Recombinant Chlamydophila caviae ATP-dependent Clp protease ATP-binding subunit ClpX (clpX)

Shipped with Ice Packs
In Stock

Description

Overview of Recombinant Chlamydophila caviae ATP-dependent Clp Protease ATP-binding Subunit ClpX (clpX)

ClpX is an ATP-dependent protein unfoldase and polypeptide translocase . In bacteria, the ATP-dependent ClpX protease participates in protein quality control, stress tolerance, virulence factor production, and binding to protein degradation tags in E. coli . ClpX, an ATPase, supplies the energy for unfolding and moving target proteins into the ClpP protease for degradation; ClpP then cleaves the unfolded target proteins .

Clp Protease Complexes and ClpX Function

Clp proteases have a two-component structure that includes multiple chaperone rings and two different proteolytic cores, allowing for several possible combinations of Clp protease complexes . The ClpP protease core can interact with different chaperones, namely ClpA, ClpC, ClpE, and ClpX, to form active chaperone-protease complexes. ClpA and ClpX of E. coli are protein unfoldases that require ATP to function and individually associate with the ClpP protease to facilitate targeted protein degradation .

ClpX in Chlamydia trachomatis (ctClpX)

The Chlamydia trachomatis (ctClpXP) ClpXP protease is formed by a hetero-tetradecameric proteolytic core composed of two distinct homologs of ClpP (ctClpP1 and ctClpP2) that associates with the unfoldase ctClpX via ctClpP2 for regulated protein degradation . The chlamydial Clp-ATPase ctClpX and the antibiotic ADEP1 interact with the hetero-tetradecameric complex ctClpP1P2 . ctClpX interacts with ctClpP1P2 and confers proteolytic activity to the complex .

Structure of ClpX

ClpX is a member of the AAA+ ATPase protein family, forming the Clp protease protein complex . Human ClpX is derived from the Clp protease protein complex structure. These hexameric HSP100/Clp proteins create ring-like structures similar to chaperonins . ClpX includes a single consensus ATP-binding site motif and has limited homology to regions of ClpA and other members of the ClpA/B/C family .

ClpXP as a Target for Antibiotics

Clp proteases have garnered interest as antibiotic targets due to their conservation among bacteria, including important human pathogens. Antibiotics of the ADEP class interfere with protease functions by preventing the interaction of ctClpX with ctClpP1P2 and activating the otherwise dormant proteolytic core for unregulated proteolysis .

ClpX and Protein Degradation

ClpXP is an ATP-dependent protease where the ClpX AAA+ motor binds, unfolds, and translocates specific protein substrates into ClpP's degradation chamber . ClpX is an ATP-dependent protein unfoldase and polypeptide translocase, while ClpP is a self-compartmentalized peptidase .

Regulation of ClpX

The regulation of ClpX and ClpP expression is complex, involving transcriptional regulators, environmental signals, and post-transcriptional modifications. Understanding how these proteins' expression is regulated can provide insights into the mechanisms by which bacteria respond to stress and maintain cellular homeostasis .

Role of ClpX in Mycobacteria

ClpX is essential and activated by single-strand DNA binding protein in mycobacteria . ClpX's rate of ATP hydrolysis increases with increasing concentrations of SSB. SSB increased ClpX’s rate of ATP hydrolysis by 100% .

Symmetry Mismatch in ClpXP Complex Formation

The formation of the ClpXP complex involves a symmetry mismatch because hexameric AAA+ rings bind axially to the opposing stacked heptameric rings of the tetradecameric ClpP .

