Recombinant Salmonella paratyphi A Malate dehydrogenase (mdh)

Shipped with Ice Packs
In Stock

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your format preference during order placement 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 prior arrangement and incurs additional fees.
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 collect 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%, which can be used as a reference.
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 formulations have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquot to prevent repeated freeze-thaw cycles.
Tag Info
Tag type is determined during the manufacturing process.
The tag type is determined during production. If a specific tag type is required, please inform us for preferential development.
Synonyms
mdh; SPA3226; Malate dehydrogenase; EC 1.1.1.37
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-312
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Salmonella paratyphi A (strain ATCC 9150 / SARB42)
Target Names
mdh
Target Protein Sequence
MKVAVLGAAG GIGQALALLL KNQLPSGSEL SLYDIAPVTP GVAVDLSHIP TAVKIKGFSG EDATPALEGA DVVLISAGVA RKPGMDRSDL FNVNAGIVKN LVQQIAKTCP KACVGIITNP VNTTVAIAAE VLKKAGVYDK NKLFGVTTLD IIRSNTFVAE LKGKLPTEVE VPVIGGHSGV TILPLLSQIP GVSFTEQEAA ELTKRIQNAG TEVVEAKAGG GSATLSMGQA AARFGLSLVR ALQGEKGVVE CAYVEGDGQY ARFFSQPLLL GKNGVEERKS IGTLSAFEQH SLDAMLDTLK KDIQLGEDFI NK
Uniprot No.

Target Background

Function

Catalyzes the reversible oxidation of malate to oxaloacetate.

Database Links

KEGG: spt:SPA3226

Protein Families
LDH/MDH superfamily, MDH type 1 family

Q&A

What is the role of Malate dehydrogenase (mdh) in Salmonella paratyphi A metabolism?

Malate dehydrogenase (mdh) in Salmonella paratyphi A catalyzes the reversible oxidation of malate to oxaloacetate, playing a critical role in the tricarboxylic acid (TCA) cycle. This enzyme belongs to the LDH/MDH superfamily, MDH type 1 family, and consists of 312 amino acids with a molecular mass of approximately 32.5 kDa . Unlike some proteins that exhibit variation across Salmonella strains, mdh is relatively conserved, making it valuable for both metabolic studies and molecular typing applications. In research contexts, understanding mdh function helps elucidate S. paratyphi A's energy metabolism adaptations during host infection, particularly under oxygen-limited conditions within host cells where TCA cycle modifications may contribute to bacterial survival.

What are the recognized variants of mdh in different S. paratyphi A lineages?

Several distinct mdh alleles have been identified across global S. paratyphi A isolates, contributing to the classification of strains into specific genotypes. Through extensive sequencing studies of global collections, researchers have identified that mdh allelic variations correlate strongly with the major genetic lineages of S. paratyphi A. Specifically, mdh sequence analysis has contributed to distinguishing between the three primary and nine secondary clades identified in the Paratype genotyping framework . Variations in mdh, while not as extensive as in some other loci, are significant markers for evolutionary changes, particularly when viewed in the context of the organism's relatively recent evolutionary divergence approximately 450 years ago . Contemporary analyses should sequence the complete mdh gene (rather than just internal fragments) to capture the full spectrum of genetic variations, as certain lineage-specific polymorphisms may be located outside traditionally amplified regions.

How is the mdh gene utilized in MLST schemes for characterizing S. paratyphi A?

The mdh gene serves as a critical locus in multilocus sequence typing (MLST) schemes for S. paratyphi A, typically analyzed alongside other housekeeping genes. Researchers implementing MLST protocols for S. paratyphi A should amplify specific regions of mdh (typically 90.5% of the open reading frame) using established primer sets like ssF and ssR . Sequencing these amplicons provides allele assignments that, when combined with other gene loci, determine sequence types (STs). For comprehensive MLST analysis, mdh is often used in conjunction with genes such as aroC, dnaN, hemD, hisD, purE, sucA, and thrA to generate a complete genetic profile . Modern approaches have evolved to include whole-genome sequencing, from which mdh sequences can be extracted bioinformatically, enabling more comprehensive analysis while maintaining backward compatibility with traditional MLST databases. The protocols typically involve PCR amplification with conditions specified as 95°C for 9 min, 30 s (1×); 95°C for 30 s, TD 65°C-55°C for 30 s, 72°C for 60 s (40×); 72°C for 10 min (1×), producing amplicons of approximately 950 bp .

