Recombinant Photorhabdus luminescens subsp. laumondii DnaA-homolog protein hda (hda)

Shipped with Ice Packs
In Stock

Description

Introduction

Photorhabdus luminescens is a bacterium known for its pathogenic effects on insects and its symbiotic relationships with nematodes of the Heterorhabditis genus . As an insect pathogen, P. luminescens releases toxins into the insect blood, effectively converting the insect into a resource that supports the growth and development of nematodes . The P. luminescens subsp. laumondii strain HP88 has a genome of 5.27-Mbp with a 42.4% G+C content, encoding 4,243 potential proteins .

Functional Overview of DnaA-Homolog Protein Hda

The DnaA-homolog protein Hda is involved in DNA replication and regulation. It shares homology with DnaA, a protein crucial for the initiation of DNA replication in bacteria. Understanding the role of Hda in P. luminescens subsp. laumondii may provide insights into bacterial replication mechanisms and potential targets for antibacterial strategies.

Hda and Bacterial Interactions

P. luminescens interacts with nematodes during transmission, which starts with the bacteria adhering to the rectal gland cells (RGC) in the nematode gut . These bacteria then replicate within the RGC, causing vacuolization and eventual lysis, which releases P. luminescens throughout the nematode's body cavity to colonize developing nematodes .

HdfR Regulator and its Impact on Transmission

The LysR-type regulator, HdfR, is essential for proper transmission in P. luminescens TTO1 . Transcriptome analysis has shown that HdfR regulates 124 genes involved in arginine metabolism, hydroxyphenylacetate catabolism, and pigment production . A mutant lacking HdfR can attach to and grow within the nematode, but later transmission stages are delayed, indicating HdfR's role in coordinating the interaction between P. luminescens and its nematode partner .

Regulation of Natural Product Biosynthesis

Bacterial enhancer-binding proteins (bEBPs) can regulate natural product biosynthesis in P. laumondii . The AAA+ ATPase core of the Sinorhizobium meliloti bEBP DctD can induce the expression of five natural product classes in P. laumondii TTO1 . Additionally, truncated copies of native P. laumondii bEBPs have shown both repression and induction across six natural product classes .

Genome-Wide Analysis of Proteases and Protease Inhibitors

A genome-wide analysis of proteases and protease inhibitors in Haemonchus contortus, a parasitic roundworm, used combined sequence- and structure-based methods to identify and classify these molecules . This approach, which uses well-curated public datasets, emphasizes the need for improved tools to annotate molecules in lesser-known organisms, such as eukaryotic pathogens .

UV-B Radiation Effects on Mirabilis himalaica

Research on Mirabilis himalaica showed that UV-B radiation induces changes in plant metabolism, particularly in the biosynthesis of rotenoids . Comprehensive analysis of protein and transcript data identified differentially expressed genes (DEGs) from plant hormone signal transduction and phosphatidylinositol signaling system pathways . These DEGs are associated with auxin and calcium signaling, suggesting their role in driving the downstream transmission of these signal transduction pathways .

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 purchasing method and location. Please consult your local distributor for precise delivery estimates.
Note: All proteins are shipped with standard blue ice packs unless dry ice shipping is specifically requested and agreed upon in advance. Additional fees apply for dry ice shipping.
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% and may 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 formulations have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquoting is essential for multiple uses. Avoid 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, and we will prioritize its development.
Synonyms
hda; plu2752; DnaA regulatory inactivator Hda
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-233
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Photorhabdus luminescens subsp. laumondii (strain DSM 15139 / CIP 105565 / TT01)
Target Names
hda
Target Protein Sequence
MNTPSQLSLP LYLPDDETFA SFFPGENATL LAAIKLAINQ PHGSYIYFWS RDSGGRSHLL HAACAELSLK DEAVGYVPLD KRAYFIPEVL DGMEHLSLVC IDNIESIAGD EEWEMAIFNL YNRILEIGRT CLLISGDRPP RQINLKLPDL ASRLDWGQIY KLQPLSDDEK LQALQLRAKL RGFELPEDVG RFLLKRLDRE MRTLFMALNQ LDHASIVAQR KLTIPFVKDI LHL
Uniprot No.

Target Background

Function

Function: This protein mediates the interaction between the DNA replication initiator protein DnaA and the DNA polymerase subunit beta sliding clamp (dnaN). It stimulates the hydrolysis of ATP-DnaA to ADP-DnaA, thus inactivating DnaA and preventing reinitiation – a process known as regulatory inhibition of DnaA (RIDA).

Database Links

KEGG: plu:plu2752

STRING: 243265.plu2752

Protein Families
DnaA family, HdA subfamily

Q&A

What experimental systems are commonly used to study the function of Hda protein in P. luminescens?

