Metallo-beta-lactamase type 2 Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to Metallo-β-Lactamase Type 2 (MBL-B2) and Antibody Development

Metallo-β-lactamases (MBLs) are zinc-dependent enzymes that hydrolyze β-lactam antibiotics, including carbapenems, rendering them ineffective. MBLs are classified into three subclasses: B1, B2, and B3, based on structural and functional differences. MBL-B2 is a distinct subclass characterized by a narrow substrate specificity, primarily targeting carbapenems, and is often chromosomally encoded in Aeromonas species .

Metallo-β-lactamase type 2 antibodies refer to immunoglobulins designed to neutralize MBL-B2 enzymes. While antibodies targeting MBL-B1 (e.g., NDM, VIM) and MBL-B3 (e.g., L1) have been studied, no peer-reviewed research explicitly describes antibodies specific to MBL-B2 in the provided sources. This gap highlights challenges in addressing subclass-specific resistance mechanisms.

Mechanistic Context of MBL-B2 and Antibody Targeting

MBL-B2 enzymes share a unique zinc-binding site configuration:

  • Zinc coordination: A single zinc ion (Zn²⁺) at the Zn1 site, with altered residues (e.g., N116, H118, or H196) compared to B1/B3 subclasses .

  • Substrate hydrolysis: Limited to carbapenems, unlike B1/B3 enzymes, which hydrolyze most β-lactams .

Antibodies targeting MBL-B2 would need to:

  • Bind active-site zinc ions or critical residues (e.g., His, Cys) to block hydrolysis.

  • Overcome structural diversity between MBL-B2 and other subclasses.

Current Antibody Research on MBLs: Focus on B1 and B3

While MBL-B2-specific antibodies are not documented, studies on B1/B3-targeting antibodies provide insights into potential strategies:

Antibody TypeTarget MBLIC₅₀ (μM)Key FindingsSource
Polyclonal camel IgGNDM-1 (B1)0.036–0.1Inhibition via binding to Zn²⁺-dependent active site; no activity against B3 (L1) .
Monoclonal rabbit IgGVIM-1 (B1)0.04Synergy with β-lactams in in vitro assays; no inhibition of B3 (L1) .
Non-β-lactam inhibitorsNDM-1 (B1)24.8–97.4Zinc chelation via phosphonate compounds; restored carbapenem efficacy in vivo .

Key observations:

  • Cross-reactivity challenges: Polyclonal antibodies against B1/B3 MBLs show limited activity across subclasses (e.g., anti-NDM-1 IgG fails to inhibit L1) .

  • Zinc chelation: Small-molecule inhibitors (e.g., PMPC derivatives) disrupt Zn²⁺ coordination, but antibody-based approaches require precise epitope targeting .

Challenges in Developing MBL-B2-Specific Antibodies

  1. Structural diversity: MBL-B2 lacks the conserved cysteine residues present in B1/B3, complicating epitope identification .

  2. Chromosomal localization: MBL-B2 is often chromosomally encoded, limiting horizontal gene transfer and clinical priority compared to plasmid-borne B1 enzymes .

  3. Therapeutic urgency: B1/B3 MBLs (e.g., NDM, VIM) dominate clinical outbreaks, redirecting research focus .

Potential Strategies for MBL-B2 Antibody Development

  1. Epitope mapping: Use crystallography to identify conserved regions in MBL-B2 (e.g., Zn1-binding residues).

  2. Synergistic approaches: Combine antibodies with β-lactamase inhibitors (e.g., DBOs like zidebactam) to enhance carbapenem efficacy .

  3. Animal models: Test antibody efficacy in Aeromonas-infected models to validate in vivo activity.

Product Specs

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

This polyclonal antibody targeting Metallo-beta-lactamase type 2 (MBL-2) is produced in rabbits immunized with recombinant Serratia marcescens MBL-2 protein (amino acids 19-246). Antibody purification is achieved via protein G affinity chromatography. This purified MBL-2 antibody is suitable for ELISA and Western blotting (WB) applications, specifically for the detection of Serratia marcescens MBL-2 protein.