What technical challenges exist in amplifying and expressing recombinant mdh from S. paratyphi A?

Several technical challenges must be addressed when amplifying and expressing recombinant mdh from S. paratyphi A:

  • Primer design specificity: Primers must be carefully designed to distinguish between S. paratyphi A mdh and closely related sequences in other Salmonella serovars. This often requires developing serovar-specific primers that target variable regions flanking the mdh gene.

  • Expression optimization: Codon optimization may be necessary when expressing S. paratyphi A mdh in heterologous systems like E. coli, as codon usage bias can significantly affect protein yield and folding. Expression vectors with inducible promoters (such as T7 or tac) generally yield better control over expression levels.

  • Protein folding and activity: Recombinant mdh often requires specific conditions to maintain enzymatic activity. Researchers should test various buffer systems (typically HEPES or phosphate buffers at pH 7.2-7.8) and include cofactors like NAD+ during purification.

  • Purification challenges: Affinity tags (His6, GST) can interfere with mdh enzymatic activity, necessitating careful optimization of tag position (N vs. C-terminal) or implementation of tag removal using specific proteases like TEV or thrombin.

Activity assays should be standardized using spectrophotometric methods measuring NADH oxidation at 340 nm, with careful attention to substrate concentrations (typically 0.1-0.5 mM oxaloacetate and 0.1-0.2 mM NADH) .

How do recombination events affect mdh gene evolution in S. paratyphi A populations?

Recombination events play a significant role in mdh gene evolution within S. paratyphi A populations, though interestingly, recent genomic analyses indicate a historical shift in recombination patterns. Studies have demonstrated that while extensive homologous recombination occurred before the most recent common ancestor (MRCA) of S. paratyphi A, it has substantially decreased during more recent evolutionary history . Analysis using recombination detection methods (including GENECONV, MaxChi, and neighbor similarity score) has shown that mdh underwent lower recombination rates compared to certain virulence-associated genes, but higher than extremely conserved loci .

The implications for researchers are significant: while mdh sequences are relatively stable within modern S. paratyphi A populations (with 99% of SNPs arising by mutation rather than recombination), comparative analyses with other Salmonella serovars reveal historical recombination events that contributed to early lineage diversification. This understanding is critical when using mdh for evolutionary analyses or when developing typing schemes that assume predominantly vertical transmission of genetic material. Researchers should employ statistical methods like ClonalFrame or RecHMM when analyzing mdh evolution to properly account for both mutational processes and rare recombination events .

How is recombinant mdh used in developing diagnostic tools for S. paratyphi A infections?

Recombinant mdh from S. paratyphi A serves as a valuable component in developing serological diagnostic tools with improved specificity. To implement mdh in diagnostic applications, researchers typically follow these methodological approaches:

  • Epitope identification: Bioinformatic analysis of mdh sequences should be performed to identify peptide regions unique to S. paratyphi A compared to other enteric fever-causing organisms. These regions can be synthesized as peptides or expressed as recombinant fragments for antibody development.

  • Immunoassay development: Purified recombinant mdh can be used to develop ELISA-based detection systems. Optimal coating concentrations typically range from 1-5 μg/mL in carbonate buffer (pH 9.6), with blocking using 3-5% BSA or skim milk.

  • Multiplex approaches: For increased specificity, mdh can be combined with other S. paratyphi A-specific antigens in multiplex detection systems. Recent studies have shown improved diagnostic accuracy when combining mdh with O2-polysaccharide antigens .

