Recombinant Legionella pneumophila subsp. pneumophila ATP-dependent Clp protease adapter protein ClpS (clpS)

Shipped with Ice Packs
In Stock

Description

Introduction to Recombinant Legionella pneumophila subsp. pneumophila ATP-dependent Clp Protease Adapter Protein ClpS (ClpS)

Recombinant Legionella pneumophila subsp. pneumophila ATP-dependent Clp protease adapter protein ClpS (ClpS) is a recombinant protein produced from the bacterium Legionella pneumophila, a causative agent of Legionnaires' disease. ClpS is an adapter protein that plays a crucial role in the Clp proteolytic system, which is essential for protein quality control and regulation in bacteria. This system involves the degradation of misfolded or damaged proteins to maintain cellular homeostasis.

Function of ClpS in the Clp Proteolytic System

ClpS acts as a specificity factor for the ClpA or ClpX ATPases, which are part of the Clp proteolytic complex. It helps in recognizing and delivering specific substrates to the ClpP protease for degradation. This process is vital for regulating protein turnover, stress response, and virulence in bacteria like Legionella pneumophila.

Production and Characteristics of Recombinant ClpS

Recombinant ClpS is produced in various expression systems, such as yeast or Escherichia coli, to ensure high purity and yield. The choice of expression host can affect the protein's characteristics, such as its folding and post-translational modifications.

Expression SystemCharacteristics
YeastHigh purity, suitable for structural studies
Escherichia coliRapid production, cost-effective

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 requested 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 consolidate the contents. Reconstitute the protein in sterile, deionized water to a concentration of 0.1-1.0 mg/mL. We recommend adding 5-50% glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our standard glycerol concentration is 50%, provided as a reference for customers.
Shelf Life
Shelf life depends on various factors, including storage conditions, buffer components, 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
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 for preferential development.
Synonyms
clpS; lpg0817ATP-dependent Clp protease adapter protein ClpS
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-111
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Legionella pneumophila subsp. pneumophila (strain Philadelphia 1 / ATCC 33152 / DSM 7513)
Target Names
clpS
Target Protein Sequence
MSKQNLEEII QTGIADTELS TEISTAIKRP RKYKVLLLND DYTPMDFVVE VLKHFFHLNE EVAIQVMLQV HFQGKGVCGV FTRDIAETKV ALVNEYARMN QHPLLSSMEP E
Uniprot No.

Target Background

Function
Involved in modulating the specificity of ClpAP-mediated ATP-dependent protein degradation.
Database Links

KEGG: lpn:lpg0817

STRING: 272624.lpg0817

Protein Families
ClpS family

Q&A

What is the role of ClpS in the Clp protease system of Legionella pneumophila?

ClpS functions as a substrate recognition and delivery component in the Clp proteolytic pathway of L. pneumophila. It specifically recognizes N-end rule substrates and delivers them to the ClpAP protease complex for degradation. Within this system, ClpP serves as the proteolytic core while ClpA acts as the ATP-dependent unfoldase that prepares substrates for degradation . The entire system is crucial for protein homeostasis in L. pneumophila, with ClpS providing specificity to the proteolytic process by selecting appropriate substrates bearing N-terminal degradation signals.

How does ClpS relate to the biphasic life cycle of Legionella pneumophila?

The biphasic life cycle of L. pneumophila consists of a nonvirulent replicative phase and a virulent transmissive phase, with the transition between these phases being tightly regulated by various factors including the Clp protease system . While the search results primarily focus on ClpP's role, ClpS likely contributes to this regulation by controlling which proteins are targeted for degradation during different phases. The completion of this biphasic life cycle and bacterial pathogenesis is greatly dependent on protein homeostasis regulated by ClpP-dependent proteolysis . As ClpS directs substrate selection for the ClpAP complex, it plays an upstream regulatory role in this process, potentially influencing which proteins are degraded at specific life cycle stages.

What is the relationship between ClpS and the ClpP protease in Legionella pneumophila?

