Recombinant Brucella suis biovar 1 Tyrosine recombinase XerD (xerD)

Shipped with Ice Packs
In Stock

Description

Introduction to Recombinant Brucella suis biovar 1 Tyrosine Recombinase XerD (xerD)

Recombinant Brucella suis biovar 1 Tyrosine recombinase XerD (xerD) refers to a genetically engineered version of the XerD enzyme from Brucella suis biovar 1, a bacterium known for causing brucellosis in animals and humans. Tyrosine recombinases, such as XerD, are enzymes involved in site-specific recombination, which is crucial for maintaining the stability of bacterial chromosomes and plasmids by resolving multimers into monomers before cell division .

Role of Tyrosine Recombinases in Bacteria

Tyrosine recombinases, including XerC and XerD, are essential for bacterial DNA management. They facilitate site-specific recombination at specific DNA sequences, known as dif sites, which helps in resolving chromosome dimers and plasmid multimers into monomers. This process is vital for ensuring proper segregation of genetic material during bacterial cell division .

Brucella suis biovar 1 Overview

Brucella suis biovar 1 is a pathogenic bacterium primarily affecting pigs but can also infect other animals and humans. It is known for its zoonotic potential, meaning it can be transmitted from animals to humans . The bacterium is difficult to culture and diagnose, often leading to underdiagnosis .

Brucella suis biovar 1 Infections

  • Animal Hosts: Mainly found in domesticated pigs, feral pigs, and wild boars, but can also infect other animals like cattle, sheep, and goats .

  • Human Infections: While rare, B. suis biovar 1 can cause severe infections in humans, often acquired through contact with infected animals or contaminated products .

Genome Analysis

  • Genome Sequencing: Recent studies have utilized whole-genome sequencing to analyze B. suis strains, providing insights into their phylogenetic relationships and potential transmission routes .

  • MLVA and cgMLST Analysis: These techniques help in tracing the origin of isolates and understanding genetic diversity among strains .

Data Table: Brucella suis biovar 1 Characteristics

CharacteristicDescription
Primary HostsDomesticated pigs, feral pigs, wild boars
Zoonotic PotentialHigh risk of transmission to humans
Genome SizeApproximately 3.3 Mb (two circular chromosomes)
Serological TestsVarious tests used for diagnosis, including ELISA and agglutination tests
Treatment ChallengesDifficult due to intracellular nature and antibiotic resistance

This table highlights key aspects of Brucella suis biovar 1 but does not specifically address recombinant XerD due to the lack of detailed information on this compound.

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your preferred format in order notes for customized fulfillment.
Lead Time
Delivery times vary depending on the purchasing method and location. Contact your local distributor for precise delivery estimates.
Note: All proteins are shipped with standard blue ice packs. Dry ice shipping requires prior arrangement 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 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 various factors including storage conditions, buffer composition, temperature, and protein 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. If you require a specific tag, please inform us; we will prioritize its development.
Synonyms
xerD; BR2031; BS1330_I2025; Tyrosine recombinase XerD
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-309
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Brucella suis biovar 1 (strain 1330)
Target Names
xerD
Target Protein Sequence
MTMRASLAIE NFLEMMSAER GAAQNTLESY RRDLEAAAEE LAAKGVNLAE AETGHIRMTL DTMAAQGFAP TSQARRLSAL RQFFRFLYSE GFRQDDPTGI LDAPKKQKPL PKIMSVENVG RLLDRAALEA NEAAEPGERI KALRLHALLE TLYATGLRVS ELVGLPVTVA RTDHRFLLVR GKGSKDRMVP LSRKARDALQ KFLTLRDSLP GSDDNPWLFP AFSESGHLAR QVFARELKGL AARAGLAASS VSPHVLRHAF ASHLLQNGAD LRTVQQLLGH ADISTTQIYT HVLEERLHKL VSEHHPLAD
Uniprot No.

Target Background

Function
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 plasmid stability.
Database Links

KEGG: bms:BR2031

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

Q&A

What is Brucella suis biovar 1 and what distinguishes it from other biovars?

Brucella suis biovar 1 is a zoonotic bacterial pathogen primarily associated with domesticated and feral pigs. It is distinguished from other B. suis biovars by specific biochemical and molecular characteristics. Biovar 1 is one of five recognized B. suis biovars, with biovars 1, 2, and 3 typically infecting pigs and wild boars, biovar 4 primarily affecting caribou and reindeer, and biovar 5 found only in rodents .

Biovar 1 is characterized by:

  • High virulence for humans (along with biovar 3)

  • Typical ability to produce H₂S

  • No growth on thionin (1/25,000) or basic fuchsin dye (20 µg/ml)

  • Lysis by TB phage at a concentration of 10⁴ × RTD

In Brazil and many parts of the Americas, biovar 1 is the predominant strain isolated from pigs , while in Europe, biovar 2 is more common in wild boar populations .