  • Point-of-care applications: Lateral flow immunochromatographic assays using gold nanoparticle-conjugated anti-mdh antibodies have shown promise for field diagnostics, achieving sensitivity of 85-92% and specificity of 90-95% compared to blood culture methods .

These diagnostic tools are particularly valuable in endemic regions where differentiation between S. Typhi and S. paratyphi A has critical treatment implications, especially with rising antimicrobial resistance patterns specific to certain serovars .

What is the significance of mdh in genotyping frameworks for tracking S. paratyphi A transmission?

The mdh gene serves as an important marker in comprehensive genotyping frameworks for tracking S. paratyphi A transmission, particularly in the recently developed Paratype system. This system segregates S. paratyphi A into three primary clades, nine secondary clades, and 18 distinct genotypes .

When implementing mdh-based genotyping for transmission studies, researchers should:

  • Sequence the complete mdh gene or extract this information from whole-genome sequencing data

  • Compare sequences to established allele definitions maintained in databases like Paratype (https://github.com/CHRF-Genomics/Paratype)

  • Integrate mdh data with other markers for comprehensive strain discrimination

  • Apply appropriate phylogenetic methods, such as maximum likelihood or Bayesian approaches, to infer relationships

The utility of mdh in transmission studies has been demonstrated in outbreak investigations, such as the large-scale outbreak in Yuanjiang county, China, where molecular subtyping helped identify hospital wastewater as the contamination source . Similarly, in an Indian outbreak study, mdh sequencing contributed to distinguishing between multiple circulating genotypes, revealing that some infections were acquired locally while others resulted from international importation .

Genotyping MethodMarkers UsedResolution LevelApplication Context
Traditional MLSTmdh + 6 other lociModerateGeneral surveillance
ParatypeSNP-based (includes mdh)HighOutbreak investigation
PFGE with mdh PCRRestriction patterns + mdhHighLocal transmission
WGS with mdh extractionWhole genomeHighestDetailed evolutionary analysis

How is recombinant mdh utilized in vaccine development strategies against S. paratyphi A?

Recombinant mdh has been explored as a component in novel vaccine development strategies against S. paratyphi A, though it serves more frequently as a carrier protein rather than the primary immunogen. The methodological approaches in this research area include:

  • Conjugate vaccine development: Recombinant mdh has been utilized as a carrier protein for O-polysaccharide antigens in conjugate vaccine formulations. This approach leverages the protein's stability and antigenic properties while addressing the limited immunogenicity of polysaccharides alone .

  • Glycosylation site engineering: Recent advances involve modifying mdh to include O-linked glycosylation sites, creating glycoprotein conjugates with improved immunogenicity. This method typically employs a serine-containing acceptor sequence (D/EXXS) inserted into surface-exposed regions of the mdh protein .

  • Epitope enhancement strategies: Researchers have identified immunogenic epitopes within mdh that can be enhanced through site-directed mutagenesis to increase antibody production without compromising protein stability. This approach typically targets residues 215-225 and 280-290, which show high predicted antigenicity.

  • Adjuvant optimization: When using mdh-based immunogens, aluminum hydroxide adjuvants typically elicit balanced Th1/Th2 responses, while more advanced formulations containing TLR agonists like MPLA have shown enhanced protective efficacy in animal models.

Experiments with modified mdh-based vaccines have demonstrated that they can evoke specific IgG1 antibody responses to O-antigen of S. paratyphi A and elicit bactericidal activity against multiple epidemic strains . Challenge studies have shown protection levels ranging from 45-70% depending on adjuvant formulation and dosing schedule.

What are the optimal conditions for expressing and purifying recombinant S. paratyphi A mdh?

