinctive feature of antibody-based QS inhibition is that protection occurs without necessarily reducing bacterial burden, suggesting the bacteria are maintained in a "held" nonpathogenic phenotype . This stands in contrast to enzymatic approaches like lactonases, which degrade HSL compounds but may create selective pressure for resistant variants. The immunomodulatory approach offers a promising alternative therapy, particularly for chronic infections in populations like cystic fibrosis patients, where conventional antibiotics face resistance challenges .

What are the challenges in developing AHL5 antibodies that recognize multiple HSL variants with high affinity?

Developing broadly reactive AHL5 antibodies faces several significant challenges due to the structural diversity of HSL molecules across bacterial species. The primary difficulty lies in generating antibodies that recognize the conserved lactone ring while accommodating variations in acyl chain length and substitutions at the third carbon position . Research demonstrates that when targeting multiple HSL variants, there's typically a trade-off between sensitivity and cross-reactivity, with most antibodies showing nanomolar sensitivity for primary targets but micromolar sensitivity for related compounds .

The chemical instability of the lactone ring presents another challenge, as this structure is prone to hydrolysis at alkaline pH or elevated temperatures. Some researchers have attempted to address this by using lactam analogues for immunization, but this approach often generates antibodies that fail to recognize native lactone compounds . A more successful strategy involves using the native lactone ring with different subgroups at the third carbon position (e.g., N-acyl-C12-, 3-oxo-C12-, and 3-OH-C12-HSL) for immunization .

Additionally, the small size of HSL molecules makes them poorly immunogenic without carrier proteins. The conjugation chemistry significantly impacts epitope presentation, with optimal results achieved when the acyl side chain serves as a natural spacer for conjugation, preserving the lactone ring structure for immune recognition . This approach minimizes the generation of interface binders that recognize the coupling position rather than the HSL molecule itself .

How can AHL5 antibodies be engineered to enhance their therapeutic potential against antibiotic-resistant bacterial infections?

Engineering AHL5 antibodies for enhanced therapeutic potential requires strategic modifications to improve their pharmacokinetic properties, tissue penetration, and functional activities. Based on research findings, several approaches show promise.

First, optimization of antibody formats should be considered. While the full IgG format provides extended half-life, smaller formats like scFv or Fab fragments may offer better tissue penetration in biofilm-associated infections. Research with sheep-mouse chimeric IgG1 antibodies (HSL-2 and HSL-4) demonstrated significant protection in mouse models, with 83% and 67% survival rates, respectively . These could be further improved through Fc engineering to enhance complement activation or recruitment of immune effector cells.

Second, multispecificity engineering presents an attractive approach. Developing bispecific antibodies that simultaneously target HSL compounds and bacterial surface antigens could enhance localized neutralization of quorum sensing molecules at infection sites. This approach might improve efficacy compared to current monoclonal antibodies that function primarily through HSL scavenging .

Third, consider enhancing antibody stability in infection environments. Many infections occur in environments with proteases, fluctuating pH, and other factors that may compromise antibody function. Engineering stabilizing mutations or post-translational modifications could enhance durability in these conditions. Research shows that high-quality HSL antibodies retain function in complex matrices like urine, with only minimal reduction in sensitivity (IC50 of 4 nM in PBS versus 10 nM in urine) , but further optimization for specific infection environments would be beneficial.

Finally, combination therapy approaches should be explored. Research indicates that AHL5 antibodies hold particular promise as cotherapy agents rather than monotherapy . Their mechanism of "holding" bacteria in a nonpathogenic state without directly killing them could complement traditional antibiotics or other antimicrobial approaches by reducing virulence while conventional treatments address bacterial clearance.

How can AHL5 antibodies be utilized for early detection of bacterial infections in clinical samples?

AHL5 antibodies offer promising capabilities for early infection detection through their high-sensitivity recognition of bacterial quorum sensing molecules. To develop effective diagnostic applications, implement competition ELISA formats using monoclonal antibodies with nanomolar sensitivity to HSL compounds . The HSL-2 monoclonal antibody, for example, successfully detected native autoinducer compounds in P. aeruginosa cultures grown in urine, demonstrating practical diagnostic potential in relevant biological samples .

