Recombinant Peptidyl-prolyl cis-trans isomerase C (ppiC)

Shipped with Ice Packs
In Stock

Description

Antioxidant Properties

Recombinant PPIC demonstrates protective effects against oxidative stress:

  • ROS Reduction: At 0.001–1 µg/ml, it decreases hydrogen peroxide (H₂O₂)-induced reactive oxygen species (ROS) in HepG2 cells .

  • Enzyme Activation: Enhances catalase (CAT), glutathione peroxidase (GPx), and thioredoxin reductase (TRR) activities, critical for ROS scavenging .

  • Gene Expression: Upregulates CAT, GPx, and TRR mRNA levels, suggesting modulation of the Keap1/Nrf2/ARE pathway .

Immunological Regulation

  • iNKT Cell Development: Murine studies show Ppic deficiency reduces invariant Natural Killer T (iNKT) cell numbers in the thymus and periphery, implicating PPIC in immune cell differentiation .

  • Calcineurin/NFAT Pathway: PPIC interacts with cyclophilin C-associated protein (CyCAP) to activate microglia and macrophages via this pathway .

Therapeutic Potential

  • Cancer Biomarker: PPIC is overexpressed in epithelial ovarian cancer (EOC) and may serve as a circulating tumor cell marker .

  • Neuroprotection: PPIC-CyCAP complexes mitigate ischemic brain damage by enhancing survival mechanisms .

  • Cardioprotection: Cyclophilins, including PPIC, are activated during ischemia-reperfusion injury, suggesting roles in tissue repair .

Antioxidant Enzyme Activities

PPIC Concentration (µg/ml)CAT ActivityGPx ActivityTRR ActivitySOD Activity
0.001↑ 45%↑ 38%↑ 28%
0.01↑ 60%↑ 52%↑ 45%↑ 30%
1↑ 53%
Data from HepG2 cells treated with H₂O₂ and recombinant PPIC .

Interaction Network

Recombinant PPIC binds multiple partners, including:

  • Cyclosporine A (CsA): Forms immunosuppressive complexes to inhibit calcineurin .

  • CyCAP: Modulates microglial activation .

  • NFATc1: Links PPIC to transcriptional regulation in immune cells .

Limitations and Future Directions

While recombinant PPIC shows promise in antioxidant and immunomodulatory studies, challenges include:

  • Concentration-Dependent Effects: High doses (e.g., 1 µg/ml) may saturate pathways, reducing efficacy .

  • Mechanistic Gaps: The exact role of PPIC in cancer progression remains unclear .
    Further studies are needed to explore its in vivo therapeutic potential and interactions with redox-sensitive pathways like PRDX-TRR .

Product Specs

Form
Lyophilized powder. We will preferentially ship the format we have in stock. If you have special format requirements, please note them when ordering, and we will fulfill your request.
Lead Time
Delivery times vary by purchase method and location. Consult your local distributor for specific delivery times. All proteins are shipped with normal blue ice packs by default. For dry ice shipping, please contact us in advance; extra fees will apply.
Notes
Avoid repeated freezing and thawing. Store working aliquots at 4°C for up to one week.
Reconstitution
Briefly centrifuge the vial before opening to collect contents at the bottom. 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 default final glycerol concentration is 50%.
Shelf Life
Shelf life depends on several factors: storage state, buffer ingredients, storage temperature, and protein stability. Generally, the liquid form has a 6-month shelf life at -20°C/-80°C. The lyophilized form has a 12-month shelf life at -20°C/-80°C.
Storage Condition
Store at -20°C/-80°C upon receipt. Aliquot for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
The tag type will be determined during the manufacturing process. If you require a specific tag type, please inform us, and we will prioritize developing it.
Synonyms
ppiC; Z5286; ECs4709; Peptidyl-prolyl cis-trans isomerase C; PPIase C; EC 5.2.1.8; Parvulin; Rotamase C
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
2-93
Protein Length
Full Length of Mature Protein
Purity
>85% (SDS-PAGE)
Species
Escherichia coli O157:H7
Target Names
ppiC
Target Protein Sequence
AKTAAALHI LVKEEKLALD LLEQIKNGAD FGKLAKKHSI CPSGKRGGDL GEFRQGQMVP AFDKVVFSCP VLEPTGPLHT QFGYHIIKVL YRN
Uniprot No.

Target Background

Function
PPIases accelerate protein folding. They prefer amino acid residues with hydrophobic side chains, such as leucine and phenylalanine, in the P1 position of peptide substrates.
Database Links

KEGG: ece:Z5286

STRING: 155864.Z5286

Protein Families
PpiC/parvulin rotamase family
Subcellular Location
Cytoplasm.