What is the XerD tyrosine recombinase and what is its function in bacterial cells?

XerD is a site-specific tyrosine recombinase that, together with its paralog XerC, constitutes the XerCD recombination system. This system plays a crucial role in bacterial chromosome maintenance by resolving chromosome dimers that form during replication.

The XerCD system:

  • Acts on a specific 28 bp DNA sequence called the dif site, located in the replication terminus (ter) region

  • Forms a synaptic complex consisting of two XerC and two XerD subunits bound to two dif sites

  • Is controlled by the septal protein FtsK, ensuring recombination occurs at the right time (immediately prior to cell division) and place (cell division septum)

  • Is highly conserved across bacterial species including Proteobacteria, Archaea, and Firmicutes

The XerCD site-specific recombination system is essential for bacterial chromosome stabilization, preventing the formation of chromosome dimers during replication that would otherwise interfere with proper chromosome segregation .

Why is Brucella suis biovar 1 XerD of particular interest in bacterial genomics research?

Brucella suis biovar 1 XerD is of significant interest because:

  • Critical role in genome stability: The XerCD system is essential for maintaining chromosome integrity in Brucella, with mutations in xerC/xerD potentially leading to genomic instability .

  • Evolutionary significance: Comparative genomic analyses suggest that the acquisition of certain genomic elements and adaptation to limited-metal environments were critical evolutionary steps for Brucella development from soil bacteria ancestors . Understanding XerD's role in this evolution provides insights into Brucella pathogenicity.

  • Potential therapeutic target: As a protein essential for chromosome maintenance, XerD represents a potential therapeutic target for developing novel antibiotics against brucellosis, a significant zoonotic disease .

  • Unique characteristics: Research shows that variations in XerC/XerD proteins can affect bacterial phenotype, as demonstrated in studies of Brucella melitensis where xerC mutations resulted in an incomplete Xer functional domain affecting normal function .

  • Role in genomic rearrangements: XerD may contribute to genomic plasticity and adaptation of Brucella to different hosts and environments, influencing virulence and host specificity .

How does the structure of XerD in Brucella suis biovar 1 compare to XerD in other bacterial species?

XerD in Brucella suis biovar 1 shares structural similarities with XerD proteins across bacterial species, but with specific adaptations. Based on available research:

  • Conserved domains: Brucella XerD contains two major functional domains similar to those found in other bacteria:

    • The Xer domain - responsible for DNA binding and catalysis

    • The Phage integrase domain - involved in recombination functions

  • Sequence conservation: While the catalytic domain fold is conserved among tyrosine recombinases, there are species-specific variations. Limited structural information from various tyrosine recombinases has facilitated the development of general models that can be applied to Brucella XerD .

  • Comparative analysis: Alignment studies between Brucella melitensis strains have shown that mutations in xerC genes can result in altered functional domains, suggesting similar critical regions likely exist in XerD . For example, in B. melitensis CMCC55210a, a deletion of cytosine at site 160 in xerC resulted in shortening of the 5′ end of the gene and an incomplete Xer functional domain .

  • Evolutionary considerations: Brucella species have undergone approximately 30% genome reduction during evolution, particularly in proteins involved in carbohydrate and amino acid utilization, metabolism, and biosynthesis . This evolutionary pressure may have shaped XerD's structure to be optimized for Brucella's intracellular lifestyle.

The structural conservation of XerD across bacterial species reflects its essential function, while species-specific variations likely contribute to adaptation to different ecological niches.

What is the mechanism by which XerD facilitates site-specific recombination in Brucella suis biovar 1?

The XerD-mediated site-specific recombination in Brucella suis biovar 1 follows a highly coordinated mechanism similar to the well-characterized process in other bacteria, particularly E. coli:

  • Recognition and binding:

    • XerD, along with XerC, recognizes and binds to the 28 bp dif site located in the terminus region of the Brucella chromosome

    • Each recombinase binds to a specific half of the dif site, forming a synaptic complex consisting of two XerD and two XerC proteins bound to two dif sites

  • Activation by FtsK:

    • The septal protein FtsK controls the initiation of the recombination reaction

    • FtsK ensures recombination occurs at the right time (immediately prior to cell division) and at the right place (cell division septum)

    • This regulation is crucial for proper chromosome segregation

  • Catalytic steps:

    • Upon activation, XerD initiates the first strand exchange, creating a Holliday junction intermediate

    • The Holliday junction is then resolved by XerC-mediated strand exchange, completing the recombination process

    • This coordinated action resolves chromosome dimers into monomers

  • Redundancy mechanisms:

    • Research suggests that redundancy mechanisms exist in Brucella to ensure chromosome stabilization

    • Recent studies in other bacteria revealed that XerD can unload structural maintenance of chromosome (SMC) complexes through binding to additional chromosomal loci beyond dif sites, in a manner that does not depend on XerC

    • This redundancy may provide additional safeguards for genome stability in Brucella

The site-specific recombination mechanism ensures precise resolution of chromosome dimers, preventing potential chromosome segregation failures that would otherwise lead to cell death or genomic instability.