Serratia marcescens produces MBL-2 as a defense mechanism against beta-lactam antibiotics. This enzyme hydrolyzes the beta-lactam ring, rendering these antibiotics ineffective and contributing to bacterial resistance.

Form
Liquid
Lead Time
Orders typically ship within 1-3 business days of receipt. Delivery times may vary depending on the shipping method and destination. Please contact your local distributor for precise delivery estimates.
Synonyms
Metallo-beta-lactamase type 2 (EC 3.5.2.6) (B2 metallo-beta-lactamase) (BLA-IMP) (IMP-1) (Beta-lactamase type II) (Metallo-beta-lactamase type II)
Target Names
N/A
Uniprot No.

Target Background

Function

MBL-2 confers resistance to various beta-lactam antibiotics (penicillins, cephalosporins, and carbapenems) through the hydrolysis of the beta-lactam ring.

Database Links

KEGG: ag:AAB30289

Protein Families
Metallo-beta-lactamase superfamily, Class-B beta-lactamase family
Subcellular Location
Periplasm.

Q&A

What are metallo-beta-lactamases and why are they clinically significant?

Metallo-beta-lactamases (MBLs) are a class of enzymes that hydrolyze β-lactam antibiotics, including carbapenems, which are often considered last-resort treatments. MBLs require zinc ions for their catalytic activity, distinguishing them from serine β-lactamases. Their clinical significance stems from their ability to confer broad-spectrum resistance to multiple β-lactam antibiotics, severely limiting treatment options .

Unlike serine β-lactamases, which can be inhibited by commercially available inhibitors like clavulanic acid, MBLs have no clinically approved inhibitors, making infections caused by MBL-producing bacteria particularly difficult to treat . The widespread dissemination of these enzymes, particularly in Pseudomonas aeruginosa, Acinetobacter species, and Enterobacteriaceae, represents a serious global health threat .

What are the major types of metallo-beta-lactamases identified in clinical isolates?

The major MBL types identified in clinical isolates include:

MBL TypeFirst ReportedCommon Bacterial HostsGeographical Distribution
IMPJapan, 1991P. aeruginosa, EnterobacteriaceaeWorldwide, endemic in Japan
VIMItaly, 1997P. aeruginosa, EnterobacteriaceaeWorldwide, endemic in Mediterranean
SPMBrazil, 1997P. aeruginosaBrazil, limited global spread
GIMGermany, 2002P. aeruginosaLimited, mainly Europe
NDMIndia, 2008Enterobacteriaceae, AcinetobacterWorldwide, endemic in Indian subcontinent

These MBLs are encoded on transferable genetic elements, typically within class 1 integrons that are embedded in transposons, facilitating their rapid dissemination among bacterial populations . The VIM-2 variant (often referred to as metallo-beta-lactamase type 2) is among the most widely distributed MBLs globally .

How do antibodies against metallo-beta-lactamases function in research applications?

Antibodies against metallo-beta-lactamases serve multiple critical functions in research:

  • Detection and Quantification: Antibodies enable specific detection of MBL proteins in bacterial isolates through immunological methods such as ELISA, western blotting, and immunofluorescence microscopy .

  • Purification: Immunoaffinity chromatography using anti-MBL antibodies allows for purification of native MBL enzymes from bacterial lysates.

  • Structural Studies: Antibodies can be used to crystallize MBL-antibody complexes for structural analysis, revealing binding epitopes and potential inhibitor design targets.

  • Diagnostic Development: Antibodies form the foundation for rapid immunological detection methods, including lateral flow assays like the NG-Test CTX-M MULTI, which can detect specific β-lactamases within 15 minutes from cultured isolates with high sensitivity (100%) and specificity (99.6%) .

Researchers typically employ polyclonal antibodies for general detection and monoclonal antibodies when higher specificity is required for distinguishing between closely related MBL variants .

What detection methods are available for metallo-beta-lactamases in clinical and research settings?