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 contact your local distributor for precise delivery estimates.
Note: All proteins are shipped with standard blue ice packs unless dry ice shipping is specifically requested. Please contact us in advance; additional fees apply.
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. 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% and serves as a reference.
Shelf Life
Shelf life depends on several factors: 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 crucial for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing.
The tag type is determined during production. If a specific tag type is required, please inform us, and we will prioritize its development.
Synonyms
clpX; CCA_00921ATP-dependent Clp protease ATP-binding subunit ClpX
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-421
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Chlamydophila caviae (strain GPIC)
Target Names
clpX
Target Protein Sequence
MNKKNLTICS FCGRSEKDVE KLIAGPSVYI CDYCIKLCSG ILDKKPTSTP SSGTPTETTP QHSDLQVLTP KEIKKHIDKY VVGQERAKKT IAVAVYNHYK RIRALLNNKH VSYGKSNVLL LGPTGSGKTL IAKTLAKILD VPFTIADATT LTEAGYVGED VENIVLRLLQ AADYNVARAE RGIIYIDEID KIGRTTANVS ITRDVSGEGV QQALLKIIEG TTANVPPKGG RKHPNQEYIR VNTENILFIV GGAFVNLDKI IAKRLGKTTI GFSDDLGDFS QEDRDHLLTK VETEDLIAFG MIPEFVGRFN CIVNCEELSL DELVAILTEP TNAIVKQYIE LFSEENVKLI FEKDALYAIA KKAKLAKTGA RALGMILENL LRDLMFEIPS DPTVEAIRIQ EDTILENKAP VIIRRTPEAI A
Uniprot No.

Target Background

Function
ATP-dependent specificity component of the Clp protease; directs the protease to specific substrates. Exhibits chaperone functions independently of ClpP.
Database Links
Protein Families
ClpX chaperone family

Q&A

What is the ClpX protein and how does it function in Chlamydophila caviae?

ClpX is an AAA+ (ATPases Associated with diverse cellular Activities) unfoldase that functions as a regulatory ATP-binding subunit of the ATP-dependent Clp protease complex in Chlamydophila caviae. It operates in conjunction with the ClpP subunit, which contains the proteolytic active site, to form the complete ClpXP complex responsible for targeted protein degradation. ClpX specifically recognizes substrate proteins, uses ATP hydrolysis to unfold these proteins, and then translocates them into the proteolytic chamber of ClpP where degradation occurs. This separation of protein recognition/unfolding (ClpX) from proteolysis (ClpP) allows for tightly regulated and specific protein degradation, which is essential for various cellular processes including quality control and developmental transitions .

How is the clpX gene organized in Chlamydophila species?

In Chlamydophila and other Chlamydia species, the clpX gene is organized in an operon with the clpP gene. Both genes are cotranscribed in a single heat-inducible mRNA transcript of approximately 2200 bases, with clpP being the promoter-proximal gene. This genetic arrangement indicates coordinated regulation of these functionally related components. The clpX gene encodes a protein with an approximate molecular weight of 46,300 Da. The protein sequence contains a single consensus ATP-binding site motif essential for its ATPase activity and shares limited homology with regions of ClpA and other members of the ClpA/B/C family of ATPases .

What evidence indicates the importance of ClpX in chlamydial development?

Multiple experimental approaches have demonstrated the critical importance of ClpX in chlamydial development. Studies have shown that overexpressing catalytic mutant isoforms of ClpX (but not wild-type) in Chlamydia has a negative impact on chlamydial growth and development, indicating a dominant-negative effect that disrupts normal function. This suggests that ClpX is involved in critical protein turnover events necessary for proper development. The obligate intracellular lifestyle of Chlamydia species, coupled with their reduced genomes, means that retention of the ClpXP system despite genome streamlining underscores its essential nature. Research with related Clp systems shows that mutations can abolish degradation of specific proteins in vivo, suggesting that the ClpXP complex regulates key processes in the biphasic developmental cycle that transitions between the infectious elementary body (EB) and the replicative reticulate body (RB) .

What are the current approaches for expressing and purifying recombinant C. caviae ClpX?

Expression and purification of recombinant Chlamydophila caviae ClpX typically follows a multi-step process optimized for obtaining functional protein. The clpX gene is initially cloned into expression vectors containing affinity tags (such as His-tag or GST-tag) for simplified purification. For expression, E. coli systems like BL21(DE3) are commonly used with optimized conditions including reduced temperatures (16-18°C), controlled IPTG concentration, and optimized induction timing to enhance soluble protein yield. Purification generally begins with affinity chromatography using the engineered tag, followed by size-exclusion chromatography to isolate the functional hexameric form of ClpX. To verify proper folding and activity, researchers typically conduct ATPase activity assays measuring ATP hydrolysis rates using methods like the malachite green assay to detect released phosphate. Functional interaction with ClpP can be assessed through in vitro degradation assays using model substrates like fluorescently tagged peptides or proteins .