ClpS and ClpP function as part of the same proteolytic machinery but with distinct roles. While ClpP forms the proteolytic core that degrades target proteins, ClpS acts as an adapter that recognizes specific substrates (particularly those following the N-end rule) and delivers them to the ClpAP complex. Research has demonstrated that ClpP-dependent proteolysis is required for normal regulation of L. pneumophila differentiation and virulence . Though the search results don't explicitly discuss ClpS, its role in substrate selection would logically influence which proteins are ultimately processed by ClpP, thereby affecting the same cellular processes controlled by ClpP-dependent proteolysis.

What are the best approaches for expressing and purifying recombinant ClpS from Legionella pneumophila?

For recombinant expression of L. pneumophila ClpS, researchers should consider the following methodology:

  • Expression System Selection: E. coli BL21(DE3) or similar strains are recommended for high-yield expression of recombinant ClpS. The gene should be cloned into vectors with strong inducible promoters such as pET series vectors with T7 promoters.

  • Optimization Protocol:

    • Transform expression plasmid into appropriate E. coli strain

    • Culture cells at 37°C to mid-log phase (OD600 of 0.6-0.8)

    • Induce expression with 0.5-1.0 mM IPTG

    • Shift temperature to 18-25°C for 16-18 hours to enhance solubility

    • Harvest cells by centrifugation at 5000×g for 15 minutes

  • Purification Strategy:

    • Lyse cells in buffer containing 50 mM Tris-HCl (pH 8.0), 300 mM NaCl, 10% glycerol, and protease inhibitors

    • Apply a His-tag or similar affinity tag for initial purification using affinity chromatography

    • Further purify using ion-exchange chromatography

    • Finalize with size-exclusion chromatography to obtain highly pure protein

    • Verify purity by SDS-PAGE and Western blotting

This approach mirrors techniques used for other Clp system components as referenced in the literature on ClpP purification and characterization .

How can researchers create and validate clpS knockout mutants in Legionella pneumophila?

Creating and validating clpS knockout mutants requires a systematic approach:

Creation Protocol:

  • Design upstream and downstream flanking sequences of the clpS gene (similar to the methodology used for clpP deletion )

  • Amplify these sequences using PCR with appropriate primer pairs

  • Perform fusion PCR to join the flanking sequences

  • Digest the fusion PCR product with appropriate restriction enzymes

  • Sub-clone into a suicide vector (such as pBRDX as used for clpP knockout )

  • Introduce the construct into wild-type L. pneumophila by electroporation

  • Select transformants on appropriate antibiotic-containing media

  • Screen for vector loss using counter-selection methods

  • Verify the deletion by PCR and sequencing

Validation Approaches:

  • Genetic Verification:

    • PCR analysis using primers that flank the deleted region

    • Whole-genome sequencing to confirm clean deletion without affecting adjacent genes

  • Protein Expression Verification:

    • Western blot analysis using antibodies against ClpS

    • Proteomics analysis to confirm absence of ClpS peptides

  • Functional Characterization:

    • Assess growth patterns in both rich media and during infection of host cells

    • Measure virulence traits including invasiveness in amoebae models and macrophages

    • Analyze protein degradation patterns of known N-end rule substrates

This methodology follows similar principles to those described for creating clpP knockout strains in L. pneumophila .

What methods are recommended for investigating ClpS-substrate interactions in Legionella pneumophila?

To investigate ClpS-substrate interactions, researchers should employ multiple complementary approaches:

  • In Vitro Binding Assays:

    • Purify recombinant ClpS and potential substrate proteins

    • Perform pull-down assays using tagged ClpS

    • Conduct surface plasmon resonance (SPR) to determine binding kinetics

    • Use isothermal titration calorimetry (ITC) to measure thermodynamic parameters

  • Substrate Identification:

    • Employ co-immunoprecipitation with anti-ClpS antibodies followed by mass spectrometry

    • Conduct bacterial two-hybrid screening to identify potential interacting partners

    • Perform comparative proteomics between wild-type and ΔclpS strains to identify accumulating substrates

  • Degradation Assays:

    • Reconstitute the ClpAPS system in vitro with purified components

    • Measure degradation of putative substrates with and without ClpS

    • Use fluorescence-based assays with labeled substrates to monitor degradation kinetics

  • In Vivo Validation:

    • Create reporter fusions with potential substrates

    • Monitor substrate stability in wild-type vs. ΔclpS backgrounds

    • Perform site-directed mutagenesis of N-terminal residues to confirm N-end rule targeting

These methodologies are extrapolated from approaches used to study ClpP-dependent proteolysis in L. pneumophila .