Multiple detection methods exist for MBLs, each with distinct advantages and limitations:

Detection ApproachMethodApplicationsTurnaround TimeSensitivity/Specificity
PhenotypicColourimetric assays (Rapid ESBL NP®, Beta Lacta Test)Clinical screening15 min - 2 hours93.9%/98.5%
Disk diffusion with EDTAClinical laboratories18-24 hoursModerate
Carba NP testClinical screening2 hoursHigh for carbapenemases
ImmunologicalLateral flow immunoassaysPoint-of-care testing15 minutes100%/99.6% for specific targets
ELISAResearch settings3-4 hoursHigh
GenotypicPCRTargeted gene detection2-4 hoursVery high for known genes
DNA MicroarraysMultiple resistance gene detection6-8 hoursHigh
Whole Genome SequencingComprehensive genetic analysis12-48 hoursHighest

Phenotypic methods detect the enzymatic activity of MBLs but cannot distinguish between different MBL types, while genotypic methods provide specific identification of MBL genes but may miss novel variants . Immunological methods, particularly those using antibodies against specific MBL types, offer a balance of specificity, sensitivity, and rapid turnaround time .

How do structural differences between metallo-beta-lactamase variants impact antibody development and specificity?

The structural diversity among MBL variants presents significant challenges for antibody development. MBLs are classified into subclasses B1, B2, and B3 based on their sequence homology and zinc coordination mechanisms. Each subclass exhibits distinct structural features that influence antibody binding:

B1 enzymes (including IMP, VIM, and NDM) contain two zinc ions in their active site and display a characteristic αβ/βα sandwich fold. Their surface-exposed epitopes often include highly variable loop regions that dictate substrate specificity but challenge cross-reactive antibody development . VIM-2 specifically contains unique loop structures near its active site that distinguish it from other B1 enzymes.

Antibody development strategies must account for:

  • Conservation of epitopes across variant types

  • Accessibility of epitopes in the folded protein

  • Potential cross-reactivity with human metallo-β-lactamase domain-containing proteins like LACTB2

Research has demonstrated that antibodies targeting conserved regions of the MBL fold can achieve broader recognition across variants, while those targeting variable loops provide higher specificity for individual types. The crystal structure data of MBLs like LACTB2 shows almost perfect alignment of the MBL domain with other ribonucleases of the MBL superfamily, highlighting regions where cross-reactivity must be carefully evaluated .

What are the methodological considerations for validating antibody specificity against different metallo-beta-lactamase variants?

Validating antibody specificity for MBL variants requires a multi-faceted approach:

1. Cross-reactivity Panel Testing
Antibodies must be tested against a comprehensive panel of:

  • Target MBL variants (e.g., VIM-1, VIM-2, VIM-4)

  • Related MBL types (IMP, NDM, SPM)

  • Serine β-lactamases (KPC, CTX-M)

  • Human MBL-domain proteins (LACTB2)

2. Epitope Mapping
Techniques to determine precise epitope recognition include:

  • Hydrogen-deuterium exchange mass spectrometry

  • Peptide array analysis

  • X-ray crystallography of antibody-antigen complexes

  • Alanine scanning mutagenesis

3. Activity Inhibition Assessment
Determining whether antibody binding affects enzymatic function:

  • Nitrocefin hydrolysis assays with/without antibody

  • Carbapenem degradation kinetics analysis

  • Zinc chelation effects on antibody binding

4. Performance in Complex Matrices
Evaluating antibody performance in:

  • Bacterial lysates from various species

  • Clinical specimens (blood, urine)

  • Mixed bacterial populations

Research indicates that antibodies targeting the active site loops may exhibit higher specificity for distinguishing between closely related variants like VIM-1 and VIM-2, but may have reduced sensitivity compared to antibodies targeting conserved structural elements .

How can researchers optimize immunoassays to differentiate between metallo-beta-lactamases and serine beta-lactamases?