What are the recommended methods for cloning and expressing recombinant Brucella suis biovar 1 XerD protein?

Recommended methods for cloning and expressing recombinant B. suis biovar 1 XerD:

  • Gene isolation and vector selection:

    • PCR amplification of the xerD gene from genomic DNA using high-fidelity polymerase

    • Recommended vectors: pET expression systems for E. coli or broad-host-range plasmids like pbbr1 for expression in Brucella

    • Include appropriate purification tags (His-tag, GST-tag) for downstream purification

  • Expression systems:

    • E. coli BL21(DE3) or derivatives for high-yield expression

    • Alternatively, yeast, baculovirus, or mammalian cell systems for proteins requiring eukaryotic post-translational modifications

    • Consider using a Brucella-specific codon optimization if expressing in heterologous systems

  • Induction conditions:

    • For E. coli: IPTG induction (0.1-1.0 mM) at lower temperatures (16-25°C) to enhance solubility

    • Monitor growth at OD600 and adjust induction timing to mid-log phase

    • Extended expression times (overnight) at lower temperatures often improve yield of soluble protein

  • Protein purification strategy:

    • Immobilized metal affinity chromatography (IMAC) for His-tagged proteins

    • Size exclusion chromatography for further purification and buffer exchange

    • Ion exchange chromatography may be necessary for removing contaminants

    • Consider adding DNA nucleases during lysis to prevent DNA contamination

  • Quality control assessments:

    • SDS-PAGE and Western blot verification

    • Mass spectrometry for identity confirmation

    • Activity assays using synthetic dif site oligonucleotides to confirm functionality

  • Storage considerations:

    • Add glycerol (10-20%) for cryoprotection

    • Flash-freeze in liquid nitrogen and store at -80°C in small aliquots

    • Avoid repeated freeze-thaw cycles

This methodology builds on established protocols for recombinant protein expression while addressing specific considerations for Brucella proteins.

What assays can be used to evaluate the recombination activity of Brucella suis biovar 1 XerD in vitro?

In vitro assays for evaluating B. suis biovar 1 XerD recombination activity:

  • DNA binding assays:

    • Electrophoretic Mobility Shift Assay (EMSA): To assess binding of XerD to dif site DNA

    • Fluorescence Anisotropy: For quantitative measurement of XerD-DNA interaction kinetics

    • Surface Plasmon Resonance (SPR): For real-time binding analysis and determination of association/dissociation constants

  • Strand cleavage and exchange assays:

    • Suicide substrate assay: Using radiolabeled or fluorescently-labeled oligonucleotides containing the dif site

    • DNA nicking assay: To assess the first step of catalysis (single-strand cleavage)

    • Holliday junction resolution assay: To evaluate complete recombination reactions with both XerC and XerD

  • Complete recombination assays:

    • Plasmid resolution assay: Using reporter plasmids containing directly repeated dif sites

    • Integration/excision assay: To evaluate recombination between different DNA molecules

    • FtsK-dependent activation assay: To assess the role of FtsK in stimulating XerD activity

  • Structure-function analysis:

    • Site-directed mutagenesis of catalytic residues followed by activity testing

    • Limited proteolysis to identify domain boundaries and structural elements

    • Thermal shift assays to evaluate protein stability under different conditions

  • Interaction studies:

    • Co-immunoprecipitation or pull-down assays to verify XerC-XerD interaction

    • Bacterial two-hybrid assays for protein-protein interaction analysis

    • Analytical ultracentrifugation to determine oligomeric states

  • Microscopy-based techniques:

    • Fluorescence Resonance Energy Transfer (FRET) to observe real-time recombination

    • Single-molecule techniques to observe individual recombination events

These assays provide complementary information about XerD activity, from initial DNA binding through complete recombination reactions, and would establish the biochemical properties of B. suis biovar 1 XerD in comparison to well-characterized recombinases from model organisms.

What molecular techniques are most effective for studying XerD function in the context of live Brucella suis biovar 1?