How can researchers study the ClpX-ClpP interaction in Chlamydophila caviae?

Studying the interaction between ClpX and ClpP in Chlamydophila caviae requires both structural and functional approaches. Structurally, cryo-electron microscopy has proven highly effective for visualizing the ClpXP complex, with recent studies achieving resolutions of 2.8-3.2 Å. This approach allows researchers to generate detailed models of the complex, using software tools like Relion for data analysis and reconstruction, followed by structure refinement using programs like PHENIX and Coot. Functionally, the interaction can be studied through in vitro reconstitution of the ClpXP complex using purified components, followed by degradation assays with model substrates. Introducing mutations in key interface regions of either protein helps identify critical residues for complex formation and function. Additionally, biophysical methods such as surface plasmon resonance (SPR) or isothermal titration calorimetry (ITC) provide quantitative measurements of binding affinity and kinetics between ClpX and ClpP, offering insights into their association mechanism and potential regulation .

What techniques are available for genetic manipulation of the clpX gene in C. caviae?

Genetic manipulation of clpX in Chlamydophila caviae presents significant challenges due to the obligate intracellular lifestyle of these bacteria, but several approaches have been developed. Chemical transformation has been reported for C. caviae, although primarily with suicide vectors. This method typically involves treating the elementary body (EB) form with calcium chloride to induce DNA uptake. Alternatively, electroporation can introduce DNA into EBs, while PAMAM dendriplexes have been employed to deliver DNA directly to the reticulate body (RB) form within infected cells. For gene disruption or modification, researchers have used transposon mutagenesis systems, though with relatively low efficiency. The discovery that inactivation of the putative ComEC homolog reduced lateral gene transfer efficiency suggests that improving DNA uptake by manipulating competence machinery could enhance transformation efficiency. Additionally, the natural competence of Chlamydia for DNA uptake allows coinfection approaches, where strains carrying different selective markers exchange DNA through recombination, providing a method for genetic manipulation without direct transformation .

What role does the ClpXP complex play in the chlamydial developmental cycle?

The ClpXP proteolytic complex plays a critical role in the chlamydial developmental cycle by mediating controlled protein degradation essential for transitions between developmental stages. In the biphasic development cycle of Chlamydia, the bacteria alternate between the infectious elementary body (EB) and the replicative reticulate body (RB) forms. ClpX, as part of the ClpXP complex, contributes to this cycle by targeting specific proteins for degradation at precise times. Research has shown that disrupting ClpX function through overexpression of catalytically inactive mutants negatively impacts chlamydial growth and development. This suggests that ClpX-mediated proteolysis regulates key factors involved in the EB-to-RB and RB-to-EB transitions. While the specific protein substrates targeted by ClpX during different developmental stages are still being investigated, they likely include regulatory proteins that control gene expression, cell division, and differentiation. The timing and regulation of ClpX activity throughout this cycle represent critical aspects of chlamydial biology and potential targets for intervention .

How does substrate specificity of ClpX affect chlamydial biology?

The substrate specificity of ClpX is a key determinant of its function in Chlamydophila caviae, as it directs the ClpXP complex to degrade specific proteins at appropriate times. ClpX recognizes various degradation signals (degrons) on substrate proteins, with the ssrA tag being one well-characterized example that marks incomplete or damaged proteins for degradation. The recognition involves specific structural elements on ClpX, such as the RKH loops, which interact with these degradation tags. Structural studies of ClpX-substrate interactions have revealed distinct conformational states during substrate recognition and processing, providing insights into the mechanism of substrate engagement. The specificity of ClpX likely governs which proteins are degraded during different stages of the C. caviae developmental cycle and under various stress conditions. The selectivity of degradation appears to be determined by interaction of ClpP with different regulatory ATPase subunits, including ClpX. This modularity allows for diverse substrate pools to be targeted by the same proteolytic core, enhancing the versatility of the system in responding to changing cellular needs during development .

What are the methodologies for identifying natural substrates of ClpX in C. caviae?