Optimizing immunoassays to differentiate between MBLs and serine β-lactamases requires strategic design considerations:

1. Differential Inhibition Controls
Incorporation of parallel sample processing with:

  • EDTA (chelates zinc, inhibits MBLs)

  • Avibactam (inhibits serine β-lactamases)

  • Combined inhibitors

2. Dual-Epitope Detection Systems
Development of sandwich ELISA or lateral flow assays utilizing:

  • Capture antibody targeting conserved MBL epitope

  • Detection antibody targeting type-specific region

  • Comparative analysis against serine β-lactamase standards

3. Assay Condition Optimization
Manipulation of buffer components to enhance specificity:

  • Zinc concentration (MBLs are zinc-dependent)

  • pH optimization (differential pH stability between enzyme classes)

  • Detergent selection for membrane preparation

4. Statistical Validation Approaches
Implementation of:

  • Receiver Operating Characteristic (ROC) analysis for cutoff determination

  • Multilevel likelihood ratio analysis

  • Bayesian modeling for predictive value assessment

Recent developments in rapid colourimetric methods demonstrate that optimization of reaction conditions can achieve excellent discrimination between enzyme classes, with assays like Rapid ESBL NP® showing 93.9% sensitivity and 98.5% specificity . For highest accuracy, many researchers employ a combinatorial approach using both inhibitor-based phenotypic tests and antibody-based detection methods .

What are the challenges in developing antibodies against emerging metallo-beta-lactamase variants?

Developing antibodies against emerging MBL variants presents several significant challenges:

Evolutionary Dynamics and Variant Diversity

  • Rapid mutational evolution generates variants with altered surface epitopes

  • Horizontal gene transfer creates chimeric enzymes with mixed epitope profiles

  • Single nucleotide polymorphisms can significantly impact antibody binding affinity

Accessibility Limitations

  • Reference materials for new variants are often limited

  • Delayed identification of novel variants in clinical settings

  • Restricted sharing of bacterial isolates across international boundaries

Cross-Reactivity Management

  • Homology between MBL variants and human MBL-domain proteins like LACTB2

  • Potential cross-reactivity with environmental MBLs of non-clinical importance

  • Distinguishing between closely related variants (e.g., VIM-2 vs. VIM-4)

Validation Complexities

  • Lack of standardized validation panels for emerging variants

  • Limited clinical isolates for performance testing

  • Difficulty establishing true negative controls

Research approaches to address these challenges include structural biology techniques to identify conserved epitopes across variant evolution, computational epitope prediction algorithms, and phage display screening against multiple variant panels simultaneously . The successful development of broadly reactive antibodies requires careful epitope selection informed by both sequence and structural analysis of the target enzyme family .

How can researchers integrate antibody-based detection with phenotypic assays for comprehensive MBL characterization?

Integration of antibody-based detection with phenotypic assays provides powerful complementary approaches for MBL characterization:

Combined Workflow Design:

  • Initial Screening Phase

    • Rapid phenotypic colourimetric assay (e.g., Carba NP test) for β-lactamase activity detection

    • Parallel EDTA inhibition testing to identify potential MBLs

    • Antibody-based lateral flow immunoassay for preliminary MBL type identification

  • Confirmation Phase

    • ELISA with type-specific antibodies for quantification and classification

    • PCR verification of corresponding MBL genes

    • Nitrocefin hydrolysis kinetics with recombinant standards

  • Characterization Phase

    • Antibody-based protein purification via immunoprecipitation

    • Enzyme kinetics determination with purified enzyme

    • Inhibitor screening against isolated enzyme

This integrated approach overcomes limitations of individual methods; phenotypic tests detect novel variants regardless of sequence, while antibody tests provide rapid type specification. Research demonstrates that such integrated workflows can reduce characterization time from days to hours while providing more comprehensive resistance profiles .

A data analysis framework incorporating results from multiple methods can be implemented:

MethodData OutputIntegration ValueLimitations
Antibody lateral flowMBL type identificationRapid typing (15 min)Limited to known variants
Colourimetric assayEnzyme activity confirmationFunctional verificationCannot distinguish types
PCRGene identificationGenetic confirmationPrimer-dependent
EDTA inhibitionMBL confirmationClass verificationNon-specific for variants

What experimental protocols yield optimal antibody specificity for MBL detection in complex bacterial samples?