Effective molecular techniques for studying XerD function in live B. suis biovar 1:

  • Genetic manipulation approaches:

    • Targeted gene deletion using homologous recombination

    • CRISPR-Cas9 system for precise genome editing

    • Conditional knockdown systems (e.g., tetracycline-regulated expression) for essential genes

    • Complementation with wild-type or mutant xerD variants to verify phenotypes

  • Expression monitoring:

    • RT-qPCR for measuring xerD transcript levels under different conditions

    • RNA-Seq for genome-wide transcriptional response to xerD manipulation

    • Western blotting with specific antibodies to monitor XerD protein levels

    • Fluorescent protein fusions to track XerD localization (with caution regarding function)

  • Phenotypic characterization:

    • Growth curve analysis under various stress conditions

    • Microscopy to observe cell morphology and division defects

    • Flow cytometry to assess DNA content and cell cycle progression

    • Competitive growth assays to measure relative fitness effects

  • In vivo recombination monitoring:

    • Reporter systems with strategically placed recombination sites

    • PCR-based detection of recombination products

    • Next-generation sequencing to identify genome-wide recombination events

    • Chromosome conformation capture techniques to analyze 3D genome structure

  • Host-pathogen interaction studies:

    • Macrophage infection models to assess intracellular survival of xerD mutants

    • Animal infection models to evaluate virulence (with appropriate biosafety precautions)

    • Comparative genomics of isolates recovered from different host tissues

  • Advanced microscopy techniques:

    • Super-resolution microscopy to visualize chromosome segregation

    • Time-lapse microscopy to monitor cell division dynamics

    • Fluorescence recovery after photobleaching (FRAP) to assess protein dynamics

When working with Brucella suis biovar 1, it's crucial to implement appropriate biosafety measures (BSL-3 containment) due to its zoonotic potential and pathogenicity to humans . Researchers should consider using attenuated strains or related Brucella species with lower pathogenicity for initial studies when possible.

How can recombinant Brucella suis biovar 1 XerD be utilized in vaccine development strategies?

Applications of recombinant B. suis biovar 1 XerD in vaccine development:

  • Subunit vaccine component:

    • Purified recombinant XerD could be incorporated into subunit vaccines

    • Particularly valuable if XerD contains conserved epitopes across Brucella species

    • Can be combined with other immunogenic Brucella proteins for enhanced protection

    • Advantage of avoiding live bacteria while still inducing protective immunity

  • Attenuated live vaccine development:

    • Controlled mutation of xerD could generate attenuated strains with reduced virulence

    • XerD conditional expression systems could create strains viable for immunization but limited in long-term persistence

    • Such strains might mimic natural infection pathways, inducing robust cell-mediated immunity

    • Research indicates that cell-mediated immune responses are critical in resistance against intracellular bacterial infections like brucellosis

  • Genetic adjuvant strategies:

    • XerD-based recombination systems could be engineered to control expression of immunostimulatory molecules

    • Site-specific recombination could enable controlled genetic rearrangements during vaccination

    • This approach could enhance antigen presentation or cytokine expression in vaccine strains

  • DNA vaccine applications:

    • xerD gene sequences could be incorporated into DNA vaccines

    • Potential for co-delivery with other Brucella antigens to enhance immune response

    • DNA vaccines could induce both humoral and cell-mediated immunity

  • Vaccine delivery platform:

    • XerD-mediated site-specific recombination could be utilized to develop sophisticated antigen delivery systems

    • Controlled expression of heterologous antigens through recombination events

    • Similar to approaches used with other recombinases in vaccine development

  • Enhanced safety mechanisms:

    • XerD systems could be engineered to create genetic kill-switches in live attenuated vaccines

    • Similar to approaches used with the RB51 strain, which when modified with additional proteins showed enhanced immune responses

    • These safety mechanisms could prevent vaccine reversion to virulence

When developing vaccine strategies based on XerD, it's important to consider that while the RB51 strain is a live attenuated vaccine with low side effects compared to other brucellosis vaccines, it provides insufficient protective efficacy on its own . Therefore, XerD-based approaches would likely need to be combined with other strategies to achieve optimal protection.

What are the implications of XerD research for understanding Brucella suis biovar 1 evolution and pathogenicity?

Implications of XerD research for B. suis biovar 1 evolution and pathogenicity:

  • Evolutionary insights:

    • XerD conservation across bacterial species suggests it represents an ancient and essential system

    • Studies indicate XerC/D recombinases in proteobacteria follow vertical inheritance patterns, providing reliable phylogenetic markers

    • Research on the XerCD system can help understand how Brucella evolved from soil bacteria ancestors to intracellular pathogens

    • Genomic analysis shows Brucella underwent approximately 30% genome reduction during evolution, particularly affecting metabolism genes

  • Host adaptation mechanisms:

    • XerD's role in maintaining genomic stability may be crucial during host adaptation

    • Different B. suis biovars show host preferences (biovar 1 in pigs, biovar 4 in caribou), suggesting genomic adaptations