Identifying natural substrates of ClpX in Chlamydophila caviae requires a multi-faceted approach combining proteomics, genetics, and biochemical techniques. A powerful strategy uses quantitative proteomics methods such as Stable Isotope Labeling with Amino acids in Cell culture (SILAC) or Tandem Mass Tag (TMT) to compare protein abundance in wild-type versus ClpX-deficient conditions. Proteins that accumulate when ClpX function is compromised represent potential substrates. This can be complemented by substrate-trapping using catalytically inactive ClpX variants (e.g., Walker B mutants) that bind but don't process substrates, coupled with pull-down assays and mass spectrometry to identify bound proteins. In vitro validation involves degradation assays using purified recombinant ClpXP and candidate substrates, with degradation monitored by SDS-PAGE, western blotting, or fluorescence-based methods. Bioinformatic approaches can identify proteins containing known ClpX recognition motifs in the C. caviae proteome. For in vivo validation, targeted proteomics or fluorescent protein fusion reporters can monitor specific candidate substrates under various conditions affecting ClpX activity. Crosslinking mass spectrometry (XL-MS) can capture transient enzyme-substrate interactions, providing direct evidence of physical association .

How can structural studies of ClpX enhance our understanding of substrate recognition?

Structural studies have significantly enhanced our understanding of how ClpX recognizes and processes substrates. Cryo-electron microscopy studies have achieved near-atomic resolution (2.8-3.2 Å) of ClpXP complexes, revealing the molecular details of substrate engagement. These structures have identified distinct conformational states during substrate processing, termed the "recognition complex" and "intermediate complex," providing insights into the dynamic nature of ClpX function. The structural data shows how specific loops in ClpX, including the RKH loops, interact with degradation tags on substrate proteins. By modeling how substrates like GFP-YALAA bind to ClpXP, researchers have visualized the initial recognition events and subsequent conformational changes that lead to substrate unfolding and translocation. These structural insights allow rational design of mutations that can alter substrate specificity or modulate ClpX activity. Additionally, comparing structures of ClpX from different bacterial species highlights conserved features essential for function as well as species-specific adaptations that might reflect specialized roles. Future structural studies focusing specifically on C. caviae ClpX could reveal unique features related to its function in the chlamydial developmental cycle .

What methodological approaches are best for studying ClpX ATP hydrolysis activity?

Studying the ATP hydrolysis activity of ClpX requires carefully designed experiments that provide quantitative and reproducible results. The recommended protocol begins with freshly purified ClpX protein (typically at 0.5-2 μM hexamer concentration) in an appropriate reaction buffer containing magnesium as a cofactor for ATP hydrolysis. The reaction is initiated by adding ATP (usually 2-5 mM), and aliquots are removed at defined time points and immediately quenched to stop the reaction. For detection of released phosphate, the malachite green assay offers high sensitivity and reproducibility. A standard curve using known phosphate concentrations allows quantification of ATP hydrolysis rates, typically expressed as moles of ATP hydrolyzed per mole of ClpX hexamer per minute. Essential controls include no-enzyme controls to account for spontaneous ATP hydrolysis and heat-denatured protein controls to confirm the activity is due to native ClpX. For kinetic analysis, performing the assay across a range of ATP concentrations allows determination of Km and Vmax values by fitting to the Michaelis-Menten equation. The effects of potential substrates, inhibitors, or ClpP on ATPase activity can provide valuable insights into regulatory mechanisms and functional interactions .

How does C. caviae ClpX compare with homologs in other bacterial species?

Chlamydophila caviae ClpX shares fundamental structural and functional features with homologs in other bacterial species while exhibiting adaptations specific to its niche as an obligate intracellular pathogen. Like other bacterial ClpX proteins, C. caviae ClpX contains the characteristic AAA+ domain with Walker A and B motifs essential for ATP binding and hydrolysis. Sequence analysis reveals that C. caviae ClpX has limited homology to regions of ClpA and other members of the ClpA/B/C family, reflecting evolutionary divergence. A notable distinction is that while some bacterial species can survive without ClpX, the Clp system appears to be essential in Chlamydia, highlighting its critical role in these obligate intracellular bacteria. The organization of clpX in an operon with clpP is conserved across many bacterial species, though regulatory mechanisms may differ. While ClpX in E. coli has been extensively studied and shown to interact with various adapter proteins that modulate substrate selection, the existence and identity of such adapters in C. caviae remain largely unknown. The substrate specificity of C. caviae ClpX may differ from other bacterial species, reflecting adaptations to the unique intracellular environment and developmental cycle of Chlamydia .

