Recombinant Bifidobacterium longum Tyrosine recombinase XerD (xerD)

Shipped with Ice Packs
In Stock

Description

General Information

XerD, along with its partner recombinase XerC, functions in the stable inheritance of the Escherichia coli chromosome and multicopy circular plasmids . These proteins ensure that circular, multimeric DNA molecules are converted to monomers . The organization of the Rin recombinase resembles tyrosine recombinases such as XerD, which contain common core-type DNA-binding domains .

Mechanism of Action

Recombination is initiated when the conserved tyrosine hydroxyl attacks the scissile phosphate, forming a 3′ phosphotyrosyl–DNA complex and a free 5′ hydroxyl . In the second step, a 5′ hydroxyl from the adjacent partner duplex attacks the phosphotyrosine to form a Holliday junction intermediate . The recombination reaction is completed by the exchange of the second pair of strands, using the same mechanism, 6–8 bp away from the site of the initial strand exchanges .

Function in Bacteria

In Escherichia coli and most bacteria, dimers are resolved to monomers by site-specific recombination, a process performed by two Chromosomally Encoded tyrosine Recombinases (XerC and XerD) . XerCD recombinases act at a 28 bp recombination site dif, which is located at the replication terminus region of the chromosome .

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your format preference in order remarks for customized preparation.
Lead Time
Delivery times vary depending on the purchase method and location. Please consult your local distributor for precise delivery estimates.
Note: Standard shipping includes blue ice packs. Dry ice shipping requires advance notification and incurs additional charges.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to consolidate the contents. Reconstitute the protein in sterile, deionized water to a concentration of 0.1-1.0 mg/mL. For long-term storage, we recommend adding 5-50% glycerol (final concentration) and aliquoting at -20°C/-80°C. Our standard glycerol concentration is 50% and can serve as a guideline.
Shelf Life
Shelf life depends on several factors, including storage conditions, buffer components, temperature, and the protein's inherent stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized forms have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Store at -20°C/-80°C upon receipt. Aliquot for multiple uses to prevent repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing.
The tag type is determined during production. Specify your desired tag type for preferential development.
Synonyms
xerD; BL1368; Tyrosine recombinase XerD
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-308
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Bifidobacterium longum (strain NCC 2705)
Target Names
xerD
Target Protein Sequence
MTNGFTRLTE QFLIHIGVER GLATATVTAY ESDIAKYIDW LETRGIHEPD AITKQDVEDY IAALDQAGES ARSKARRLAS IHEFHRFALG QHAVTADVSA AVKAPKGAST LPDVLTVDEV TRLLDAAAVG GSTDPVVLRD KALLEFMYAT GCRVSEATGA NLDDIDLDEH IARLMGKGSK QRLVPLGSYA CRAITAYLNA GRGELEQRSS AKIPERRALF LNKRGKRISR QSVWEIVKAT GERAGITKPL HPHTLRHSFA THLIQGGADV RTVQELLGHA SVTTTQIYTH VSPETLIETY LTSHPRAR
Uniprot No.

Target Background

Function
Recombinant Bifidobacterium longum Tyrosine recombinase XerD (xerD) is a site-specific tyrosine recombinase that catalyzes the cutting and rejoining of DNA molecules. The XerC-XerD complex is crucial for resolving bacterial chromosome dimers into monomers, enabling proper segregation during cell division. It also contributes to the segregational stability of plasmids.
Database Links

KEGG: blo:BL1368

STRING: 206672.BL1368

Protein Families
'phage' integrase family, XerD subfamily
Subcellular Location
Cytoplasm.

Q&A

What is Bifidobacterium longum and why is it suitable for recombinant protein expression?

Bifidobacterium longum is a gram-positive, anaerobic bacterium that stands as one of the most abundant microorganisms in the human intestinal tract. It naturally produces lactic and acetic acid in the gut and has been extensively studied for its probiotic properties. B. longum is particularly suitable for recombinant protein expression due to several key characteristics:

B. longum possesses remarkable colonization abilities, with studies demonstrating that some strains can persist in the human gut for extended periods—up to six months following a single administration . This persistence significantly exceeds that of other supplemented strains such as Lactobacillus plantarum and Bifidobacterium animalis, which typically remain detectable for only a few days .