How does ClpS contribute to the virulence regulation in Legionella pneumophila?

ClpS likely contributes to virulence regulation through targeted protein degradation, though its specific role must be inferred from what we know about the ClpP system:

Virulence Regulation Mechanisms:

  • Effector Protein Control: Similar to ClpP's role in regulating effector translocation , ClpS may influence which effector proteins are targeted for degradation, thereby affecting the T4BSS (Type IVB Secretion System) function that is essential for L. pneumophila pathogenesis.

  • Life Cycle Phase Transitions: The ClpP-dependent proteolysis system is crucial for the transition between replicative and transmissive phases . ClpS likely participates in this regulation by controlling the degradation of specific substrates involved in phase transitions.

  • Global Regulator Processing: ClpP regulates the global regulator CsrA at both transcriptional and protein levels . ClpS may be involved in recognition and delivery of key transcriptional regulators to the ClpAP protease complex.

Experimental approaches to investigate this aspect would include:

  • Comparative virulence assays between wild-type and ΔclpS strains in various host cell models

  • Proteomics analysis of differentially degraded virulence factors

  • Investigation of the escape from endosome-lysosomal pathway in ΔclpS mutants

  • Analysis of T4BSS effector translocation efficiency in the absence of ClpS

These investigations would build upon findings that ClpP is required for L. pneumophila to escape the endosome-lysosomal pathway and for efficient translocation of certain effector proteins .

What is the interplay between ClpS and other proteolytic systems in regulating Legionella pneumophila physiology?

The regulation of L. pneumophila physiology involves complex interactions between multiple proteolytic systems:

Cross-System Interactions:

Proteolytic SystemInteraction with ClpSPhysiological Impact
Lon proteasePotential substrate overlapCell division, stress response
HslUV proteaseComplementary substrate specificityProtein quality control
FtsH membrane proteaseCompartmentalized proteolysis coordinationMembrane protein turnover
Proteasome-like proteasesHierarchical degradation decisionsResponse to specific stresses

Research approaches should include:

  • Creating double knockouts (ΔclpS plus another proteolytic component)

  • Comparative proteomics across single and double knockout strains

  • Global analysis of protein half-lives in various protease mutant backgrounds

  • Investigation of stress responses in protease mutant combinations

These studies would extend understanding beyond the known roles of ClpP-dependent proteolysis in regulating L. pneumophila differentiation and virulence .

How do post-translational modifications affect ClpS function in substrate recognition and delivery?

Post-translational modifications (PTMs) likely play significant roles in regulating ClpS activity:

Potential ClpS Modifications and Their Effects:

  • Phosphorylation: May alter substrate binding affinity or interaction with ClpA. Key residues likely include conserved serine and threonine residues in the substrate-binding domain.

  • Acetylation: Could modify the charged surface of ClpS, affecting its interaction with N-degrons of substrate proteins.

  • Oxidation: Under stress conditions, oxidation of key cysteine residues might serve as a regulatory mechanism to adjust ClpS activity in response to oxidative stress.

Investigation Methodology:

  • Mass spectrometry analysis of purified ClpS to identify PTMs

  • Site-directed mutagenesis of modified residues to create phosphomimetic or non-modifiable variants

  • In vitro reconstitution assays comparing native and modified ClpS

  • Temporal analysis of ClpS modifications during the biphasic life cycle

This research direction is particularly relevant given the findings that the biphasic life cycle of L. pneumophila requires precisely timed proteolysis events , suggesting that regulation of proteolytic adapter proteins like ClpS is likely subject to sophisticated control mechanisms.