What insights can be gained from studying the evolution of ClpX in obligate intracellular bacteria?

Studying ClpX evolution in obligate intracellular bacteria like Chlamydophila caviae provides valuable insights into how protein quality control systems adapt to specialized niches. The retention of the ClpXP system in the reduced genomes of obligate intracellular bacteria underscores its essential function, suggesting strong selective pressure to maintain this proteolytic machinery despite genome streamlining. Comparative genomic analyses of clpX genes across different Chlamydia species can reveal patterns of conservation and divergence that correlate with host range, tissue tropism, or disease manifestations. For instance, specific adaptations in substrate recognition domains might reflect different protein degradation requirements in various host environments. The co-evolution of ClpX with its proteolytic partner ClpP and with key substrates represents another fascinating area of research. Analysis of selection pressures on different domains of ClpX can identify regions under positive selection, potentially indicating adaptation to new functions or substrates, versus regions under purifying selection, suggesting conserved critical functions. This evolutionary perspective can also inform our understanding of how these bacteria have adapted their protein quality control systems to the unique challenges of intracellular life .

What does the conservation of ClpX across Chlamydia species suggest about its functional importance?

The high degree of conservation of ClpX across diverse Chlamydia species strongly suggests its fundamental importance to chlamydial biology. Despite the genomic reduction that typically occurs in obligate intracellular bacteria as they adapt to their specialized niche, the retention of the clpX gene indicates strong selective pressure to maintain this function. This conservation extends across species with different host ranges and tissue tropisms, from human pathogens like C. trachomatis to animal pathogens like C. caviae, suggesting that ClpX serves core functions essential to the chlamydial lifestyle rather than host-specific adaptations. The organization of clpX in an operon with clpP is also conserved, indicating the importance of coordinated expression of these functionally linked components. Research has demonstrated that disrupting ClpX function negatively impacts chlamydial growth and development, providing experimental support for its essential role. Additionally, the finding that certain transposon mutants affecting protein quality control showed reduced infectivity in animal models highlights the importance of these systems in natural infection settings. Together, these observations reinforce the view that ClpX-mediated proteolysis represents a core process essential for the viability and virulence of Chlamydia species .

What statistical approaches should be used when analyzing ClpX activity data?

Analyzing ClpX activity data requires appropriate statistical approaches to ensure robust and interpretable results. For ATPase activity assays measuring the rate of ATP hydrolysis, researchers should employ regression analysis to determine initial velocities from time-course data, followed by fitting to enzyme kinetic models (such as Michaelis-Menten) to extract parameters like Km and Vmax. When comparing wild-type ClpX to mutant variants or examining the effects of potential inhibitors, analysis of variance (ANOVA) with appropriate post-hoc tests (such as Tukey's or Dunnett's) is recommended for multiple comparisons, while t-tests may be sufficient for simpler two-condition comparisons. For substrate degradation assays, which often produce non-linear degradation curves, non-linear regression analysis should be used to determine degradation rates. In all cases, researchers should report both the effect size (percent change, fold difference) and statistical significance (p-values), while also considering the practical significance of observed differences. Biological replicates (typically n ≥ 3) performed with independently prepared protein batches are essential for robust statistical analysis, as is the inclusion of appropriate controls such as no-enzyme and no-ATP conditions. For more complex experimental designs examining multiple factors simultaneously, multifactorial ANOVA or mixed-effects models may be more appropriate .

How can researchers determine the specificity of substrate degradation by ClpXP?

Determining the specificity of substrate degradation by ClpXP requires systematic experiments that establish both the selectivity and mechanism of substrate recognition. In vitro degradation assays should compare multiple potential substrates under identical conditions, quantifying degradation rates for each. Competition experiments where multiple substrates are present simultaneously can reveal preferential degradation, indicating substrate hierarchies. Mutagenesis of suspected degradation signals on substrate proteins can identify specific recognition elements and their relative contributions. For example, systematic mutation of C-terminal residues in an ssrA-tagged substrate can reveal the sequence requirements for efficient recognition. Control experiments with other AAA+ unfoldases (such as ClpA) are essential to distinguish ClpX-specific degradation from general susceptibility to AAA+ proteases. The influence of potential adapter proteins or other regulatory factors can be assessed by adding these components to purified degradation systems. Structural studies using techniques like hydrogen-deuterium exchange mass spectrometry can identify regions of the substrate that interact with ClpX during recognition. Kinetic analyses comparing Km and kcat values for different substrates provide quantitative measures of specificity. Together, these approaches can establish both the scope of ClpX substrate specificity and the molecular mechanisms underlying substrate selection .

