ldhA E. coli

Fermentative D-lactate Dehydrogenase, NAD-Dependent E.Coli Recombinant
Shipped with Ice Packs
In Stock

Description

Gene Structure and Regulatory Mechanisms

The ldhA gene in E. coli is regulated by pH, carbon sources, and metabolic intermediates. Key regulatory factors include:

FactorRole in RegulationExperimental Evidence
Low pHInduces transcription via acid-responsive promoters.Transcriptional and translational fusions show 5–10-fold induction at acidic pH .
Anaerobic ConditionsEssential for activation; oxygen suppresses expression.Expression abolished under aerobic conditions .
ArcBModulates expression (3–5-fold reduction in mutants).Disruption of arcB reduces ldhA activity .
MlcRepresses in the absence of glucose; disruption abolishes expression.mlc mutants show near-complete loss of ldhA expression .
CsrAModulates post-transcriptional regulation (moderate reduction in mutants).csrA mutants reduce expression of ldhA fusions .

Enzyme Characteristics and Production

The recombinant ldhA protein (D-lactate dehydrogenase) is expressed in E. coli with specific biochemical properties:

PropertyDetailReference
Molecular Mass39.1 kDa (353 amino acids, including a 24-amino acid His-tag)
Purity>95% (SDS-PAGE)
Activity>200 units/mg (1 unit = 1 µmol pyruvate to D-lactate/min at pH 7.5, 37°C)
Optimal ConditionsNADH-dependent, cytochrome c as electron acceptor

Production Challenges: Overexpression of ldhA in high-copy plasmids leads to poor growth due to pyruvate depletion, mitigated by supplementing alanine or pyruvate .

Role in Metabolism and Cellular Function

ldhA is central to E. coli’s anaerobic fermentation, particularly under acid stress:

Metabolic ContextFunctionConsequence
Low pH, AnaerobicConverts pyruvate to D-lactate, regenerating NAD⁺.Prevents redox imbalance; allows growth in acidic environments .
Pyruvate Pool ManagementExcess LDH activity depletes pyruvate, limiting downstream metabolites.Growth defects in minimal media; rescued by pyruvate supplementation .

Persister Cell Formation and SOS Response

ldhA overexpression induces persister cells via an unconventional mechanism:

MechanismDetailsImplications
SOS Response ActivationDNA damage (e.g., from fluoroquinolones) triggers SOS, consuming ATP.Persister cells enter dormancy, surviving antibiotic stress .
Nucleotide MetabolismDe novo purine biosynthesis upregulated to repair DNA.Maintains viability for regrowth post-antibiotic removal .

Post-Translational Modifications and Activity Regulation

Lysine acetylation modulates ldhA activity and lactate production:

Acetylation SiteEffect on ActivityIndustrial Relevance
K9 (K9R mutant)Increases lactate titer by 1.74-foldEnhanced D-lactate yield in fermentation .
K154 and K248Inhibits lactate accumulationRedirects flux to 3-hydroxypropionate (3HP) .

Applications in Industrial Biotechnology

ldhA engineering enables tailored metabolic pathways:

ApplicationStrategyOutcome
D-Lactate ProductionOverexpress ldhA in pflB mutants to block alternative pathways.High-yield D-lactate fermentation .
L-Lactate SynthesisReplace ldhA with heterologous L-LDH genes (e.g., ldhL from Pediococcus).Converts pyruvate to L-lactate for bioplastic precursors .

Table 1: Regulatory Mutants and Their Impact on ldhA Expression

MutationldhA ExpressionObserved Effect
arcB::Kan↓ 3–5-foldReduced transcriptional and translational activity
mlc::Kan→ Near-abolishedLoss of pH-dependent induction
csrA::Kan↓ ModerateAltered post-transcriptional regulation

Table 2: Acetylation-Site Mutants and Their Effects

MutantLactate Titer3HP ProductionMechanism
K9R↑ 1.74×N/AEnhanced enzyme stability/activity .
K154Q-K248QRedirects carbon flux to 3HP .