To study the function of Hda protein in P. luminescens, researchers typically employ several experimental approaches:

  • Gene expression systems: Recombinant expression of hda in expression hosts like yeast or E. coli to produce the protein for biochemical studies .

  • Mutant analysis: Creating mutant strains with alterations in the dnaN gene (e.g., dnaN-G157C) to study how changes in the β clamp affect Hda function and DNA replication .

  • Luciferase reporter assays: These can be used to monitor gene expression changes that result from alterations in DnaA activity regulated by Hda .

  • ATP hydrolysis assays: Biochemical assays to directly measure the rate of ATP hydrolysis by DnaA in the presence and absence of Hda protein.

  • Bioluminescence techniques: Since P. luminescens is naturally bioluminescent, researchers can use bioluminescence to monitor cellular ATP levels, which can be affected by Hda-mediated regulation .

How is recombinant P. luminescens Hda protein typically produced and purified for research?

The recombinant Hda protein from P. luminescens is typically produced and purified using the following methodological approach:

  • Expression system selection: The protein is commonly expressed in yeast systems, which allows for proper folding and potential post-translational modifications .

  • Vector construction: The full-length hda gene (coding for all 233 amino acids) is cloned into an appropriate expression vector, often with an affinity tag to facilitate purification.

  • Expression induction: The recombinant protein expression is induced under optimal conditions for the chosen host system.

  • Cell lysis and initial purification: Cells are lysed, and the lysate is clarified by centrifugation.

  • Affinity chromatography: The protein is purified using affinity chromatography based on the tag attached.

  • Quality control: SDS-PAGE analysis is performed to ensure purity (>85% purity is typically achieved) .

  • Storage: The purified protein is stored with 5-50% glycerol (typically 50%) at -20°C/-80°C to maintain stability. The shelf life is approximately 6 months for liquid form and 12 months for lyophilized form .

  • Reconstitution (if lyophilized): The protein is reconstituted in deionized sterile water to a concentration of 0.1-1.0 mg/mL before use .

How does the DnaA-ATP/DnaA-ADP ratio affect DNA replication in P. luminescens, and what role does Hda play in maintaining this balance?

The DnaA-ATP/DnaA-ADP ratio is a critical regulatory mechanism that controls DNA replication initiation in P. luminescens and other bacteria. This balance is intricately maintained by several factors, with Hda playing a central role:

  • Mechanism of action: Hda promotes the hydrolysis of ATP bound to DnaA, converting active DnaA-ATP to inactive DnaA-ADP. This conversion occurs through a process called RIDA (Regulatory Inactivation of DnaA) .

  • β clamp interaction: Hda's activity requires interaction with the β clamp (encoded by the dnaN gene), which is the replication processivity factor. This interaction is essential for the hydrolysis reaction to occur efficiently .

  • Cellular effects of imbalance: Research with a dnaN-G157C allele demonstrates that alterations in this interaction can lead to an under-replication phenotype due to increased accumulation of DnaA-ADP. This suggests that proper Hda function is required for maintaining the correct DnaA-ATP/DnaA-ADP ratio .

  • Transcriptional regulation: The DnaA-ATP/DnaA-ADP ratio also affects gene expression beyond replication initiation. For example, expression of the iraD gene in related bacteria is elevated when the dnaN-G157C mutation promotes more regulatory inactivation of DnaA, favoring DnaA-ADP over DnaA-ATP .

The methodological approach to studying this balance typically involves creating specific mutations in the dnaN gene or hda gene and observing the effects on replication timing, cell cycle progression, and gene expression patterns.

What is the relationship between Hda protein function and temperature restriction in P. luminescens?

P. luminescens displays notable temperature restrictions in its growth, typically being unable to grow at temperatures above 35°C on solid media . While the direct relationship between Hda protein and temperature restriction has not been fully elucidated, several lines of evidence suggest potential connections:

  • Replication control at different temperatures: Hda's role in regulating DnaA activity might be temperature-dependent, as replication initiation dynamics can vary with temperature.

  • Temperature-sensitive mutants: Studies have isolated mutants of P. luminescens DJC with abilities to grow between 36 and 37°C . Whole genome sequencing of these temperature-tolerant clones might reveal mutations in genes related to replication control, potentially including hda or its interacting partners.

  • TRL operon connection: The temperature restriction locus (TRL) has been identified in P. luminescens, with gene expression showing up-regulation upon shift to 36°C . The relationship between this operon and replication control proteins like Hda warrants investigation.