What analytical approaches help distinguish direct from indirect effects of ClpX manipulation?

Distinguishing direct from indirect effects of ClpX manipulation requires a multi-faceted analytical approach. Direct interactions between ClpX and potential substrates or effector molecules should be demonstrated through techniques such as co-immunoprecipitation, bacterial two-hybrid assays, or in vitro binding studies with purified components. Complementary to this, in vitro degradation assays with reconstituted ClpXP complexes can confirm direct proteolytic targeting of specific substrates. Time-course studies are crucial for establishing causality by determining the temporal sequence of events following ClpX manipulation—direct effects typically occur rapidly, while indirect effects emerge later as downstream consequences. To identify indirect effects, comprehensive analyses such as proteomics can identify changes in protein abundance beyond direct ClpX substrates, while transcriptomics can reveal altered gene expression patterns that may contribute to observed phenotypes. Statistical methods such as path analysis or structural equation modeling can help dissect direct from indirect relationships in complex datasets. For genetic approaches, complementation experiments where wild-type ClpX is reintroduced into a mutant background should rescue direct effects more efficiently than indirect ones. Additionally, dose-response relationships typically show different patterns for direct versus indirect effects, with direct effects often showing more linear relationships with the level of ClpX activity .

What emerging technologies could advance our understanding of ClpX function in C. caviae?

Several emerging technologies hold great promise for advancing our understanding of ClpX function in Chlamydophila caviae. Single-molecule techniques, such as optical tweezers or magnetic tweezers, could provide unprecedented insights into the mechanics of ClpX-mediated protein unfolding and translocation at the molecular level. Cryo-electron tomography of intact Chlamydia within host cells could reveal the spatial organization of ClpXP complexes in their native cellular context. Advanced genome editing approaches, such as base editing or prime editing, might overcome current limitations in generating precise genetic modifications in Chlamydia. Microfluidic platforms for single-cell analysis could enable real-time monitoring of ClpX activity during the developmental cycle, potentially revealing cell-to-cell heterogeneity in proteolytic function. Proximity labeling approaches like APEX2 or TurboID could identify the dynamic interactome of ClpX throughout infection, revealing temporal changes in protein-protein interactions. Machine learning algorithms applied to multi-omics datasets could predict ClpX substrates and regulatory networks with greater accuracy than current bioinformatic approaches. These technological advances, combined with improvements in genetic manipulation of Chlamydia, are poised to resolve many outstanding questions regarding ClpX function in this important pathogen .

What key unresolved questions about ClpX function should researchers prioritize?

Despite significant progress, several unresolved questions about ClpX function in Chlamydophila caviae represent priorities for future research. The comprehensive identification of natural ClpX substrates throughout the developmental cycle remains incomplete, limiting our understanding of how ClpX-mediated proteolysis regulates chlamydial development. The specific mechanisms by which ClpX recognizes its substrates in C. caviae, including the potential role of adapter proteins that might modulate substrate selection, are largely unknown. The regulation of ClpX activity during different developmental stages and stress conditions, including potential post-translational modifications or interactions with regulatory molecules, represents another key knowledge gap. The precise contribution of ClpX to chlamydial virulence and host-pathogen interactions, particularly how ClpX-mediated proteolysis might influence immune evasion or host cell manipulation, requires further investigation. Additionally, the potential interactions between the ClpXP system and other proteolytic systems in Chlamydia remain poorly understood, leaving questions about functional redundancy or cooperation. Addressing these questions will require integrated approaches combining structural biology, proteomics, genetics, and cell biology, ultimately advancing our understanding of this essential component of chlamydial biology .

What is the optimal protocol for assessing ClpXP activity against model substrates?