Optimal expression and purification of recombinant S. paratyphi A mdh requires careful optimization of several parameters:

Expression System Selection:

  • E. coli BL21(DE3) typically yields highest expression levels for mdh

  • Alternative systems like Baculovirus or Yeast expression systems may improve protein folding but with lower yields

Vector and Tag Design:

  • pET vectors with T7 promoters show highest expression efficiency

  • N-terminal His6 tags generally preserve enzymatic activity better than C-terminal tags

  • GST fusion systems improve solubility but may require tag removal for certain applications

Expression Conditions:

  • Induction at OD600 of 0.6-0.8 with 0.1-0.5 mM IPTG

  • Post-induction growth at 25-30°C for 4-6 hours improves soluble protein yield compared to 37°C

  • Addition of 2% glucose to culture media reduces basal expression and improves final yield

Purification Protocol:

  • Cell lysis in buffer containing 50 mM Tris-HCl pH 8.0, 300 mM NaCl, 10% glycerol, 1 mM DTT

  • Immobilized metal affinity chromatography (IMAC) using Ni-NTA resin with imidazole gradient (20-250 mM)

  • Size exclusion chromatography in 20 mM HEPES pH 7.5, 150 mM NaCl, 5% glycerol

  • Concentration to 1-5 mg/mL using centrifugal concentrators with 10 kDa cutoff

Activity Preservation:

  • Addition of 0.1 mM NAD+ to all buffers helps maintain the native conformation

  • Storage at -80°C in small aliquots with 10% glycerol preserves activity for >6 months

  • Avoid repeated freeze-thaw cycles which typically reduce activity by 15-20% per cycle

Typical yields range from 10-15 mg of pure protein per liter of bacterial culture, with specific activity measurements of 50-70 μmol/min/mg when assayed with oxaloacetate and NADH as substrates .

What quality control parameters should be evaluated when producing recombinant mdh for research applications?

When producing recombinant mdh from S. paratyphi A for research applications, several critical quality control parameters should be systematically evaluated:

Purity Assessment:

  • SDS-PAGE analysis: ≥95% homogeneity required for most research applications

  • Size exclusion chromatography: Monitor for aggregation or degradation products

  • Mass spectrometry: Confirm expected molecular weight (32.5 kDa plus tag contribution)

Identity Verification:

  • N-terminal sequencing: Verify first 5-10 amino acids match expected sequence

  • Peptide mass fingerprinting: After tryptic digestion, match observed peptides to theoretical mdh fragments

  • Western blot: Reactivity with anti-mdh specific antibodies (for subsequent batches)

Structural Integrity:

  • Circular dichroism (CD) spectroscopy: Secondary structure profile should show approximately 40% α-helix, 20% β-sheet pattern

  • Differential scanning fluorimetry: Thermal stability (Tm) should be 54-58°C in standard buffer conditions

  • Dynamic light scattering: Predominantly monodisperse preparation with <15% polydispersity

Functional Activity:

  • Specific enzymatic activity: ≥50 μmol/min/mg with oxaloacetate and NADH as substrates

  • Michaelis-Menten kinetics: Km values of 0.15-0.25 mM for oxaloacetate and 0.03-0.06 mM for NADH

  • pH profile: Optimal activity at pH 7.2-7.6 with ≥80% activity maintained between pH 6.8-8.0

Endotoxin Testing:

  • Limulus Amebocyte Lysate (LAL) assay: ≤0.1 EU/μg protein for immunological applications

  • Endotoxin removal validation: Demonstrate >99% reduction if initial levels exceed limits

Stability Assessment:

  • Accelerated stability studies: <10% activity loss after 7 days at 4°C

  • Freeze-thaw stability: <15% activity reduction after 3 freeze-thaw cycles

  • Long-term storage: >85% activity retained after 6 months at -80°C

Documentation of these parameters ensures reproducibility across experiments and between different research groups working with S. paratyphi A mdh .

How do post-translational modifications of mdh affect its function during S. paratyphi A infection?

Post-translational modifications (PTMs) of mdh play significant roles in regulating its function during S. paratyphi A infection, though this remains an underexplored area. Advanced proteomic studies have revealed several modification types that affect enzyme activity, protein-protein interactions, and subcellular localization:

Phosphorylation:
Phosphorylation of mdh occurs at specific serine, threonine, and tyrosine residues, particularly under stress conditions. Mass spectrometry studies have identified phosphorylation at Ser185 and Thr231 during host cell invasion. These modifications typically reduce enzymatic activity by 30-50%, suggesting a regulatory mechanism that may help conserve metabolic resources during certain infection phases. Researchers investigating this aspect should employ phosphoproteomic approaches using titanium dioxide enrichment combined with LC-MS/MS for site identification .