To study this relationship, researchers typically employ the following methodological approaches:

  • Creation of temperature-tolerant mutants through experimental evolution

  • Whole genome sequencing to identify mutations

  • Promoter reporter constructs to monitor gene expression changes at different temperatures

  • Analysis of replication dynamics at varying temperatures

How do mutations in the dnaN gene affect Hda function and what are the consequences for cellular physiology in P. luminescens?

Mutations in the dnaN gene, which encodes the β clamp processivity factor that interacts with Hda, can significantly alter Hda function with cascading effects on cellular physiology:

  • Enhanced RIDA activity: The dnaN-G157C mutation in related bacteria has been shown to increase regulatory inactivation of DnaA (RIDA), resulting in higher levels of DnaA-ADP compared to DnaA-ATP .

  • Under-replication phenotype: This mutation leads to an under-replication phenotype, indicating disrupted control of DNA replication initiation .

  • Gene expression changes: Changes in the DnaA-ATP/DnaA-ADP ratio due to altered Hda-β clamp interaction affect the expression of various genes. For example, expression of iraD was observed to be elevated by the dnaN-G157C mutation, strongly in exponential phase and to a lesser extent upon entry to stationary phase .

  • Growth effects: While specific to P. luminescens, mutations affecting replication control often impact growth characteristics and cell cycle progression.

Research methodologies to study these effects include:

  • Construction of specific dnaN mutants in P. luminescens

  • Analysis of replication dynamics using flow cytometry or microscopy

  • Gene expression profiling using RNA-seq or reporter constructs

  • Growth curve analysis under various conditions

  • Biochemical assessment of Hda-mediated ATP hydrolysis in the presence of wild-type and mutant β clamp proteins

What are the optimal conditions for assaying Hda protein activity in vitro?

The optimal conditions for assaying Hda protein activity in vitro focus on measuring its ability to stimulate ATP hydrolysis by DnaA in the presence of the β clamp. A comprehensive methodological approach includes:

  • Buffer composition:

    • Tris-HCl (pH 7.5-8.0): 25-50 mM

    • MgCl₂: 5-10 mM (essential for ATP hydrolysis)

    • NaCl or KCl: 50-150 mM

    • DTT or β-mercaptoethanol: 1-5 mM (to maintain reducing conditions)

    • BSA: 0.1-0.5 mg/ml (as a stabilizer)

  • Reaction components:

    • Purified recombinant P. luminescens Hda protein (typically 50-200 nM)

    • Purified DnaA protein (50-200 nM)

    • β clamp (dnaN gene product) (50-200 nM)

    • ATP (typically radiolabeled [α-³²P]-ATP at 1-2 μM)

    • DNA (optional, can enhance activity)

  • Detection methods:

    • Thin-layer chromatography to separate ATP from ADP

    • Liquid scintillation counting for quantification of radiolabeled products

    • Malachite green assay for phosphate release

    • Coupled enzyme assays that link ATP hydrolysis to NADH oxidation

  • Controls:

    • DnaA alone to establish baseline ATP hydrolysis

    • Heat-inactivated Hda to control for contaminating ATPase activity

    • Varying concentrations of components to establish dose-dependency

  • Optimization considerations:

    • Temperature (typically 25-30°C, reflecting P. luminescens optimal growth temperature)

    • Incubation time (usually 15-60 minutes)

    • Order of addition of components (typically pre-incubating β clamp with DNA before adding other components)

How can researchers effectively study the interaction between Hda and the β clamp in P. luminescens?

Studying the interaction between Hda and the β clamp (dnaN gene product) in P. luminescens requires multiple complementary approaches:

  • Protein-protein interaction assays:

    • Pull-down assays: Using tagged versions of either Hda or β clamp to capture interaction partners

    • Surface Plasmon Resonance (SPR): For real-time monitoring of binding kinetics

    • Isothermal Titration Calorimetry (ITC): To determine binding affinity and thermodynamics

    • Fluorescence resonance energy transfer (FRET): By labeling Hda and β clamp with appropriate fluorophores

  • Structural studies:

    • X-ray crystallography: To determine the 3D structure of the Hda-β clamp complex

    • Cryo-electron microscopy: For visualization of larger complexes

    • NMR spectroscopy: For dynamic interaction studies

  • Mutational analysis:

    • Alanine scanning mutagenesis: To identify critical residues for interaction

    • Domain swapping: Between P. luminescens Hda and homologs from other species

    • Analysis of natural variants: Like the dnaN-G157C that affects RIDA activity

  • In vivo approaches:

    • Bacterial two-hybrid assays: To confirm interactions in a cellular context

    • Fluorescence microscopy: Using fluorescently tagged proteins to observe co-localization

    • Co-immunoprecipitation: From P. luminescens lysates followed by Western blotting

  • Functional assays:

    • RIDA assays: To measure the effect of mutations on Hda-stimulated ATP hydrolysis by DnaA

    • DNA replication assays: To assess the functional consequences of altered interactions

What techniques can be used to investigate how Hda protein levels influence genome stability and replication control in P. luminescens?