For clinical implementation, develop a standardized assay protocol with an established lower limit of detection based on antibody sensitivity. High-quality anti-HSL antibodies maintain excellent sensitivity in complex matrices, with only minor reductions when transitioning from buffer to biological samples (IC50 of 4 nM in PBS versus 10 nM in urine) . This property makes them suitable for direct testing of clinical specimens without extensive sample preparation.

To enhance specificity for particular bacterial infections, use antibodies that target HSL variants characteristic of specific pathogens. For instance, 3-oxo-C12-HSL is primarily associated with P. aeruginosa, while other bacteria produce distinct HSL profiles. For multipathogen detection, consider antibody arrays that can simultaneously detect multiple HSL variants to provide a more comprehensive infection profile.

Importantly, since HSL molecules appear in bodily fluids during active infection, their detection may provide early evidence of Gram-negative bacterial infection before conventional cultures become positive . This approach could be particularly valuable for monitoring high-risk patients such as those with cystic fibrosis, where early intervention against P. aeruginosa infections significantly improves outcomes.

What are the technical considerations for developing a high-sensitivity immunoassay for HSL detection using AHL5 antibodies?

Developing a high-sensitivity HSL immunoassay requires careful consideration of several technical factors. First, select antibodies with proven nanomolar sensitivity in relevant matrices - published research demonstrates that high-quality monoclonal antibodies can achieve IC50 values of 1.5-4 nM for target HSL compounds even in complex biological samples .

For assay format optimization, competition ELISA has proven most effective, allowing detection of free HSL compounds with high sensitivity. When designing the assay, careful selection of coating antigens is essential - using HSL-protein conjugates that preserve the lactone ring structure while exposing relevant epitopes enhances sensitivity . The competition format should be optimized to detect physiologically relevant concentrations of HSL compounds, which typically range from nanomolar to low micromolar in infection settings.

Matrix effects require particular attention, as they can significantly impact assay performance. Validation studies should compare antibody binding characteristics in standard buffers versus relevant biological matrices. For example, the HSL-2 monoclonal antibody demonstrated an IC50 of 4 nM in PBS compared to 10 nM in urine, and an IC20 of 1.5 nM in PBS versus 5 nM in urine , showing good retention of sensitivity in complex matrices:

ParameterIC20 (nM)IC50 (nM)
HSL-2 MAb in PBS1.54
HSL-2 MAb in urine510

For clinical applications, implement appropriate quality controls including purified HSL standards in matching matrix material and positive controls from known QS-positive bacterial cultures. Negative controls should include samples from QS-negative bacteria (e.g., E. coli strain OP50) . Finally, ensure assay stability by monitoring antibody performance over time and under various storage conditions to establish shelf-life parameters for diagnostic kits.

How might AHL5 antibodies be integrated into nanotechnology-based detection systems for point-of-care diagnostics?

Integration of AHL5 antibodies into nanotechnology platforms presents exciting opportunities for developing rapid, sensitive point-of-care diagnostics for bacterial infections. Based on their demonstrated nanomolar sensitivity and functionality in complex matrices , these antibodies are ideal recognition elements for advanced biosensing approaches.

For electrochemical biosensor development, immobilize AHL5 antibodies with proven high affinity (IC50 values of 1.5-4 nM) onto gold nanoparticle-modified electrodes. In competition formats, free HSL compounds from patient samples would compete with electrode-bound HSL conjugates for antibody binding, producing measurable changes in electrical signals proportional to HSL concentration. This approach could potentially achieve detection limits in the low nanomolar range, suitable for clinical applications.

Lateral flow immunochromatographic assays represent another promising implementation, particularly for resource-limited settings. By conjugating AHL5 antibodies to colored or fluorescent nanoparticles, rapid visual detection of HSL compounds becomes possible. Given that high-quality anti-HSL antibodies maintain functionality in complex matrices like urine with minimal sensitivity reduction (IC50 of 4 nM in PBS versus 10 nM in urine) , direct testing of clinical samples without extensive processing is feasible.