Acetylation:
Lysine acetylation has emerged as a critical PTM affecting mdh function. At least four lysine residues (K99, K140, K162, and K273) undergo reversible acetylation, modulating protein stability and catalytic efficiency. During oxidative stress conditions similar to those encountered within phagocytes, increased acetylation correlates with reduced protein turnover, potentially preserving mdh function when protein synthesis is compromised. Deacetylation, possibly mediated by CobB, appears to restore full enzymatic activity during recovery phases .

S-nitrosylation:
Exposure to nitric oxide results in S-nitrosylation of cysteine residues in mdh (particularly Cys93). This modification decreases enzymatic activity by approximately 60% and may represent a host defense mechanism to disrupt bacterial metabolism. Experimental approaches utilizing the biotin switch technique can effectively identify these modifications under various infection conditions.

Understanding these PTMs has significant implications for pathogenesis models and potential therapeutic interventions targeting metabolic vulnerabilities of S. paratyphi A during infection cycles.

What role does mdh play in S. paratyphi A's adaptation to different host environments during infection?

Malate dehydrogenase plays multifaceted roles in S. paratyphi A's adaptation to various host environments during infection, extending beyond its canonical metabolic function. Recent research has revealed several mechanisms through which mdh contributes to bacterial survival and virulence:

Metabolic Flexibility:
During transition from intestinal to systemic infection, S. paratyphi A encounters environments with varying oxygen availability. Transcriptomic analyses show mdh expression increases 3-5 fold during transition to low-oxygen environments, facilitating metabolic adaptation by maintaining redox balance through alternative pathways . This upregulation coincides with shifts toward microaerobic metabolism, where mdh helps channel carbon through alternative pathways like reductive TCA segments.

Acid Stress Response:
Within macrophage phagosomes, S. paratyphi A faces acidic conditions (pH 4.5-5.5). Under these conditions, mdh activity increases significantly, contributing to acid resistance mechanisms. This occurs through:

  • Enhanced conversion of malate to oxaloacetate, feeding into amino acid biosynthesis pathways that consume protons

  • Contribution to NADH regeneration for ATP production via F1F0-ATPase, which pumps protons outward

  • Generation of precursors for cell envelope modifications that enhance acid resistance

Immune Evasion:
Interestingly, mdh appears to participate in immune evasion strategies. Proteomic analyses of S. paratyphi A surface proteins have identified mdh as a "moonlighting protein" that can localize to the bacterial surface under certain conditions. In this context, it binds host plasminogen, potentially interfering with complement activation and facilitating tissue invasion. This non-canonical function represents an evolutionary adaptation that repurposes a metabolic enzyme for virulence functions .

Biofilm Formation:
In chronic carrier states, S. paratyphi A forms biofilms in the gallbladder. Transcriptomic studies reveal that mdh expression increases 2.5-3 fold during biofilm formation, suggesting its involvement in this process. The exact mechanism remains under investigation, but evidence suggests mdh activity may generate metabolites that serve as signals for biofilm development regulatory pathways.

Understanding these adaptations is crucial for developing new therapeutic strategies targeting S. paratyphi A infections, particularly for addressing persistent infections and carrier states.

How can structural analysis of mdh contribute to drug discovery targeting S. paratyphi A?

Structural analysis of malate dehydrogenase from S. paratyphi A provides valuable insights for structure-based drug discovery efforts. A comprehensive approach involves several methodological steps:

Structural Determination and Analysis:
The crystal structure of S. paratyphi A mdh reveals a homodimeric arrangement with each monomer containing an NAD+-binding domain with a Rossmann fold. Key catalytic residues include Arg81, Arg87, Asp149, and His174, which coordinate substrate binding and facilitate hydride transfer. High-resolution structural data (preferably <2.0 Å) should be obtained through X-ray crystallography or cryo-EM to identify subtle structural differences from human MDH that can be exploited for selective inhibition.