Investigating the influence of Hda protein levels on genome stability and replication control in P. luminescens requires a comprehensive set of techniques:

  • Modulation of Hda expression:

    • Inducible expression systems: Using vectors with controllable promoters to overexpress Hda

    • CRISPR-Cas9 gene editing: To create defined mutations or deletions in the hda gene

    • Antisense RNA strategies: To reduce Hda levels post-transcriptionally

  • Replication dynamics analysis:

    • Marker frequency analysis: Using qPCR or next-generation sequencing to measure ori:ter ratios

    • DNA fiber analysis: To visualize individual replication forks and measure fork progression rates

    • Flow cytometry: To assess DNA content distribution in cell populations

  • Genome stability assessment:

    • Mutation rate measurements: Using fluctuation tests with selective markers

    • DNA damage response monitoring: Through reporters for SOS response activation

    • Whole genome sequencing: To identify mutations or structural variations arising from altered Hda levels

  • Cell cycle analysis:

    • Time-lapse microscopy: To track cell division patterns and timing

    • Synchronization methods: To examine replication events at specific cell cycle stages

    • BrdU incorporation: To label newly synthesized DNA

  • Molecular approaches:

    • ChIP-seq: To map DnaA binding sites across the genome and how they change with Hda levels

    • RNA-seq: To determine global transcriptional changes resulting from altered Hda levels

    • ATP/ADP ratio measurements: Using luciferase-based assays to monitor cellular energy status

How does the Hda protein from P. luminescens compare structurally and functionally to its homologs in other bacterial species?

The Hda protein from P. luminescens shares structural and functional similarities with homologs in other bacterial species, but also exhibits unique characteristics:

  • Sequence conservation:

    • The P. luminescens Hda protein consists of 233 amino acids

    • Key domains are conserved across bacterial species, particularly the regions involved in β clamp binding and ATPase stimulation

    • Specific sequence variations may account for species-specific regulation

  • Functional conservation and differences:

    • Core function: All Hda homologs participate in RIDA (Regulatory Inactivation of DnaA) by stimulating ATP hydrolysis by DnaA in a β clamp-dependent manner

    • Regulatory distinctions: The exact conditions under which Hda activity is modulated may differ between species

    • Temperature sensitivity: P. luminescens has specific temperature restrictions for growth , which may be reflected in the thermal stability and activity profile of its Hda protein

  • Structural comparison with E. coli Hda:

    • While the specific structure of P. luminescens Hda has not been fully characterized, based on homology to E. coli:

    • An N-terminal AAA+ domain that interacts with DnaA

    • A distinctive clamp-binding motif that mediates interaction with the β clamp

  • Species-specific interactions:

    • Potential interactions with P. luminescens-specific proteins involved in its unique lifecycle as an entomopathogenic bacterium that lives in mutualistic association with soil nematodes

  • Evolutionary perspective:

    • Conservation patterns suggest strong selective pressure on core functions

    • Divergence in regulatory regions may reflect adaptation to different ecological niches

Methodological approaches for comparative analysis include:

  • Multiple sequence alignment of Hda proteins from diverse bacterial species

  • Homology modeling based on known structures

  • Heterologous expression and complementation studies

  • Exchange of domains between Hda proteins from different species to identify functional determinants

What role might Hda play in the life cycle of P. luminescens, particularly during the transition between insect pathogenesis and nematode symbiosis?

P. luminescens has a complex life cycle involving symbiosis with nematodes and pathogenicity toward insects . The role of Hda in these transitions may be multifaceted:

  • Regulation of replication during host transitions:

    • Hda-mediated control of DnaA activity may help coordinate DNA replication with the changing metabolic demands during transitions between hosts

    • Different growth rates in nematode versus insect environments may require distinct replication control mechanisms

  • Coordination with environmental sensing:

    • Temperature-dependent regulation: P. luminescens exhibits temperature restrictions , and Hda may contribute to sensing and responding to temperature changes encountered during host transitions

    • Nutritional status sensing: ATP/ADP ratios change with nutrient availability, potentially linking Hda activity to the variable nutrient environments of different hosts

  • Interaction with virulence systems:

    • P. luminescens produces various toxins and uses multiple secretion systems for insect pathogenesis

    • Replication control through Hda may be coordinated with virulence factor expression