Product Specs

Introduction
D-lactate dehydrogenase (ldha) is an enzyme that belongs to the D-isomer specific 2-hydroxyacid dehydrogenase family. It catalyzes the conversion of (D)-lactate to pyruvate using ferricytochrome c as an electron acceptor. In this reaction, ferricytochrome c is reduced to ferrocytochrome c.
Description
Recombinant ldhA from E. coli is a single, non-glycosylated polypeptide chain consisting of 353 amino acids (residues 1-329). It has a molecular weight of 39.1 kDa. This ldhA protein is expressed with a 24 amino acid His-tag at the N-terminus and purified using proprietary chromatographic methods.
Physical Appearance
Clear, colorless solution that has been sterilized by filtration.
Formulation
The ldhA protein is supplied in a solution with a concentration of 1mg/ml. The solution contains 20mM Tris-HCl buffer at pH 8.0, 10% glycerol, and 100mM NaCl.
Stability
For short-term storage (up to 4 weeks), the ldhA solution can be stored at 4°C. For long-term storage, it is recommended to store the solution at -20°C. Adding a carrier protein like 0.1% HSA or BSA is recommended for long-term storage. Avoid repeated cycles of freezing and thawing.
Purity
The purity of the ldhA protein is greater than 95% as determined by SDS-PAGE analysis.
Synonyms
D-lactate dehydrogenase, D-LDH, Fermentative lactate dehydrogenase, ldhA, hslI, htpH, b1380, JW1375.
Source
E.coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSHMKLAVY STKQYDKKYL QQVNESFGFE LEFFDFLLTE KTAKTANGCE AVCIFVNDDG SRPVLEELKK HGVKYIALRC AGFNNVDLDA AKELGLKVVR VPAYDPEAVA EHAIGMMMTL NRRIHRAYQR TRDANFSLEG LTGFTMYGKT AGVIGTGKIG VAMLRILKGF GMRLLAFDPY PSAAALELGV EYVDLPTLFS ESDVISLHCP LTPENYHLLN EAAFEQMKNG VMIVNTSRGA LIDSQAAIEA LKNQKIGSLG MDVYENERDL FFEDKSNDVI QDDVFRRLSA CHNVLFTGHQ AFLTAEALTS ISQTTLQNLS NLEKGETCPN ELV.

Q&A

What is the ldhA gene in E. coli and what is its primary function?

The ldhA gene in Escherichia coli encodes the fermentative D-lactate dehydrogenase (D-LDH), an NADH-linked enzyme that catalyzes the conversion of pyruvate to D-lactate under anaerobic conditions. This enzyme plays a crucial role in the microorganism's ability to maintain redox balance during fermentative growth, particularly when oxygen is limited. The enzyme is notably more active under low pH conditions, showing approximately 10-fold increased expression compared to neutral pH environments. Unlike many other bacterial species that produce L-lactate, E. coli's native LdhA specifically produces the D-isomer of lactate . The genetic sequence analysis reveals that ldhA in E. coli shares high homology with other D-lactate-specific dehydrogenases but shows no significant sequence similarity to L-lactate-specific enzymes . This fundamental difference in substrate stereoselectivity has important implications for both the biochemical function of the enzyme and potential biotechnological applications.

How can researchers experimentally confirm ldhA function in E. coli strains?

To experimentally verify ldhA function in E. coli, researchers typically employ gene disruption techniques followed by phenotypic analysis. A methodological approach involves inserting an antibiotic resistance cassette (such as a kanamycin resistance gene) into the unique KpnI restriction site within the ldhA coding region, creating an ldhA::Kan construct. When this disrupted gene is transferred to the chromosome via homologous recombination, it completely abolishes D-LDH synthesis, which can be confirmed through enzyme activity assays . Another confirmatory method involves measuring lactate production using HPLC or enzymatic assays under anaerobic conditions, comparing wild-type strains with ldhA knockout mutants. Researchers can further validate gene function through complementation studies, where reintroducing the functional ldhA gene on a plasmid restores lactate production in knockout strains. These methodological approaches are essential for definitively establishing the relationship between the ldhA gene and D-lactate production in experimental systems.