Q&A

What is peptidyl-prolyl cis-trans isomerase C and what is its primary function?

Peptidyl-prolyl cis-trans isomerase C (ppiC) belongs to a family of enzymes that catalyze the cis/trans isomerization of peptidyl-prolyl peptide bonds in proteins and oligopeptides. This isomerization represents a critical rate-limiting step in protein folding processes. The primary function of ppiC is to accelerate protein folding by catalyzing the interconversion between cis and trans conformations of the peptide bond preceding proline residues. This catalytic activity is essential for generating properly folded, functionally active proteins in both prokaryotic and eukaryotic systems .

The isomerization reaction catalyzed by ppiC involves a 180° rotation around the peptidyl-prolyl bond, which significantly impacts the three-dimensional structure of proteins. Without PPIases like ppiC, some denatured proteins would refold extremely slowly (minutes to hours), whereas the presence of these enzymes can dramatically accelerate the process to milliseconds or seconds .

How does ppiC differ from other members of the PPIase family?

The PPIase superfamily consists of three main families: cyclophilins (Cyps), FK506-binding proteins (FKBPs), and parvulins. Each family has distinct structural features, substrate specificities, and inhibitor sensitivities:

  • Cyclophilins: These are receptors for the immunosuppressive drug cyclosporin A (CsA). They have a broad substrate specificity and are abundant in cells, constituting approximately 0.4% of cellular dry mass .

  • FK506-binding proteins: These bind to immunosuppressive drugs FK506 and rapamycin. They often contain additional functional domains that mediate various cellular processes .

  • Parvulins: The family to which ppiC typically belongs. These enzymes show distinct substrate preferences compared to the other families and are not sensitive to CsA or FK506 .

ppiC has specific structural features and catalytic properties that distinguish it from other PPIases, including substrate specificity toward particular proline-containing sequence motifs. Understanding these differences is crucial for developing specific research applications or therapeutic targeting strategies.

What standard assays are used to measure ppiC enzymatic activity?

Several methodological approaches are available to measure ppiC enzymatic activity:

Spectrophotometric Assay:
The most common assay uses chromogenic peptide substrates containing a proline residue (e.g., succinyl-Ala-Phe-Pro-Phe-4-nitroanilide). The reaction sequence involves:

  • Allowing the substrate to reach cis/trans equilibrium

  • Adding ppiC to catalyze isomerization

  • Adding chymotrypsin, which specifically cleaves after the proline residue only in the trans conformation

  • Measuring the released 4-nitroanilide by monitoring absorbance at 390 nm

The specificity constant can be calculated from this assay, with active PPIases typically showing values in the range of 10^7 M^-1s^-1. For example, NifM from A. vinelandii showed a specificity constant of 1.09 × 10^7 M^-1s^-1 .

Rheological Measurements:
For assessing the impact of ppiC on extracellular matrix (ECM) properties, rheological measurements can be used to evaluate:

  • Storage modulus (stiffness)

  • Gelation dynamics

  • Self-healing properties of biomaterials

Cell Surface PPIase Activity Assay:
A specialized assay for detecting PPIase activity on living cell surfaces can be correlated with ECM development and cellular physiological states .

What expression systems are most effective for producing recombinant ppiC with high activity?

The optimal expression system for recombinant ppiC production depends on research objectives and downstream applications. Based on established protocols for similar PPIases, the following systems have proven effective:

Bacterial Expression (E. coli):

  • Most commonly used for PPIases due to high yield and simplicity

  • Typical vectors include pET series (especially pET21d+) with T7 promoter and lac operator for inducible expression

  • Expression can be induced using IPTG

  • Fusion tags like 6×His facilitate purification

Protocol outline for E. coli expression:

  • PCR amplification of the ppiC gene

  • Cloning into expression vector downstream of T7 promoter

  • Transformation into expression host (typically BL21(DE3) or similar)

  • Culture growth to optimal density followed by IPTG induction

  • Cell harvesting and lysis

  • Protein purification

Yeast Expression Systems:

  • Provide eukaryotic post-translational modifications

  • Suitable when proper folding is challenging in bacterial systems

  • S. cerevisiae or P. pastoris are commonly used hosts

Mammalian Cell Expression:

  • For applications requiring mammalian-specific modifications

  • HEK293 or CHO cells are preferred for complex proteins

Yield optimization factors include growth temperature (often reduced to 16-25°C during induction), induction time, and media composition. Codon optimization of the ppiC sequence for the host organism can significantly improve expression levels.