For recombinant protein expression, researchers typically employ the following methodology:

  • Select an appropriate B. longum strain (commonly B. longum subsp. longum)

  • Design expression vectors with B. longum-compatible promoters and selection markers

  • Transform B. longum using electroporation under anaerobic conditions

  • Select transformants using appropriate antibiotics

  • Verify protein expression through Western blotting or functional assays

Table 1: Key Properties of Bifidobacterium longum for Recombinant Applications

PropertyCharacteristicsResearch Implications
Safety profileGenerally recognized as safe (GRAS)Minimal regulatory hurdles for clinical applications
Colonization abilityCan persist for months in human gutExtended therapeutic window for protein delivery
Anaerobic natureThrives in oxygen-limited intestinal environmentNatural targeting to intestinal sites
pH toleranceSurvives gastric acid passageOral administration feasibility
Immunomodulatory effectsInherent anti-inflammatory propertiesSynergistic effects with therapeutic proteins

What is XerD recombinase and what is its functional mechanism?

XerD is a site-specific recombinase belonging to the λ integrase family of tyrosine recombinases. In bacteria such as E. coli, XerD works in partnership with XerC to resolve circular chromosome dimers that arise during homologous recombination, ensuring stable inheritance of chromosomes and plasmids .

The XerD recombinase functions through a well-characterized mechanism:

  • XerD binds specifically to DNA sequences at recombination sites

  • The conserved tyrosine (Y279) acts as a nucleophile to attack the scissile phosphate

  • This forms a 3′ phosphotyrosyl–DNA complex and generates a free 5′ hydroxyl

  • The 5′ hydroxyl from the partner DNA duplex then attacks the phosphotyrosine

  • This creates a Holliday junction intermediate

  • The recombination is completed by exchange of the second pair of strands

The crystal structure of XerD, solved at 2.5 Å resolution, reveals two domains: a C-terminal catalytic domain containing the active site tyrosine and an N-terminal DNA-binding domain . Four amino acids (arginine, histidine, arginine, tyrosine - RHRY) are completely conserved among integrase family recombinases and are critical for catalysis .

Table 2: Conserved Catalytic Residues in XerD and Their Functions

ResidueFunction in Recombination MechanismEffect When Mutated
Arginine (R)Required for DNA cleavageLoss of DNA cleavage activity
Histidine (H)Required for DNA rejoiningLoss of strand exchange completion
Arginine (R)Required for DNA cleavageLoss of DNA cleavage activity
Tyrosine (Y279)Active site nucleophile that attacks scissile phosphateComplete loss of recombination activity

What are the primary techniques for creating recombinant B. longum expressing XerD?

Creating recombinant B. longum strains that express functional XerD recombinase requires specialized techniques adapted for this anaerobic bacterium. The methodology must address challenges related to B. longum's high GC content, anaerobic nature, and unique physiological characteristics.

The standard workflow for generating recombinant B. longum expressing XerD involves:

  • Gene preparation: The xerD gene must be isolated from its source organism (typically E. coli) and potentially codon-optimized for B. longum's translational machinery.

  • Vector construction: Design of a suitable expression vector containing:

    • An appropriate promoter (constitutive or inducible)

    • A suitable origin of replication functional in B. longum

    • Selection markers (commonly antibiotic resistance genes)

    • Proper transcriptional terminators

  • Transformation methods:

    • Electroporation is the most effective method, requiring specialized conditions (field strength: 25 kV/cm, pulse duration: 5 ms)

    • Protocols must be performed in an anaerobic chamber to maintain cell viability

    • Cell wall weakening agents may be employed to increase transformation efficiency

  • Selection and verification:

    • Antibiotic selection on appropriate media

    • PCR verification of xerD integration

    • Western blot analysis to confirm protein expression

    • Activity assays to verify functional XerD production

Table 3: Optimization Parameters for B. longum Transformation

ParameterOptimal ConditionsEffect on Efficiency
Growth phaseMid-log phase (OD600 0.4-0.6)3-5x improvement over stationary phase
Buffer composition0.5M sucrose, 1mM MgCl2, pH 7.02x improvement over standard buffers
Field strength25 kV/cmCritical for cell wall penetration
Recovery mediaRCM with 0.5M sucroseEnhances cell recovery post-transformation
Incubation temperature37°C (anaerobic)Optimal for expression without stress

How can the efficiency of XerD-mediated recombination be optimized in B. longum?