What are the standard laboratory conditions for studying ldhA expression in E. coli?

When investigating ldhA expression in E. coli, researchers should consider several critical experimental parameters. The gene shows differential expression depending on oxygen availability, with maximal expression under anaerobic conditions. Temperature typically remains at 37°C for optimal E. coli growth, while pH significantly impacts expression levels. For studying pH-dependent regulation, researchers often use controlled bioreactors or buffered media systems that maintain specific pH levels (typically testing the range of pH 5.0-7.5) to observe the approximately 10-fold increase in ldhA expression at lower pH values . Media composition also influences expression patterns - minimal media versus complex media yield different results, with cells expressing high levels of D-LDH growing particularly poorly in minimal media unless supplemented with alanine or pyruvate . To monitor ldhA expression, researchers commonly use gene fusion constructs such as ldhA-cat (chloramphenicol acetyltransferase) or fluorescent protein reporters, coupled with enzyme activity assays that measure the NADH-dependent reduction of pyruvate to D-lactate spectrophotometrically at 340nm. These standardized conditions facilitate reproducible results when examining ldhA regulation and function.

How is ldhA expression regulated in E. coli?

The regulation of ldhA expression in E. coli involves complex mechanisms responsive to environmental conditions. The gene demonstrates substantial pH-dependent regulation, with expression increasing approximately 10-fold under low pH conditions compared to neutral pH . This adaptation likely helps E. coli maintain cytoplasmic pH homeostasis during fermentative growth when organic acids accumulate. Research using ldhA-cat gene fusion constructs has identified potential regulatory elements, with some mutants showing altered pH-dependent regulation. Importantly, mutations in the pta gene (encoding phosphotransacetylase) affect ldhA regulation, suggesting that acetyl phosphate may serve as a signaling molecule in this regulatory pathway . Additionally, when the ldhA gene is present in high copy numbers, it escapes negative regulation, resulting in overexpression, which indicates the presence of repressor elements that control native expression levels . The regulatory complexity extends to carbon source availability, as demonstrated by glucose supplementation significantly altering acetylation patterns of the LdhA protein, which in turn affects enzyme activity . This intricate regulatory network ensures appropriate expression of ldhA in response to changing environmental conditions, particularly oxygen availability, pH, and carbon source.

What methods are most effective for manipulating ldhA expression for research purposes?

Several methodological approaches have proven effective for manipulating ldhA expression in E. coli research contexts. For overexpression studies, cloning the ldhA gene into multicopy vectors with strong promoters (such as T7 or tac) allows for significant upregulation, though researchers should note that extreme overexpression can negatively impact cell growth . Site-directed mutagenesis following Stratagene protocols enables targeted modification of specific residues, particularly useful when investigating the functional importance of lysine residues subject to acetylation (K9, K70, K154, and K248) . For chromosomal manipulation, P1 vir-mediated transduction using donor strains from collections like the Keio collection facilitates precise gene knockouts . In situ gene replacement using suicide plasmid-mediated homologous recombination (such as with pRE112) allows for introducing specific mutations directly into the chromosomal ldhA locus . For studying regulatory mechanisms, construction of reporter gene fusions (ldhA-cat, ldhA-lacZ, or ldhA-gfp) permits quantitative measurement of gene expression under various conditions. When precise control is required, CRISPR-Cas9 methodologies offer advantages for creating scarless mutations or deletions. These approaches provide researchers with a comprehensive toolkit for manipulating ldhA expression across various experimental objectives.

What is known about the interplay between ldhA and other metabolic genes in E. coli?