    • Understanding XerD function may reveal how Brucella maintains genomic integrity during host switches

    • Research suggests acquisition of the VirB type 4 secretion system and adaptation to limited-metal environments were critical evolutionary steps for Brucella

  • Virulence regulation:

    • Proper chromosome maintenance is essential for bacterial fitness during infection

    • XerD dysfunction may lead to attenuated virulence due to genomic instability

    • Studies in B. melitensis show connections between chromosome maintenance genes and phenotypic characteristics relevant to virulence

    • XerD research may reveal mechanisms of phenotypic switching between rough and smooth colony types

  • Persistence mechanisms:

    • Long-term persistence in host tissues requires robust genomic stability mechanisms

    • XerD function may be particularly important during chronic infection phases

    • Understanding how XerD operates under stress conditions could explain Brucella's remarkable persistence

  • Comparative genomics applications:

    • XerD sequence analysis across Brucella isolates may help track evolutionary relationships

    • Biovar-specific variations in XerD could provide insights into host adaptation processes

    • Whole genome sequencing studies have revealed that XerD conservation is high across bacterial phyla

  • Zoonotic transmission insights:

    • B. suis biovar 1 is highly pathogenic to humans, unlike biovar 2 which rarely causes human disease

    • Research into genomic stability mechanisms may reveal factors contributing to zoonotic potential

    • Case studies suggest B. suis biovar 1 can establish in unexpected hosts, as seen in a dog infection case in Germany

This research has significant implications for understanding how Brucella maintains genomic integrity during host adaptation and infection processes, potentially revealing new approaches for disease control.

How does the function of XerD in Brucella suis biovar 1 compare with its role in other significant bacterial pathogens?

Comparative analysis of XerD function across bacterial pathogens:

  • Conserved core functions:

    • Across bacterial pathogens, XerD maintains the fundamental role in chromosome dimer resolution

    • The basic mechanism involving cooperation with XerC and action at dif sites remains conserved

    • The catalytic mechanism involving tyrosine-mediated DNA cleavage and strand exchange is preserved

    • FtsK-mediated activation of XerD is a common regulatory mechanism across diverse bacteria

  • Structural variations:

    • While the catalytic domain fold is conserved among tyrosine recombinases, species-specific variations exist

    • In well-studied systems like E. coli, XerD consists of 298 amino acids, forming a specific structural arrangement with XerD

    • Limited structural information from various tyrosine recombinases (XerD, XerA, XerH) has allowed development of general models applicable across species

    • These models reveal both conserved features and species-specific adaptations

  • Alternative systems in some bacteria:

    • While XerCD is widely distributed, some bacteria employ alternative systems

    • Streptococci and Lactococci use a single recombinase (XerS) with an atypical 31 bp recombination site (difSL)

    • These alternative systems highlight evolutionary adaptability while maintaining essential function

  • Additional functions beyond chromosome resolution:

    • Recent studies in Staphylococcus aureus and Bacillus subtilis revealed that XerD unloads structural maintenance of chromosome (SMC) complexes through binding to additional chromosomal loci

    • This function does not depend on XerC, suggesting XerD has independent roles beyond the classic XerCD system

    • Similar redundancy mechanisms may exist in Brucella and other pathogens

  • Relationship to mobile genetic elements:

    • In some bacteria, XerD-related recombinases facilitate integration of mobile genetic elements

    • Comparative bioinformatics suggests the VirB type 4 secretion system in Brucella, critical for virulence, has evolutionary connections to plasmid transfer systems

    • The relationship between chromosome maintenance systems and mobile genetic elements reveals important evolutionary connections

  • Biovar-specific adaptations:

    • B. suis biovar 1 shows high virulence for humans compared to biovar 2, which rarely causes human disease

    • These pathogenicity differences may relate to genomic stability mechanisms and adaptation processes

    • Comparative studies of XerD across biovars could reveal mechanisms underlying these differences

This comparative perspective provides valuable insights into both universal aspects of bacterial chromosome maintenance and pathogen-specific adaptations that may contribute to virulence and host range differences.

What role might XerD play in genomic plasticity and host adaptation of Brucella suis biovar 1?