How can researchers resolve contradictory findings regarding ClpS specificity in different bacterial models versus Legionella pneumophila?

Resolving contradictory findings requires systematic comparative approaches:

Reconciliation Strategy:

  • Direct Comparison Studies:

    • Clone and express ClpS from different bacterial species in the same expression system

    • Conduct side-by-side substrate binding assays using identical methodologies

    • Perform cross-complementation studies by expressing foreign ClpS in L. pneumophila ΔclpS

  • Structural Analysis:

    • Determine crystal structures of L. pneumophila ClpS alone and in complex with substrates

    • Compare with structures from other bacterial species

    • Identify key residues that differ in the substrate-binding pocket

  • Evolutionary Context:

    • Perform phylogenetic analysis of ClpS across bacterial species

    • Correlate sequence divergence with lifestyle and pathogenicity

    • Identify selective pressures on substrate specificity regions

  • Experimental Validation:

    • Create chimeric ClpS proteins with domains from different species

    • Test substrate specificity of these chimeras

    • Use directed evolution to shift specificity of L. pneumophila ClpS toward that of other species

This approach acknowledges that adaptation to different ecological niches may have driven the evolution of distinct substrate preferences in L. pneumophila ClpS compared to other bacterial species.

What methodological challenges exist in studying the temporal dynamics of ClpS-mediated proteolysis during the Legionella pneumophila life cycle?

Studying temporal dynamics of ClpS activity faces several methodological challenges:

Key Challenges and Solutions:

ChallengeSolution Approach
Asynchronous bacterial culturesDevelop synchronization protocols using nutrient shifts or density gradients
Low abundance of ClpSCreate conditionally tagged versions that maintain function but allow enrichment
Transient substrate interactionsEmploy photocrosslinking with modified ClpS to capture fleeting interactions
Determining in vivo degradation ratesDevelop fluorescent timer fusion proteins with potential substrates
Distinguishing ClpS-dependent from independent degradationCreate specialized substrate reporters with N-degron modifications

Advanced Methodological Framework:

  • Implement ribosome profiling paired with proteomics to distinguish between translational and post-translational regulation

  • Develop single-cell tracking of ClpS activity using fluorescence resonance energy transfer (FRET)-based sensors

  • Employ microfluidic devices to monitor individual bacterial cells through life cycle transitions while measuring ClpS activity

This methodological approach builds on insights from temporal expression studies of regulatory proteins like CsrA that are controlled by ClpP-dependent proteolysis during the L. pneumophila life cycle .

How might the understanding of ClpS function inform the development of targeted antimicrobial strategies against Legionella pneumophila?

Leveraging ClpS research for antimicrobial development presents unique opportunities:

Therapeutic Strategy Framework:

  • Direct ClpS Inhibition Approach:

    • Identify small molecules that bind to the substrate-binding pocket of ClpS

    • Develop peptide mimetics that occupy the ClpS-ClpA interaction interface

    • Create N-degron analogs that bind ClpS but resist delivery to ClpAP

  • Pathway Modulation Strategy:

    • Design molecules that hyperactivate ClpS to cause excessive degradation of essential proteins

    • Develop compounds that alter ClpS specificity to target non-canonical substrates

    • Create synthetic substrates that hijack the ClpS-ClpAP system to deplete cellular energy

  • Selectivity Considerations:

    • Focus on unique structural features of L. pneumophila ClpS not present in human cells or beneficial bacteria

    • Target virulence-specific functions rather than growth-essential roles to reduce selection pressure

    • Exploit differences in substrate recognition between L. pneumophila ClpS and other bacterial species

  • Experimental Validation Path:

    • Screen candidate molecules using in vitro reconstituted ClpS-ClpAP systems

    • Validate hits in cellular infection models

    • Assess resistance development through long-term evolution experiments

This approach builds on the understanding that disruption of the Clp protease system impairs L. pneumophila virulence , suggesting that targeted interference with ClpS function could provide a novel therapeutic avenue while potentially reducing selective pressure compared to broad-spectrum antibiotics.