The assessment of ClpXP activity against model substrates requires a carefully designed protocol that ensures reproducible and quantitative results. Begin with freshly purified ClpX (0.5-1 μM hexamer) and ClpP (1-2 μM 14-mer) in an appropriate reaction buffer (typically 50 mM Tris-HCl pH 7.5, 100 mM KCl, 10 mM MgCl2, 1 mM DTT). Include an ATP regeneration system consisting of 5 mM ATP, 16 mM creatine phosphate, and 0.032 mg/ml creatine kinase to maintain consistent ATP levels throughout the assay. For fluorescence-based assays, GFP-ssrA is an excellent model substrate, with degradation monitored by the decrease in fluorescence over time (excitation 467 nm, emission 511 nm). Alternatively, for gel-based assays, samples can be withdrawn at defined time points, quenched with SDS-PAGE loading buffer, and analyzed by gel electrophoresis followed by densitometry. Essential controls include reactions without ATP or with ATPγS (a non-hydrolyzable ATP analog) to confirm ATP dependence, reactions without ClpP to demonstrate the requirement for the proteolytic component, and reactions with known ClpX inhibitors as positive controls for inhibition. For kinetic analysis, vary the substrate concentration while keeping enzyme concentrations constant, and fit the resulting degradation rates to the Michaelis-Menten equation. When comparing different substrate variants or testing potential inhibitors, calculate relative degradation rates normalized to a standard substrate or condition .

What experimental design best reveals the developmental regulation of ClpX in the Chlamydia life cycle?

To reveal the developmental regulation of ClpX across the Chlamydia life cycle, researchers should employ a temporal sampling approach with synchronized infections. Begin by infecting host cells (typically epithelial cells) with purified elementary bodies (EBs) at a defined multiplicity of infection, then remove unattached EBs after a short adsorption period to synchronize the infection. Collect samples at key timepoints spanning the developmental cycle: early (2-6 hours post-infection, representing EB-to-RB conversion), mid (12-24 hours, active RB replication), and late (30-48 hours, RB-to-EB conversion and EB release). For each timepoint, analyze both ClpX expression levels (using quantitative RT-PCR for mRNA and western blotting for protein) and activity (using cell lysates in ATP-dependent degradation assays with model substrates). Immunofluorescence microscopy with anti-ClpX antibodies can reveal the subcellular localization of ClpX at different developmental stages. To assess ClpX function during development, inducible expression systems can introduce wild-type or dominant-negative ClpX at specific timepoints, followed by analysis of developmental progression using markers for EBs and RBs. Complementary approaches include proteomic analysis to identify changes in the abundance of potential ClpX substrates across the developmental cycle, and ChIP-seq to identify potential transcriptional regulators of clpX expression. This multi-faceted approach can provide a comprehensive view of how ClpX expression, activity, and function are regulated throughout the chlamydial developmental cycle .

How can researchers develop and validate specific inhibitors of C. caviae ClpX?

Developing and validating specific inhibitors of C. caviae ClpX requires a systematic approach spanning in silico, in vitro, and in vivo methods. Begin with structure-based virtual screening using the crystal or cryo-EM structure of ClpX (or a homology model if specific C. caviae structures are unavailable) to identify compounds predicted to bind to functional sites such as the ATP-binding pocket or ClpP interaction interface. Alternatively, high-throughput screening of diverse chemical libraries using ATPase activity assays can identify inhibitory compounds. Once potential inhibitors are identified, validate their direct binding to ClpX using biophysical methods such as isothermal titration calorimetry, surface plasmon resonance, or thermal shift assays. Assess functional inhibition through in vitro assays measuring ATPase activity and substrate degradation, establishing dose-response relationships and IC50 values. Determine specificity by testing activity against related AAA+ ATPases and human homologs. For the most promising candidates, assess cellular activity by measuring their ability to inhibit chlamydial growth in infected cell cultures, correlating growth inhibition with molecular effects on ClpX function. Develop structure-activity relationships by testing analogs of lead compounds to optimize potency and specificity. Finally, evaluate the most promising inhibitors in animal models of chlamydial infection, assessing both efficacy in reducing bacterial burden and potential toxicity. Throughout this process, medicinal chemistry approaches can improve pharmacokinetic properties and reduce potential off-target effects .

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.