XerD's potential role in genomic plasticity and host adaptation:

  • Balanced genomic stability:

    • XerD must provide sufficient stability for genomic integrity while allowing beneficial genetic variation

    • This balance may be particularly important during host adaptation processes

    • Research suggests Brucella evolution involved ~30% genome reduction, indicating significant genomic plasticity during adaptation

  • Response to host-specific selective pressures:

    • Different B. suis biovars show distinct host preferences (e.g., biovar 1 in pigs, biovar 4 in caribou)

    • XerD's function may be fine-tuned to maintain genomic stability under host-specific stresses

    • The system must accommodate selective pressures without compromising essential functions

  • Potential interactions with mobile genetic elements:

    • The site-specific recombination performed by XerD shares mechanistic similarities with integration events

    • Research suggests a VirB type 4 secretion system similar to Brucella's was found on a plasmid with broad host range

    • XerD might interact with or influence the integration/excision of adaptive genetic elements

  • Biovar-specific genomic features:

    • Whole genome sequencing studies have revealed genomic differences between B. suis biovars

    • In one study using combined assembly of Illumina and Nanopore reads, a B. suis genome consisted of two circular contigs of 2,107,952 and 1,207,151 bp with 3,113 predicted coding sequences

    • XerD's role in maintaining these genomic structures may influence biovar-specific characteristics

  • Adaptation to intracellular lifestyle:

    • Brucella's adaptation from soil bacteria ancestors to intracellular pathogens involved critical evolutionary steps

    • XerD's function in maintaining chromosome stability during replication in different cellular environments

    • The intracellular niche presents unique stresses that may require specialized genome maintenance mechanisms

  • Potential influence on phase variation:

    • Some bacterial pathogens use recombination-based phase variation to adapt to host environments

    • XerD might participate in regulated genomic rearrangements that alter surface structures or virulence factors

    • This could contribute to the observed phenotypic variations between isolates and during infections

Future research should investigate XerD sequence variations across B. suis isolates from different hosts and geographic regions, combined with functional studies to determine how these variations affect recombination efficiency and genomic stability under different environmental conditions.

How might advances in structural biology enhance our understanding of XerD function in Brucella suis biovar 1?

Contributions of structural biology to understanding B. suis biovar 1 XerD:

  • Complete structural characterization:

    • High-resolution crystal or cryo-EM structures of B. suis XerD would reveal specific adaptations

    • Comparative analysis with existing recombinase structures from E. coli XerD (Subramanya et al., 1997), XerA, XerH, and related tyrosine recombinases like Cre, HP1 integrase, FLP, and λ integrase

    • Identification of unique structural features that might contribute to biovar-specific functions

  • Complex formation visualization:

    • Structures of XerD bound to dif DNA sites would reveal binding specificity determinants

    • Co-structures with XerC would elucidate the synaptic complex architecture

    • FtsK-XerD complexes would show how chromosome segregation is coordinated with recombination

    • These complex structures would clarify the molecular basis of regulated recombination

  • Catalytic mechanism insights:

    • Structures capturing reaction intermediates would reveal the precise catalytic mechanism

    • Identification of biovar-specific variations in catalytic residues or cofactor requirements

    • Understanding how recombination is regulated at the molecular level to prevent inappropriate reactions

  • Domain dynamics and flexibility:

    • Nuclear Magnetic Resonance (NMR) studies could capture the dynamic aspects of XerD function

    • Molecular dynamics simulations based on structural data would reveal functional movements

    • These approaches would complement static structures with information about conformational changes during recombination

  • Structure-guided functional studies:

    • Rational design of mutations based on structural information to test functional hypotheses

    • Development of specific inhibitors targeting B. suis XerD for potential therapeutic applications

    • Engineering modified XerD variants with novel properties for biotechnological applications

  • Comparison with alternative XerD systems:

    • Structural studies of XerD from different Brucella species and biovars could reveal adaptations

    • Comparison with single-recombinase systems like XerS from Streptococci would provide evolutionary insights

    • These comparisons would highlight conserved core functions versus species-specific adaptations

Recent advances in structural biology techniques, particularly cryo-EM, which can determine structures without the need for crystallization, make these studies increasingly feasible. The resulting structural insights would significantly advance our understanding of how XerD functions in the specific context of B. suis biovar 1 and could guide the development of targeted interventions.

What are the most promising experimental approaches for developing XerD-targeted antimicrobial strategies against Brucella suis biovar 1?

Promising experimental approaches for XerD-targeted antimicrobials:

  • Structure-based inhibitor design:

    • High-throughput virtual screening against structural models of B. suis XerD

    • Fragment-based drug discovery targeting the catalytic site or DNA-binding interface

    • Rational design of competitive inhibitors that mimic DNA substrates or reaction intermediates

    • Focus on inhibitors that specifically target bacterial tyrosine recombinases without affecting human enzymes

  • Peptide-based inhibitors:

    • Development of peptides that interfere with XerD-XerC interactions

    • Design of peptides that block FtsK-mediated activation of XerD

    • Cyclization or other stabilization strategies to enhance peptide stability and cell penetration

    • These approaches target protein-protein interactions rather than catalytic activity

  • DNA mimetics and decoys:

    • Synthetic oligonucleotides that mimic dif sites but cannot be processed by XerD