The ldhA gene in E. coli operates within an interconnected metabolic network, particularly within central carbon metabolism and fermentation pathways. Research has identified significant interactions with several key metabolic genes. One important relationship exists with the pta gene encoding phosphotransacetylase, as mutations in pta alter ldhA regulation, suggesting acetyl phosphate's role as a potential signaling molecule in the regulatory pathway . The findings indicate metabolic cross-talk between acetate and lactate fermentation pathways. Multiple gene deletion studies have demonstrated functional relationships between ldhA and other fermentation genes including focA-pflB (encoding pyruvate formate-lyase), frdBC (encoding fumarate reductase), adhE (encoding alcohol dehydrogenase), and ackA (encoding acetate kinase) . These deletions redirect carbon flux through remaining pathways, indicating significant metabolic plasticity. During anaerobic growth, the absence of functional pyruvate formate-lyase (pflB) and lactate dehydrogenase (ldhA) severely impairs E. coli's ability to metabolize glucose in mineral media without an alternative NADH oxidation pathway . This interdependence highlights the essential nature of redox balance maintenance in anaerobic metabolism. Understanding these metabolic interactions provides insights into both fundamental bacterial physiology and enables strategic pathway engineering for biotechnological applications.

What is known about the structural characteristics of E. coli LdhA and how do they relate to its function?

E. coli LdhA functions as a homotetramer with allosteric properties, demonstrating positive cooperativity in substrate binding . Each monomer consists of two distinct domains: a catalytic domain and an NAD-binding domain, connected by two linker regions . The enzyme belongs to the D-isomer-specific 2-hydroxyacid dehydrogenase family, in which homologous members adopt a closed conformation when their catalytic domains bind substrates or substrate analogs . While the complete complex structure of E. coli LdhA with pyruvate and NADH has not been definitively resolved, structural modeling based on the highly homologous D-lactate dehydrogenase from Fusobacterium nucleatum (sharing 47% sequence identity) provides insights into the likely conformational arrangement. This homology modeling approach is appropriate because both enzymes demonstrate similar positive cooperativity in substrate binding . The understanding of LdhA's quaternary structure helps explain how subtle modifications—such as lysine acetylation—can significantly impact enzyme activity through allosteric effects and conformational changes that propagate through the tetramer. These structural insights prove valuable for interpreting experimental results and guiding site-directed mutagenesis studies targeting specific functional domains or interfaces within the enzyme complex.

How can E. coli ldhA deletion or modification be utilized for metabolic engineering purposes?

The manipulation of the ldhA gene in E. coli offers valuable strategies for metabolic engineering applications. Complete deletion of ldhA eliminates D-lactate production, redirecting carbon flux through alternative pathways and potentially increasing yields of target compounds that compete with lactate for pyruvate as a precursor. This approach has been successfully employed in metabolic engineering projects targeting products like 3-hydroxypropionate (3HP), where an engineered LdhA variant (K154Q-K248Q) inhibited lactate accumulation and improved 3HP production . Another sophisticated approach involves replacing the native D-lactate-producing ldhA with heterologous genes encoding L-lactate dehydrogenases, such as the ldhL gene from Pediococcus acidilactici. This substitution transforms E. coli from a D-lactate producer to an L-lactate producer, as demonstrated in strain SZ85, which achieved 93-95% yield of L-lactic acid with 98% purity and greater than 99% optical purity . Methodologically, these strain developments require precise genetic techniques including chromosomal gene replacements via homologous recombination and careful screening for improved variants. The SZ85 strain, for example, emerged from selecting for improved growth after the initial chromosomal replacement, resulting in mutations that increased L-LDH activity 30-fold . These approaches demonstrate how fundamental knowledge of ldhA can be translated into valuable biotechnological applications.

What role does LdhA play in E. coli adaptation to the inflammatory gut environment?