Achieving optimal antibody specificity for MBL detection in complex bacterial samples requires carefully developed protocols:

Antibody Selection Criteria

  • Monoclonal antibodies for highest specificity to individual MBL types

  • Validation against panel of recombinant MBL standards

  • Epitope mapping to confirm targeting of distinguishing regions

  • Cross-adsorption against related MBL types for reduced cross-reactivity

Signal Amplification Strategies

  • Biotin-streptavidin systems for enhanced sensitivity

  • Tyramide signal amplification for low-abundance detection

  • Quantum dot conjugates for multiplexed detection

  • Microfluidic concentration for enhanced limit of detection

Validation and Standardization

  • Inclusion of recombinant MBL standards as positive controls

  • Matrix-matched negative controls

  • Statistical determination of signal-to-noise thresholds

  • Multi-laboratory validation of protocol robustness

Research indicates that targeted epitope selection combined with optimized sample preparation can achieve detection limits as low as 10 ng/mL of specific MBL types in complex bacterial lysates, representing approximately 103 CFU/mL in clinical samples .

How can structural data on metallo-beta-lactamases inform epitope selection for antibody development?

Structural data provides critical insights for rational epitope selection in MBL antibody development:

Structure-Based Epitope Mapping Approach

Crystal structures of MBLs reveal distinct regions for antibody targeting:

RegionStructural FeaturesAntibody Development Implications
Active Site LoopsHighly variable, surface exposedType-specific detection, potential enzyme inhibition
Zinc-Binding MotifsConserved HXHXD sequenceBroad MBL detection, zinc-dependent epitopes
β-Sheet CoreConserved structural scaffoldPan-MBL detection, stable epitopes
C-terminal RegionVariable accessibilityType-specific detection if accessible

The crystal structure of human LACTB2 shows almost perfect alignment of the MBL domain with other ribonucleases of the MBL superfamily, highlighting regions where cross-reactivity with human proteins must be avoided .

Molecular Dynamics Simulation for Epitope Accessibility

Molecular dynamics simulations can predict:

  • Epitope flexibility in solution

  • Solvent accessibility under physiological conditions

  • Conformational changes upon substrate binding

  • Potential masking effects in membrane-associated states

Sequence-Structure Correlation Analysis

Bioinformatic approaches combining sequence and structural data:

  • Identification of conserved surface patches across variants

  • Prediction of immunodominant epitopes

  • Assessment of evolutionary conservation to target stable regions

  • Identification of regions unique to bacterial MBLs versus human homologs

Structural Considerations for Diagnostic Applications

  • Target conformational epitopes spanning multiple loops for specificity

  • Select regions distant from the active site for detection without inhibition

  • Identify epitopes maintained across variant evolution

  • Avoid regions involved in protein-protein interactions in vivo

Research on LACTB2 and other MBL family proteins has demonstrated that careful structural analysis can identify unique surface-exposed epitopes that distinguish between closely related MBL variants while avoiding cross-reactivity with human proteins .

What quality control measures are essential for validating antibodies against metallo-beta-lactamases in research applications?

Comprehensive quality control measures are essential for validating antibodies used in MBL research:

Analytical Validation Framework

ParameterMethodologyAcceptance Criteria
SpecificityWestern blot against panel of 10+ MBL variants<5% cross-reactivity with non-target MBLs
SensitivityLimit of detection using purified standardsDetection of ≤10 ng/mL target MBL
PrecisionIntra/inter-assay CV with replicate testingCV ≤15% across assay runs
AccuracyRecovery from spiked samples80-120% recovery of known additions
LinearitySerial dilution analysisR² ≥0.98 across working range
RobustnessPerformance across buffer/temperature variationsConsistent results (±15%) across conditions

Biological Validation Requirements

  • Confirmation of specificity using knockout bacterial strains

  • Comparison with orthogonal detection methods (PCR, enzymatic activity)