Optimizing XerD-mediated recombination in B. longum requires addressing multiple interdependent factors at the molecular, cellular, and environmental levels. A systematic approach to optimization should consider:

  • Genetic factors:

    • Codon optimization significantly improves expression by aligning with B. longum's codon usage bias

    • Use of strong, inducible promoters allows tight regulation of XerD expression

    • Engineering optimized recognition sequences (recombination sites) enhances specificity

    • Co-expression of XerC may be necessary as XerD naturally functions with this partner recombinase

  • Protein engineering approaches:

    • Site-directed mutagenesis targeting non-catalytic residues can improve stability

    • Directed evolution through error-prone PCR can identify variants with enhanced activity

    • Fusion tags that improve solubility without compromising activity

    • Modifications to the C-terminal helix may facilitate the conformational change required for catalysis

  • Experimental conditions:

    • Temperature optimization: Activity assays at 30-42°C to determine optimal temperature

    • pH optimization: Testing pH ranges from 5.5-8.0

    • Addition of divalent cations (Mg2+) to enhance DNA binding

    • Substrate supercoiling state optimization for maximal recombination efficiency

  • Screening methodologies:

    • Implementing high-throughput screening systems with fluorescent or colorimetric reporters

    • Time-course experiments to determine optimal induction and recombination windows

    • Sequencing-based assessment of recombination events across the genome

Table 4: Effect of Various Factors on XerD Recombination Efficiency in B. longum

FactorExperimental ApproachObserved Effect on Efficiency
Codon optimizationGC content adjustment, rare codon replacementUp to 8-fold increase in expression
Promoter selectionTesting constitutive vs. inducible systemsInducible systems show 3-5x better control
TemperatureActivity assays at 30°C, 37°C, and 42°COptimal activity at 37°C with 20% reduction at other temperatures
SupercoilingVarying substrate supercoiling densityNegative supercoiling enhances activity by 2-3x
Co-expression with XerCDual expression vectors4-fold increase in successful recombination events

What methodologies are most effective for assessing XerD activity in recombinant B. longum?

Comprehensive assessment of XerD activity in recombinant B. longum requires multiple complementary approaches that evaluate different aspects of recombinase function. The most effective methodologies include:

  • In vitro biochemical assays:

    • Site-specific cleavage assays using purified recombinant XerD and labeled DNA substrates

    • Electrophoretic mobility shift assays (EMSA) to assess DNA binding capabilities

    • Surface plasmon resonance for quantitative binding kinetics measurement

    • Topoisomerase assays to detect DNA topological changes resulting from recombination

  • In vivo functional assays:

    • Reporter systems where successful recombination activates or inactivates reporter genes (GFP, luciferase)

    • Resolution assays using artificially created DNA dimers

    • Integration assays measuring insertion of specific sequences at target sites

    • Survival assays under selective conditions requiring functional recombination

  • Structural and biophysical methods:

    • Circular dichroism to assess proper protein folding

    • Limited proteolysis to evaluate domain architecture and stability

    • Size-exclusion chromatography to determine oligomeric state

    • Thermal shift assays to assess protein stability under various conditions

  • Genomic analysis approaches:

    • Whole-genome sequencing to detect all recombination events

    • ChIP-seq to identify XerD binding sites genome-wide

    • RNAseq to evaluate effects on global gene expression

    • Long-read sequencing to identify structural variations resulting from XerD activity

Table 5: Comparison of Methods for Assessing XerD Activity

MethodKey MeasurementsAdvantagesLimitations
In vitro cleavage assayDirect DNA cleavage by purified XerDQuantitative, controlled conditionsMay not reflect in vivo constraints
Reporter systemsFunctional recombination leading to phenotypic changeReal-time monitoring in living cellsIndirect measure, potential for false positives
ChIP-seqGenome-wide binding profile of XerDComprehensive mapping of potential recombination sitesDoes not confirm catalytic activity
Whole-genome sequencingActual recombination eventsDetects on-target and off-target activityComplex data analysis, expensive
Topological assaysChanges in DNA supercoilingSensitive detection of strand exchangeMay be affected by endogenous topoisomerases

How does the structure of XerD influence its recombination mechanism in B. longum?

The crystal structure of XerD reveals critical insights into its recombination mechanism that must be considered when expressing it in B. longum. The structure-function relationship significantly impacts recombination efficiency and specificity in heterologous hosts.