The role of lactate metabolism in E. coli adaptation to inflammatory gut environments represents an emerging research area with significant implications for understanding host-microbe interactions. During intestinal inflammation, the metabolic landscape of the gut undergoes profound changes, including increased lactate levels in the gut lumen . This lactate is derived in part from colonic epithelial cells, which modify their metabolism during inflammation. E. coli, a representative member of the Enterobacteriaceae family that expands during inflammation-associated dysbiosis, can utilize this increased lactate through respiratory pathways, particularly via the L-lactate dehydrogenase LldD . This capacity to utilize lactate as an alternative carbon source during inflammation contributes to E. coli fitness in the inflamed gut environment. Research methodologies to study this phenomenon include animal models of both infectious and non-infectious colitis, comparative metagenomic sequencing to assess changes in the gut microbiota's coding capacity, and genetic approaches using bacterial mutants lacking specific lactate utilization pathways. These studies reveal that bacterial genes encoding putative dehydrogenases are enriched during colitis, and that bacterial mutants lacking L-lactate dehydrogenase activity show decreased fitness in the inflamed gut . These findings highlight the complex interplay between host inflammatory responses, metabolic alterations, and bacterial adaptations that collectively shape the gut microbiome during disease states.

What techniques can be used to study ldhA transcriptional regulation under different environmental conditions?

Investigating ldhA transcriptional regulation under varying environmental conditions requires a comprehensive methodological toolkit. RNA-based approaches form the foundation, with quantitative reverse transcription PCR (qRT-PCR) serving as the standard method for quantifying ldhA transcript levels under different conditions, including varying pH, oxygen availability, and carbon sources. For genome-wide perspectives, RNA-seq provides transcriptome-wide insights into how ldhA expression fits within broader transcriptional networks. Promoter fusion constructs represent another powerful approach, where the ldhA promoter region is fused to reporter genes such as lacZ (β-galactosidase), cat (chloramphenicol acetyltransferase), or gfp (green fluorescent protein), enabling quantitative measurement of promoter activity through colorimetric, fluorometric, or luminescence assays . To identify specific transcription factors involved in regulation, techniques such as electrophoretic mobility shift assays (EMSA), DNase footprinting, and chromatin immunoprecipitation (ChIP) can be employed. For studying pH-dependent regulation specifically, researchers should establish precise pH control systems using appropriate buffering systems and monitor pH throughout experiments. The construction of regulatory mutants, as demonstrated in studies isolating mutants that no longer show pH-dependent regulation of ldhA, provides valuable insights into regulatory mechanisms . These comprehensive approaches collectively enable detailed characterization of the complex regulatory networks controlling ldhA expression across diverse environmental conditions.

What are the current limitations in understanding ldhA function and regulation in E. coli?

Despite significant research progress, several key limitations remain in our understanding of ldhA function and regulation in E. coli. One major gap concerns the complete elucidation of the complex regulatory network controlling ldhA expression. While pH-dependence and potential involvement of acetyl phosphate have been identified , the precise transcriptional regulators, promoter elements, and signaling cascades remain incompletely characterized. The complete three-dimensional structure of E. coli LdhA in complex with its substrates and cofactors has not been definitively resolved, necessitating reliance on homology modeling based on related enzymes . This structural uncertainty limits our understanding of the precise molecular mechanisms underlying allosteric regulation and substrate binding. The physiological significance of lysine acetylation on LdhA under different natural growth conditions remains poorly understood, as most acetylation studies have been conducted under laboratory conditions that may not fully reflect natural environments . The evolutionary advantages of producing D-lactate rather than L-lactate (as produced by many other bacteria) remain speculative. Additionally, the comprehensive metabolic consequences of ldhA deletion or modification beyond the immediately connected pathways have not been systematically characterized using global metabolomics approaches. Addressing these knowledge gaps will require integrated approaches combining structural biology, systems biology, evolutionary analysis, and physiological studies under more naturalistic conditions.

What new methodologies are emerging for studying ldhA and lactate metabolism in E. coli?