    • DNA adenine methyltransferase (Dam) affects P. luminescens motility and virulence , potentially interacting with Hda-mediated pathways

  • Response to stress conditions:

    • Insects deploy immune responses that create stressful conditions for invading bacteria

    • Hda may help regulate replication during stress response, preventing detrimental over-initiation

Research approaches to investigate these relationships include:

  • Transcriptomic and proteomic analysis of hda expression during different life cycle stages

  • Creation of conditionally active Hda variants to study effects at specific points in the life cycle

  • In vivo imaging of fluorescently tagged Hda to track localization during host transitions

  • Analysis of replication dynamics in wild-type versus hda mutant strains during insect infection and nematode colonization

How can knowledge about P. luminescens Hda be applied to develop novel antimicrobial strategies?

Understanding the function of Hda in P. luminescens could inform novel antimicrobial development strategies:

  • Targeting bacterial replication control:

    • Small molecule inhibitors of the Hda-β clamp interaction could disrupt replication control, leading to replication stress and potential cell death

    • Compounds that affect the DnaA-ATP/DnaA-ADP balance could similarly disrupt essential cell cycle processes

  • Comparative analysis for broad-spectrum applications:

    • Identifying conserved features of Hda across pathogenic bacteria could lead to broad-spectrum antimicrobials

    • Targeting unique features of P. luminescens Hda could provide specific control agents for this insect pathogen

  • Synthetic biology approaches:

    • Engineered Hda variants could be introduced into bacteria to create sensitized strains for biological control

    • Temperature-sensitive Hda variants could expand the range of conditions under which P. luminescens can be applied for insect control

  • Screening methodologies:

    • In vitro screening: Using purified Hda protein in ATP hydrolysis assays to identify inhibitory compounds

    • Cell-based screening: Using reporter systems that respond to replication stress to identify compounds affecting Hda function

    • Structure-based drug design: Using the known or predicted structure of Hda to design targeted inhibitors

  • Applications in antimicrobial resistance research:

    • Hda targets a conserved and essential bacterial process, potentially avoiding existing resistance mechanisms

    • Combination therapies targeting both Hda function and other cellular processes could reduce resistance development

What methods can be developed to use recombinant Hda protein as a tool for studying bacterial replication in diverse species?

Recombinant Hda protein from P. luminescens can serve as a valuable research tool for studying bacterial replication:

  • Developing in vitro replication systems:

    • Purified Hda, along with DnaA and the β clamp, can be used to reconstitute RIDA activity in vitro

    • These systems can be used to study the molecular mechanisms of replication control under defined conditions

  • Creating biotinylated or fluorescently labeled Hda variants:

    • These modified proteins can be used as probes to identify interaction partners

    • They can also serve as trackers for visualizing replication dynamics in live cells

  • Cross-species complementation studies:

    • P. luminescens Hda can be expressed in other bacterial species to determine functional conservation

    • Chimeric Hda proteins combining domains from different species can identify species-specific functional determinants

  • Development of Hda-based biosensors:

    • Engineered Hda variants fused to reporter proteins could serve as sensors for replication stress

    • These biosensors could be valuable tools for screening antimicrobial compounds

  • Methodological protocols for activity assays:

    • Standardized protocols for expressing and purifying active recombinant Hda

    • Optimized assay conditions for measuring Hda-dependent stimulation of DnaA ATPase activity

    • Controls and reference standards for comparative studies across different laboratories

How might the temperature restriction mechanisms in P. luminescens, possibly involving Hda, be utilized in biotechnological applications?

The temperature restriction mechanisms in P. luminescens, potentially involving Hda, offer interesting biotechnological applications:

  • Temperature-controlled gene expression systems:

    • Using temperature-responsive elements from P. luminescens to develop temperature-inducible expression systems

    • The TRL operon shows up-regulation upon shift to 36°C and could be harnessed for biotechnological applications

  • Containment strategies for engineered microorganisms:

    • Creating organisms that can only replicate within defined temperature ranges

    • Using knowledge of temperature restriction mechanisms to design biological safeguards

  • Thermosensitive biological control agents:

    • Developing strains with modified temperature restrictions for insect pest control in specific environmental conditions

    • Creating versions that are automatically limited by environmental temperature changes

  • Tools for synthetic biology:

    • Temperature-sensitive replication modules could serve as environmental switches in synthetic circuits

    • Integration of temperature sensing with other cellular processes for programmed responses

  • Methodological approaches for development:

    • Directed evolution to select for variants with desired temperature properties

    • Genetic engineering of the TRL operon and related components

    • Creation of chimeric proteins combining temperature-sensitive domains with functional domains from other proteins

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.