What emerging technologies could advance our understanding of the ClpS interactome in Legionella pneumophila?

Several cutting-edge technologies show promise for mapping the complete ClpS interactome:

Advanced Technological Approaches:

  • Proximity-dependent Biotinylation (BioID or TurboID):

    • Create ClpS-BioID fusion proteins expressed in L. pneumophila

    • Identify proteins that come into proximity with ClpS during different life cycle phases

    • Distinguish between substrates, cofactors, and regulatory proteins

  • Cryo-Electron Microscopy:

    • Visualize ClpS-ClpA-substrate complexes at near-atomic resolution

    • Capture different states of the substrate delivery process

    • Determine structural changes during substrate recognition and handoff

  • Single-Molecule Tracking:

    • Visualize individual ClpS molecules in living bacteria using photoactivatable fluorescent proteins

    • Track dynamics of ClpS localization during different growth phases

    • Measure diffusion rates to infer complex formation and substrate binding

  • Integrative Multi-omics:

    • Combine transcriptomics, proteomics, and metabolomics in wild-type and ΔclpS strains

    • Construct network models of ClpS-dependent processes

    • Identify metabolic pathways indirectly affected by ClpS activity

These technologies would extend current understanding of protein degradation in L. pneumophila beyond what has been established for ClpP-dependent proteolysis .

How might systems biology approaches help integrate ClpS function into broader Legionella pneumophila regulatory networks?

Systems biology offers powerful frameworks for contextualizing ClpS function:

Integration Strategies:

  • Mathematical Modeling:

    • Develop ordinary differential equation models of protein degradation kinetics

    • Create stochastic models of ClpS-substrate encounters

    • Build genome-scale models incorporating proteolysis into metabolic networks

  • Network Analysis:

    • Construct protein-protein interaction networks centered on ClpS

    • Identify regulatory hubs connected to ClpS-mediated degradation

    • Map feedback loops involving proteolysis and gene expression

  • Multi-scale Integration:

    • Connect molecular events (substrate degradation) to cellular outcomes (virulence)

    • Model temporal dynamics across different timescales (seconds for binding, hours for life cycle transitions)

    • Link proteostasis to other cellular systems including metabolism and virulence

  • Predictive Applications:

    • Identify potential intervention points for disrupting pathogenesis

    • Predict compensatory mechanisms when ClpS function is compromised

    • Forecast evolutionary trajectories under different selective pressures

This systems approach would build upon findings that ClpP-dependent proteolysis spans a broad physiological spectrum involving key metabolic pathways that regulate the transition of the biphasic life cycle and bacterial virulence .

What are the implications of ClpS research for understanding bacterial adaptation to environmental stresses and host defense mechanisms?

ClpS research has broad implications for bacterial stress adaptation:

Cross-cutting Research Themes:

  • Environmental Persistence:

    • Investigate how ClpS contributes to survival in water systems through targeted protein turnover

    • Determine if ClpS function changes in response to disinfectants or temperature shifts

    • Explore ClpS role in biofilm formation and maintenance

  • Host-Pathogen Interface:

    • Examine how ClpS-mediated proteolysis responds to host defense mechanisms

    • Investigate whether ClpS targets host-derived proteins during infection

    • Determine if ClpS activity is modulated by host-derived signals

  • Evolutionary Considerations:

    • Compare ClpS function across Legionella species with different host ranges

    • Investigate selective pressures on ClpS in environmental versus clinical isolates

    • Identify genetic variations in clpS associated with enhanced virulence

  • Broader Bacterial Physiology:

    • Explore commonalities and differences in ClpS function across bacterial pathogens

    • Investigate conservation of substrate recognition mechanisms

    • Determine if ClpS represents a convergent evolutionary solution to similar selective pressures

This research direction connects to the observation that ClpP-dependent proteolysis facilitates L. pneumophila adaptation to both aquatic and intracellular niches , suggesting that ClpS likely plays a role in this adaptability through its substrate selection function.

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.