  • Performance testing in clinical isolate panels of known MBL status

  • Stability assessment under intended storage conditions

Production Consistency Controls

  • Lot-to-lot comparison of antibody performance

  • Epitope mapping confirmation for monoclonal antibodies

  • Isotype verification and fragmentation assessment

  • Endotoxin testing for research applications

Application-Specific Validation

  • For immunoprecipitation: Pull-down efficiency measurement

  • For ELISA: Matrix effect characterization

  • For immunohistochemistry: Fixation method compatibility

  • For lateral flow: Time-to-result consistency

The most reliable antibody validation incorporates multiple detection formats and comparison with established reference methods. Recent advances in rapid detection methods have demonstrated that properly validated antibodies can achieve sensitivity and specificity exceeding 99% for specific MBL variants in clinical samples .

How might antibodies against metallo-beta-lactamases be incorporated into novel inhibitor discovery pipelines?

Antibodies against MBLs offer innovative approaches to inhibitor discovery:

Epitope-Guided Inhibitor Design

  • Crystal structures of antibody-MBL complexes reveal key binding pockets

  • Computational fragment-based design targeting antibody-defined epitopes

  • Antibody-competitive binding assays for high-throughput screening

  • Development of antibody mimetics as potential therapeutic agents

Antibody-Displacement Screening Platforms

  • Fluorescently labeled antibodies as competition probes

  • Displacement assays to identify small molecules binding to specific epitopes

  • Surface plasmon resonance for real-time binding kinetics

  • Flow cytometry-based screening using bacterial surface display

Structure-Function Relationship Studies

  • Antibodies as crystallization chaperones for MBL structural studies

  • Identification of conformational changes upon inhibitor binding

  • Mapping of allosteric sites for non-competitive inhibition

  • Determination of zinc coordination preferences for chelator optimization

Combinatorial Approaches

  • Bispecific antibodies targeting MBL and bacterial surface markers

  • Antibody-drug conjugates for targeted delivery of inhibitors

  • Synergistic screening of antibody fragments with small molecule libraries

  • Nanobody-based inhibitor scaffold development

The development of effective MBL inhibitors remains an urgent clinical need as highlighted in recent reviews, and antibody-based approaches may provide valuable insights that complement traditional medicinal chemistry strategies . Current clinical trials of compounds like taniborbactam and xeruborbactam (QPX7728) could benefit from structure-guided antibody studies to further optimize inhibitor binding and specificity .

What emerging technologies might enhance the sensitivity and specificity of antibody-based MBL detection?

Several emerging technologies show promise for enhancing antibody-based MBL detection:

Advanced Biosensor Platforms

  • Surface acoustic wave (SAW) biosensors for label-free detection

  • Graphene field-effect transistor (GFET) immunosensors

  • Localized surface plasmon resonance (LSPR) nanoparticle-enhanced detection

  • Electrochemical impedance spectroscopy with antibody-modified electrodes

Digital and Single-Molecule Detection

  • Digital ELISA platforms with single-molecule resolution

  • Droplet microfluidics for absolute quantification

  • Single-molecule pull-down assays for ultra-sensitive detection

  • Super-resolution microscopy for spatial localization of MBLs

Multiplexed Detection Systems

  • Antibody arrays for simultaneous detection of multiple MBL types

  • Spectral flow cytometry with quantum dot-labeled antibodies

  • Mass cytometry (CyTOF) for multi-parameter analysis

  • Next-generation sequencing of antibody-captured targets

Artificial Intelligence Integration

  • Machine learning algorithms for pattern recognition in complex signal data

  • Deep learning for image analysis of immunofluorescence

  • Predictive modeling of antibody-antigen interactions

  • Automated quality control assessment and standardization

These technologies offer potential for detecting MBLs at concentrations well below current clinical thresholds, potentially enabling earlier detection of resistance and better monitoring of treatment efficacy. Recent advances in rapid colourimetric and immunological lateral flow assays demonstrate the clinical value of improved detection methods, with assays now capable of detecting specific β-lactamases within 15 minutes with excellent sensitivity and specificity .

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.