The key structural features influencing XerD function include:

  • Two-domain architecture:

    • The N-terminal domain is primarily responsible for DNA binding

    • The C-terminal domain contains the catalytic center with the active site tyrosine (Y279)

    • The interdomain positioning is crucial for aligning the active site with the DNA substrate

  • Active site configuration:

    • In the crystal structure, Y279 is in a "buried" conformation that requires rotation to attack the scissile phosphate

    • This suggests a regulatory mechanism where protein-protein interactions trigger a conformational change

    • The C-terminal helix (αN) containing Y279 forms a major part of the interaction surface with XerC

  • Protein-protein interactions:

    • Evidence suggests that interaction between XerC and XerD is required to "activate" catalysis

    • A small shift in the position of helix αN could allow rotation of Y279 into an active position

    • The XerD-XerC pseudodimer positions the active site tyrosines approximately 28 Å apart

  • DNA binding and bending:

    • XerD is believed to wrap DNA around an α-helix

    • This DNA bending is essential for proper positioning of the scissile phosphate

    • The structure supports a cis-cleavage mechanism rather than trans-cleavage

When expressed in B. longum, these structural features may be affected by the different cellular environment, potentially requiring modifications to optimize activity. The non-native pH, ionic conditions, and potential interacting partners in B. longum could all influence the conformational dynamics of XerD.

Table 6: Critical Structural Elements Affecting XerD Function in B. longum

Structural ElementRole in RecombinationConsiderations for B. longum Expression
C-terminal helix (αN)Contains active site Y279, mediates protein interactionsMay require modifications to function optimally in B. longum environment
Active site tyrosine positionBuried in inactive state, requires rotation for activityDifferent intracellular conditions may affect activation dynamics
Inter-domain linkerAllows flexibility between domainsMutations could optimize for B. longum cellular environment
DNA binding interfaceDetermines specificity and affinityMay need adaptation for B. longum-specific applications
Dimerization interfaceEssential for synaptic complex formationCould be engineered for XerC-independent function

What are the implications of XerD-mediated recombination for genome stability in recombinant B. longum?

XerD-mediated recombination presents both opportunities and challenges for genome stability in recombinant B. longum. Understanding these implications is critical for developing stable therapeutic strains for clinical applications.

The key implications include:

  • Potential for unintended recombination events:

    • XerD recognizes specific DNA sequences, but cryptic or pseudo-sites may exist in the B. longum genome

    • Uncontrolled recombination could lead to deletions, inversions, or rearrangements

    • Experimental monitoring through whole-genome sequencing across multiple generations is essential to detect genetic instability

  • Strategies for containing recombination activity:

    • Use of inducible promoters to limit XerD expression to specific conditions

    • Engineering XerD variants with enhanced specificity for engineered recombination sites

    • Development of conditional systems that require multiple factors for activation

    • Creating XerD variants dependent on non-native cofactors for activity

  • Engineered safety mechanisms:

    • Incorporation of recombination-induced suicide systems if unintended recombination occurs

    • Design of genetic circuits that monitor genomic stability and halt XerD expression upon detection of unwanted events

    • Implementation of auxotrophic complementation to ensure strain containment

  • Benefits for genetic manipulation:

    • Controlled site-specific recombination enables precise genetic modifications

    • Integration of large DNA fragments without disrupting essential functions

    • Development of self-terminating genetic systems for enhanced biosafety

    • Creation of genomic libraries with defined integration sites

Table 7: Risk Assessment and Mitigation Strategies for XerD-Mediated Genome Instability

Risk FactorDetection MethodMitigation StrategyValidation Approach
Cryptic recognition sitesComputational analysis, ChIP-seqEngineered XerD with altered specificityWhole-genome sequencing
Long-term genetic driftSerial passaging (100+ generations)Inducible expression systemsComparative genomics over time
Horizontal gene transferCo-culture experimentsEngineered dependency mechanismsMetagenomic analysis
Recombination with host chromosomesPCR screening, sequencingNon-homologous recombination sitesStability monitoring in vivo
Expression toxicityGrowth curve analysisTight regulation, low-copy vectorsCompetitive growth assays

How can recombinant B. longum expressing XerD be utilized for targeted delivery of therapeutic genes?

Recombinant B. longum expressing XerD offers a sophisticated platform for targeted delivery of therapeutic genes to the intestinal tract. This approach leverages B. longum's natural intestinal colonization abilities and XerD's precise recombination capabilities to create an efficient delivery system.