Emerging methodologies are expanding our capacity to investigate ldhA and lactate metabolism in E. coli with unprecedented precision and comprehensiveness. CRISPR-Cas9 genome editing technologies now permit rapid, scarless modification of ldhA and related genes, enabling more sophisticated genetic manipulations than traditional methods. Metabolic flux analysis using 13C-labeled substrates coupled with mass spectrometry provides quantitative insights into how carbon flows through lactate pathways under different conditions, revealing redirections in metabolic flux following ldhA manipulation. Advanced protein structural analysis techniques, including cryo-electron microscopy and time-resolved X-ray crystallography, offer opportunities to visualize LdhA conformational changes during catalysis and regulatory events. Single-cell technologies such as microfluidics combined with fluorescent reporters allow researchers to observe ldhA expression dynamics at the individual cell level, revealing population heterogeneity that may be masked in bulk measurements. Multi-omics approaches integrating transcriptomics, proteomics, metabolomics, and fluxomics provide systems-level understanding of how ldhA functions within broader metabolic networks. Protein acetylation can now be studied more comprehensively using acetylomics approaches that quantify acetylation patterns across the proteome . Miniaturized bioreactors with precise environmental control enable high-throughput screening of ldhA function under diverse conditions. Collectively, these advanced methodologies promise to address existing knowledge gaps and reveal new dimensions of ldhA biology and lactate metabolism in E. coli.

Table 2: Acetylated Lysine Sites in E. coli LdhA and Their Effects

Lysine ResidueEffect of Acetylation on Enzyme ActivityLocation in Protein StructureMethodological Evidence
K9Mutation to mimic non-acetylated state (K9R) improved lactate production 1.74-foldN-terminal regionMass spectrometry; site-directed mutagenesis; fermentation analysis
K70Identified as acetylation siteWithin protein structureMass spectrometry detection
K154When combined with K248Q mutation, inhibited lactate accumulationPart of catalytic domainSite-directed mutagenesis; production analysis
K248When combined with K154Q mutation, inhibited lactate accumulationNear active siteSite-directed mutagenesis; production analysis

Product Science Overview

Role and Mechanism

In the absence of oxygen, E. coli relies on fermentative pathways to generate energy. One of the key pathways involves the reduction of pyruvate to lactate by lactate dehydrogenase. The NAD-dependent D-lactate dehydrogenase specifically catalyzes the conversion of pyruvate to D-lactate, using NADH as a cofactor, which is oxidized to NAD+ in the process. This reaction is vital for maintaining the redox balance within the cell and ensuring a continuous supply of NAD+ for glycolysis.

Recombinant Production

The recombinant production of D-lactate dehydrogenase in E. coli involves the insertion of the gene encoding this enzyme into the bacterial genome or a plasmid vector. This genetic modification allows for the overexpression of the enzyme, facilitating its study and potential industrial applications. Recombinant E. coli strains engineered to overproduce D-LDH can be used in various biotechnological processes, including the production of D-lactic acid, which is a valuable chemical in the food, pharmaceutical, and polymer industries .

Applications and Significance
  1. Industrial Production: D-lactic acid produced by recombinant E. coli is used in the synthesis of biodegradable plastics, such as polylactic acid (PLA). PLA is an environmentally friendly alternative to conventional plastics derived from petrochemicals.
  2. Biochemical Research: The study of D-LDH and its recombinant forms provides insights into the metabolic pathways of E. coli and other bacteria. This knowledge can be applied to metabolic engineering efforts aimed at optimizing the production of various biochemicals.
  3. Medical and Pharmaceutical: D-lactic acid has potential applications in the medical field, including its use as a precursor for the synthesis of certain pharmaceuticals.
Challenges and Future Directions

While the recombinant production of D-LDH in E. coli has shown promise, there are several challenges that need to be addressed:

  • Metabolic Burden: Overexpression of recombinant proteins can impose a metabolic burden on the host cells, potentially affecting their growth and productivity.
  • Optimization: Further optimization of the fermentation conditions and genetic constructs is necessary to maximize the yield and efficiency of D-lactic acid production.
  • Scale-Up: Translating laboratory-scale processes to industrial-scale production requires careful consideration of various factors, including cost, scalability, and regulatory compliance.

Future research efforts are focused on addressing these challenges and exploring new applications for D-lactic acid and other products derived from recombinant E. coli strains. Advances in metabolic engineering, synthetic biology, and fermentation technology will play a crucial role in realizing the full potential of these biotechnological innovations .

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 2024 Thebiotek. All Rights Reserved.