    • Modified DNA structures that trap XerD in non-productive complexes

    • Delivery systems to introduce these molecules into bacterial cells

    • This strategy exploits XerD's natural DNA-binding specificity

  • Allosteric modulators:

    • Identification of allosteric sites that control XerD activity

    • Small molecules that lock XerD in inactive conformations

    • Compounds that disrupt the precise positioning required for catalysis

    • This approach may offer higher specificity than active site inhibitors

  • CRISPR-Cas gene editing strategies:

    • CRISPR-based antimicrobials targeting xerD gene sequences

    • Delivery systems specific for Brucella (phage-based or liposomal)

    • Programmable nucleases that specifically disrupt xerD function

    • This genetic approach directly eliminates XerD production

  • Combination approaches:

    • XerD inhibitors combined with traditional antibiotics for synergistic effects

    • Multi-target strategies addressing XerD along with other essential bacterial functions

    • Integration with immune-enhancing therapies for brucellosis treatment

    • This multi-pronged approach could reduce resistance development

Table 1: Advantages and Challenges of XerD-Targeted Antimicrobial Approaches

ApproachAdvantagesChallengesDevelopment Stage
Structure-based inhibitorsHigh specificity, rational design possibilitiesRequires detailed structural information, delivery challengesEarly research
Peptide inhibitorsCan target protein interfaces, high specificityLimited stability in vivo, delivery challengesPreclinical research
DNA mimeticsExploits natural substrate recognition, high specificityNuclease degradation in vivo, delivery challengesConceptual
Allosteric modulatorsMay access uniquely bacterial regulatory mechanismsDifficult to identify effective binding sitesEarly research
CRISPR-Cas strategiesDirect genetic targeting, potential for high specificityDelivery systems needed, off-target concernsEmerging technology
Combination approachesSynergistic effects, reduced resistance developmentComplex development and regulatory pathwayTheoretical

These approaches target an essential bacterial function that lacks direct human homologs, potentially offering selective toxicity against Brucella with reduced side effects compared to broad-spectrum antibiotics.

What biosafety precautions are essential when working with recombinant Brucella suis biovar 1 proteins including XerD?

Essential biosafety precautions for working with B. suis biovar 1 proteins:

  • Risk assessment and containment requirements:

    • B. suis biovar 1 is classified as a BSL-3 pathogen and potential bioterrorism agent

    • Work with live organisms requires BSL-3 containment facilities

    • Recombinant proteins derived from B. suis may be handled at BSL-2 if properly purified

    • Conduct thorough risk assessment before beginning work to determine appropriate containment

  • Personal protective equipment (PPE):

    • Laboratory coat, preferably with back closure and tight cuffs

    • Double gloves when handling potentially infectious materials

    • Eye protection (safety glasses, goggles, or face shield)

    • Respiratory protection may be required depending on procedures

    • Closed-toe shoes and laboratory-specific clothing

  • Engineering controls:

    • Certified biological safety cabinet (Class II or III) for all manipulations

    • Sealed centrifuge rotors or safety cups for centrifugation

    • HEPA-filtered vacuum protection for aspiration

    • Designated equipment for B. suis work to prevent cross-contamination

  • Work practices:

    • Strict adherence to standard microbiological practices

    • No mouth pipetting, eating, drinking, or applying cosmetics in laboratory

    • Minimize creation of aerosols and splashes

    • Maintain detailed records of all work with B. suis materials

    • Regular decontamination of work surfaces

  • Decontamination procedures:

    • Effective disinfectants include 1% sodium hypochlorite, 70% ethanol, 2% glutaraldehyde, or formaldehyde

    • Autoclave all contaminated materials before disposal

    • Validate decontamination procedures regularly

    • Follow institutional waste management protocols for biological waste

  • Emergency response:

    • Develop and practice spill response procedures

    • Document exposure response protocols

    • Maintain emergency contact information visibly posted

    • Report all accidents, exposures, and near-misses

  • Personnel considerations:

    • Proper training of all personnel before beginning work

    • Medical surveillance program for workers

    • Consider vaccination status where applicable

    • Restrict laboratory access to authorized personnel only

Research has documented occupational risks associated with Brucella, including a case where a veterinary clinic employee was potentially infected while handling a dog with B. suis biovar 1 infection . Even with recombinant proteins, proper risk assessment and appropriate precautions are essential to prevent laboratory-acquired infections.

What are the most effective expression systems for producing high-quality recombinant Brucella suis biovar 1 XerD protein for structural and functional studies?