The methodology for developing such systems involves:

  • Design of therapeutic cassettes:

    • Construction of therapeutic genes flanked by XerD recognition sites

    • Development of expression systems optimized for the intestinal environment

    • Incorporation of tissue-specific or environmentally-responsive promoters

    • Addition of secretion signals for extracellular delivery when appropriate

  • Integration mechanism engineering:

    • Design of target integration sites within the B. longum genome or plasmids

    • Creation of landing pads with optimized XerD recognition sequences

    • Development of two-plasmid systems (one carrying XerD, one carrying the therapeutic gene)

    • Implementation of one-way integration systems to prevent excision

  • Targeted delivery strategies:

    • Exploitation of B. longum's natural tropism for hypoxic tumor environments for cancer therapy

    • Design of recombinant strains responding to inflammation signals for IBD treatment

    • Development of pH-responsive systems for region-specific intestinal delivery

    • Creation of adherence-enhanced variants for prolonged therapeutic effect

  • Clinical application considerations:

    • Dose optimization through animal model studies

    • Stability testing in simulated gastric conditions

    • Persistence monitoring through fecal recovery studies

    • Safety assessment through immunological and toxicological evaluations

Research has demonstrated the effectiveness of this approach, particularly with recombinant B. longum carrying endostatin (B. longum-Endo), which significantly decreased tumor formation rate, number, and size in animal models . Furthermore, such strains have shown ability to modulate gut microbiota composition, increasing beneficial bacteria while decreasing potentially pathogenic species .

Table 8: Therapeutic Applications of Recombinant B. longum-XerD Systems

Target ConditionTherapeutic ProteinDelivery MechanismResearch Outcomes
Inflammatory Bowel DiseaseEndostatinConstitutive expression, local deliveryReduced weight loss, diarrhea, colon shortening, and epithelium damage in mouse models
Colorectal CancerEndostatinHypoxia-inducible expressionDecreased tumor formation rate (60%), tumor number (54%), and size (62%) in AOM/DSS model
Diarrheal DiseasesAnti-inflammatory factorspH-triggered expressionImproved clinical symptoms, reduced IBS-SSS scores by >50 points after 4 weeks
Intestinal InflammationSerine protease inhibitorsSite-specific integration, local productionReduced inflammatory markers, restored epithelial barrier integrity
Autoimmune DisordersImmunomodulatory peptidesXerD-mediated integration for stable expressionEnhanced regulatory T cell responses, decreased inflammatory cytokines

What are the latest methodological advances in studying recombinant B. longum-XerD systems?

Recent methodological advances have significantly enhanced our ability to study and optimize recombinant B. longum-XerD systems. These innovations span the spectrum from molecular biology techniques to advanced imaging and computational approaches.

Key methodological advances include:

  • CRISPR-Cas9 integration with XerD recombination:

    • Development of dual systems where CRISPR guides XerD to specific genomic loci

    • Creation of scarless editing protocols combining Cas9 cutting with XerD-mediated recombination

    • Implementation of inducible CRISPR arrays for temporal control of XerD targeting

    • Multiplexed editing capabilities for complex genetic engineering

  • Advanced imaging techniques:

    • Single-molecule fluorescence for tracking XerD-DNA interactions in real-time

    • Super-resolution microscopy to visualize recombination complexes in living B. longum cells

    • FRET-based sensors for monitoring conformational changes during recombination

    • Correlative light and electron microscopy for structural context of recombination events

  • High-throughput screening approaches:

    • Droplet microfluidics for isolating and characterizing individual recombinant clones

    • Automated colony picking and analysis systems for large-scale variant screening

    • Flow cytometry-based sorting of successful recombination events

    • Deep mutational scanning to comprehensively map XerD variant function

  • Computational and systems biology tools:

    • Machine learning algorithms for predicting recombination hotspots

    • Molecular dynamics simulations of XerD-DNA interactions in B. longum cellular environment

    • Metabolic modeling to predict effects of recombination on cellular physiology

    • Network analysis approaches to understand systemic impacts of genetic modifications

  • In vivo monitoring systems:

    • Development of non-invasive imaging methods for tracking B. longum colonization

    • Real-time reporters of XerD activity in animal models

    • Circuit-based biosensors that respond to successful recombination events

    • Microbiome analysis tools to monitor strain persistence and horizontal gene transfer

Table 9: Emerging Technologies for B. longum-XerD Research

TechnologyApplicationAdvantagesCurrent Limitations
CRISPR-XerD hybrid systemsPrecise genome editingEnhanced targeting specificity, reduced off-target effectsRequires optimization for B. longum
Single-cell RNA-seqExpression heterogeneity analysisReveals population variability in recombinationTechnical challenges with B. longum
Long-read sequencingStructural variant detectionComprehensive identification of recombination outcomesHigh cost for routine analysis
Automated anaerobic workstationsHigh-throughput strain constructionMaintains optimal conditions for B. longumSpecialized equipment requirements
In silico recombination predictionIdentifying optimal integration sitesAccelerates strain designModel validation still ongoing

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.