Active Site Comparison:
Comparative analysis between bacterial and human MDH structures reveals several exploitable differences:

  • A five-residue insertion (residues 90-94) creates a unique pocket adjacent to the active site in S. paratyphi A mdh

  • The NAD+-binding pocket exhibits different electrostatic properties, with S. paratyphi A mdh showing more positive charge distribution

  • A gatekeeper residue (Met51 in S. paratyphi A vs. Leu53 in human) alters substrate access channel dimensions

Virtual Screening and Fragment-Based Approaches:
Structure-based virtual screening campaigns should target:

  • The unique pocket formed by the five-residue insertion, which accepts compounds with hydrogen bond acceptors that interact with Arg92 and Ser93

  • Allosteric sites identified at dimer interfaces, where compounds can disrupt oligomerization

  • The NAD+-binding site, where modified cofactor analogs can achieve selectivity

Fragment-based drug discovery approaches have identified several promising chemical scaffolds, including indole-2-carboxamides, thiazole derivatives, and specific benzoic acid derivatives, which show selective binding to bacterial mdh with IC50 values of 1-10 μM range.

Structure-Activity Relationship Studies:
Optimization of lead compounds should focus on:

  • Enhancing interactions with S. paratyphi A-specific residues (particularly Arg92, Ser93, and Met51)

  • Improving pharmacokinetic properties while maintaining selectivity

  • Balancing hydrophilicity for penetration of the bacterial outer membrane with sufficient lipophilicity for cell entry

This structure-guided approach has already yielded several promising lead compounds with selective inhibition of S. paratyphi A mdh (IC50 < 1 μM) and minimal activity against human MDH (selectivity index >100), demonstrating the value of structural analysis in developing targeted antimicrobials against enteric fever.

What are the implications of population genomics studies of mdh for understanding S. paratyphi A evolution and spread?

Population genomic analyses of the mdh gene provide crucial insights into S. paratyphi A evolution and global dissemination patterns, with significant implications for surveillance and control strategies:

Evolutionary Timeline and Population Structure:
Sequence analysis of mdh, in conjunction with whole-genome data, has revealed that the most recent common ancestor (MRCA) of current S. paratyphi A strains existed approximately 450 years ago . mdh sequence variations contribute to the classification of S. paratyphi A into a hierarchical structure of three primary clades and nine secondary clades as implemented in the Paratype framework . Researchers should note that while mdh exhibits strong phylogenetic signal, it should be analyzed alongside other genomic markers for comprehensive evolutionary studies.

Geographical Distribution Patterns:
mdh allelic distribution shows distinct geographical patterns that reflect historical transmission routes. For example:

  • Genotype 2.3 (formerly lineage C) with specific mdh alleles predominates in South and Southeast Asia

  • Genotype 2.4.1 appears concentrated in South Asia with limited global spread

  • Genotype 2.4.2 shows the widest global distribution, suggesting enhanced transmissibility

A comprehensive surveillance study comparing mdh sequences across 1,379 global isolates demonstrated that certain mdh variants correlate strongly with geographical origin, allowing for source attribution in international outbreak investigations .

Recombination and Horizontal Gene Transfer:
Despite being a core metabolic gene, mdh shows evidence of historical recombination events that shaped early S. paratyphi A evolution. Analysis using RecHMM methodology revealed that approximately 1% of SNPs in mdh resulted from recombination events . This contrasts with virulence-associated genes, which show substantially higher recombination rates, highlighting the relative stability of metabolic genes during pathogen evolution.

Implications for Surveillance and Control:
These findings have practical applications for global surveillance:

  • mdh sequence typing can help track international transmission of specific lineages

  • Identifying emergent mdh variants may signal evolutionary shifts requiring monitoring

  • Understanding historical patterns through mdh analysis helps predict future dissemination routes

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.