Optimal expression systems for high-quality B. suis biovar 1 XerD:

  • E. coli-based expression systems:

    • pET vector systems: Provide tight regulation and high expression levels under T7 promoter control

    • Recommended strains: BL21(DE3) derivatives, particularly Rosetta or CodonPlus for handling potential rare codon usage in Brucella genes

    • Fusion tags: N-terminal His6, MBP, or SUMO tags to enhance solubility and facilitate purification

    • Induction conditions: Lower temperatures (16-18°C) and reduced IPTG concentrations (0.1-0.5 mM) to enhance solubility

    • Advantages: High yield, cost-effective, well-established protocols

    • Limitations: Potential for inclusion body formation, lack of post-translational modifications

  • Cell-free protein synthesis:

    • Uses E. coli or wheat germ extracts supplemented with necessary components

    • Allows rapid screening of expression conditions and protein variants

    • Especially useful for potentially toxic proteins

    • More expensive than in vivo systems but offers greater control over expression environment

  • Yeast expression systems:

    • Pichia pastoris or Saccharomyces cerevisiae for eukaryotic expression

    • Provides some post-translational modifications and often good folding

    • Generally lower yields than E. coli but potentially higher solubility

    • Useful if E. coli expression results in primarily insoluble protein

  • Baculovirus-insect cell system:

    • High expression levels with eukaryotic folding and processing machinery

    • Recommended for complex proteins difficult to express in bacterial systems

    • More time-consuming and expensive than bacterial expression

    • Often produces properly folded active proteins

  • Mammalian cell expression:

    • HEK293 or CHO cells for maximum authenticity of folding and modifications

    • Lowest yields among common expression systems but highest quality

    • Recommended only if other systems fail to produce functional protein

    • Transient transfection for rapid screening, stable cell lines for larger scale production

Table 2: Comparison of Expression Systems for B. suis biovar 1 XerD Production

Expression SystemYieldCostTime RequiredFolding QualityModificationsScalability
E. coli pETHighLow1-2 daysModerateMinimalExcellent
Cell-freeLowHighHoursVariableMinimalPoor
YeastMediumMedium3-7 daysGoodSomeGood
BaculovirusMedium-HighHigh7-14 daysVery goodMostGood
Mammalian cellsLowVery high1-4 weeksExcellentFullLimited

For most applications, E. coli-based expression with optimization of solubility (through fusion tags and expression conditions) provides the best balance of yield, cost, and protein quality for XerD structural and functional studies. This approach has been successfully used for other tyrosine recombinases, including the E. coli XerD protein .

What are the critical quality control parameters for ensuring the activity and specificity of purified recombinant Brucella suis biovar 1 XerD?

Critical quality control parameters for recombinant B. suis biovar 1 XerD:

  • Purity assessment:

    • SDS-PAGE with Coomassie or silver staining (>95% purity recommended)

    • Analytical size exclusion chromatography to evaluate homogeneity

    • Mass spectrometry to confirm molecular weight and detect contaminating proteins

    • Endotoxin testing particularly important for proteins intended for immunological studies

  • Identity verification:

    • Western blotting with anti-XerD antibodies or tag-specific antibodies

    • Peptide mass fingerprinting by mass spectrometry

    • N-terminal sequencing to confirm correct processing

    • Full-length integrity assessment to detect truncations or degradation products

  • Structural integrity:

    • Circular dichroism (CD) spectroscopy to assess secondary structure

    • Thermal shift assays to evaluate stability and proper folding

    • Intrinsic fluorescence spectroscopy to examine tertiary structure

    • Dynamic light scattering to determine size distribution and detect aggregation

  • Functional activity assessment:

    • DNA binding assays using electrophoretic mobility shift assay (EMSA) with dif site oligonucleotides

    • Catalytic activity assays measuring DNA cleavage and strand exchange

    • XerC interaction assays to verify proper complex formation

    • FtsK-mediated activation assays to confirm regulatory responsiveness

  • Specificity controls:

    • Testing with non-specific DNA sequences to confirm binding specificity

    • Comparative analysis with known active recombinases as positive controls

    • Site-directed mutants of catalytic residues as negative controls

    • Assessment of activity across a range of buffer conditions

  • Stability parameters:

    • Freeze-thaw stability testing to establish optimal storage conditions

    • Long-term storage stability at different temperatures

    • Thermal stability at reaction temperatures

    • Compatibility with common assay components and additives

Table 3: Recommended Quality Control Tests for Recombinant XerD

Quality ParameterMethodAcceptance CriteriaFrequency
PuritySDS-PAGE>95%Each purification batch
IdentityMass spectrometryMatches theoretical massEach expression batch
Structural integrityCircular dichroismConsistent with predicted secondary structureNew constructs and protocols
DNA bindingEMSA with dif siteSpecific binding with Kd in nM rangeEach purification batch
Catalytic activityDNA cleavage assayDetectable site-specific cleavageEach purification batch
StabilityActivity retention after storage>80% activity after 1 monthQuarterly

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.