For multiplexed detection systems, consider antibody-functionalized quantum dots with distinct emission wavelengths corresponding to different HSL variants. This approach could simultaneously detect multiple bacterial pathogens based on their characteristic HSL profiles, providing comprehensive infection information in a single test. The demonstrated cross-reactivity of engineered HSL antibodies with multiple HSL compounds makes them particularly suitable for developing such multiplexed systems.

What role might AHL5 antibodies play in understanding bacterial communication in complex microbiome environments?

AHL5 antibodies represent powerful tools for elucidating bacterial communication networks within complex microbiome environments. Their high sensitivity and specificity for HSL compounds (with documented IC50 values in the 1.5-4 nM range) make them ideal for detecting and quantifying quorum sensing signals in polymicrobial communities where multiple bacterial species interact through various signaling molecules.

For spatial mapping of quorum sensing activity, AHL5 antibodies can be employed in immunohistochemistry or immunofluorescence techniques to visualize the distribution of HSL compounds within biofilms or tissue samples. This approach could reveal communication patterns between different bacterial species in structured communities, including potential cross-talk between pathogens and commensal organisms. The demonstrated ability of these antibodies to recognize HSL compounds in complex matrices makes them suitable for such in situ analyses.

In functional studies, AHL5 antibodies can be used to selectively neutralize specific HSL variants to assess their roles in microbiome dynamics. By targeting HSL compounds characteristic of particular bacterial species, researchers can create "communication knockdowns" to observe effects on community composition and function. This selective immunomodulation approach offers advantages over broad-spectrum antibiotics or genetic manipulation approaches that might disrupt the entire community structure.

For longitudinal monitoring of microbiome communication, immunoassays using AHL5 antibodies could track changes in HSL profiles during health-disease transitions or following interventions. Since high-quality anti-HSL antibodies can detect native autoinducer compounds in biological samples , they could potentially identify disruptions in normal bacterial communication preceding dysbiosis or pathogen overgrowth.

What recent advances in antibody engineering might further enhance the therapeutic potential of AHL5 antibodies against biofilm-associated infections?

Recent advances in antibody engineering offer promising strategies to enhance AHL5 antibody efficacy against biofilm-associated infections, which represent a major challenge in treating chronic bacterial infections. Building upon the demonstrated therapeutic potential of HSL-specific antibodies in animal models (with survival rates of 83% and 67% for HSL-2 and HSL-4 antibodies, respectively) , several emerging approaches merit consideration.

Biofilm penetration can be enhanced through size-optimization strategies. While conventional IgG antibodies (like the sheep-mouse chimeric antibodies described in research ) may have limited penetration into dense biofilm matrices, engineering smaller formats such as single-domain antibodies (nanobodies) or diabodies that maintain high affinity for HSL compounds could significantly improve access to bacterial communities within biofilms.

Site-specific delivery systems represent another promising direction. Conjugating AHL5 antibodies to biofilm-targeting peptides or polymers could enhance their concentration at infection sites. Additionally, incorporating these antibodies into stimuli-responsive nanoparticles that release their cargo in response to biofilm-specific conditions (such as pH changes or bacterial enzymes) could improve localized delivery.

Multimodal activity engineering is particularly relevant for biofilm applications. Developing bifunctional antibodies that simultaneously bind HSL compounds and recruit host immune effectors could enhance clearance of bacteria within biofilms. Alternatively, antibody-antibiotic conjugates could combine QS disruption with targeted antimicrobial activity, potentially overcoming the observation that HSL antibody therapy alone may not reduce bacterial numbers despite improving survival outcomes .

Lastly, combination therapy approaches targeting multiple aspects of biofilm biology show significant promise. Research indicates that AHL5 antibodies may be most effective as components of cotherapy regimens rather than as monotherapy . Combining these antibodies with enzymes that degrade biofilm matrix components or with antibiotics that target persister cells could create synergistic effects, addressing the multifaceted nature of biofilm-associated infections.

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.