What purification strategies yield the highest purity and activity for recombinant ppiC?

A multi-step purification protocol typically yields the highest purity and activity for recombinant ppiC:

Immobilized Metal Affinity Chromatography (IMAC):

  • Primary purification step for His-tagged ppiC

  • Ni-NTA or Co-based resins are commonly used

  • Imidazole gradient elution improves purity

  • Typical yield: 10-20 mg protein per liter of bacterial culture

Size Exclusion Chromatography (SEC):

  • Secondary purification step for separating monomeric ppiC from aggregates

  • Also provides information on the oligomeric state of ppiC (typically monomeric in its native form)

Ion Exchange Chromatography:

  • Optional additional step for removing remaining impurities

  • Selection of cation or anion exchange depends on ppiC's isoelectric point

Quality Control Methods:

  • SDS-PAGE and Western blotting with anti-His or specific anti-ppiC antibodies

  • Activity assays using chromogenic substrates

  • Circular dichroism to confirm proper secondary structure

  • Fluorescence spectroscopy to verify tertiary structure integrity

The purification protocol should include protease inhibitors throughout the process, and care should be taken to maintain an appropriate buffer system to preserve activity. Typically, a phosphate or Tris buffer at pH 7.0-8.0 with 100-150 mM NaCl provides a suitable environment for ppiC stability.

How can researchers verify the proper folding and structural integrity of purified recombinant ppiC?

Verification of proper folding and structural integrity of recombinant ppiC involves several complementary biophysical techniques:

Circular Dichroism (CD) Spectroscopy:

  • Far-UV CD (190-260 nm) reveals secondary structure content

  • Properly folded ppiC typically shows characteristic negative minima at 208 nm and 222 nm, indicating alpha-helical content

  • Thermal denaturation studies can assess protein stability

Fluorescence Spectroscopy:

  • Intrinsic tryptophan fluorescence indicates tertiary structure integrity

  • Emission maximum shifts reflect the local environment of aromatic residues

  • Properly folded protein shows defined fluorescence spectrum that changes upon denaturation

Size Exclusion Chromatography (SEC):

  • Confirms homogeneity and oligomeric state

  • Recombinant ppiC typically exists as a monomer in its native state

Enzymatic Activity Assay:

  • The most functional verification of proper folding

  • Standard protease-coupled assay using chromogenic peptides

  • Active site integrity can be confirmed by inhibitor binding studies

Thermal Shift Assay (Differential Scanning Fluorimetry):

  • Measures protein stability and can detect if ligands or substrates bind to the protein

  • Uses fluorescent dyes that bind to hydrophobic regions exposed during unfolding

A properly folded, active recombinant ppiC should demonstrate both the expected structural characteristics by spectroscopic methods and catalytic activity comparable to the native enzyme.

How can recombinant ppiC be used to study extracellular matrix (ECM) dynamics?

Recombinant ppiC can serve as a valuable tool for investigating ECM dynamics through several experimental approaches:

Rheological Analysis of ECM Biomaterials:
Researchers can directly assess the impact of ppiC on the mechanical properties of various ECM components:

  • Fibrin gelation: Addition of recombinant ppiC (1-10 μM) to fibrinogen before thrombin-induced polymerization can significantly enhance the storage modulus (stiffness) of the resulting hydrogel

  • Collagen gelation: Both pH-induced and temperature-induced gelation of collagen can be modulated by ppiC, affecting matrix organization

  • Self-healing properties: ppiC influences the recovery rate and extent after mechanical disruption of ECM networks

Cell-ECM Interaction Assays:

  • Viscosity measurements of cell-protein mixtures: ppiC can alter the interaction between cells and ECM proteins, detectable through changes in the viscometric properties of the suspension

  • Adhesion dynamics: Controlling ppiC activity (through addition of recombinant protein or specific inhibitors) during cell-ECM attachment can reveal kinetic aspects of this interaction that are prolyl isomerization-dependent

ECM Remodeling Studies:
Recombinant ppiC can be used to understand how prolyl isomerization contributes to:

  • Folding and assembly of ECM proteins

  • Structural dynamics of dense polymer networks

  • Temporal control of ECM maturation

This approach offers unique insights by isolating prolyl isomerization from other dynamic events in the complex ECM environment, providing a molecular-level understanding of matrix regulation.

What methods can be used to study the interaction between ppiC and potential binding partners?

Several methods are available to characterize interactions between ppiC and its binding partners:

Co-immunoprecipitation (Co-IP):

  • Uses antibodies against ppiC or the suspected binding partner

  • Can identify novel protein-protein interactions in cellular contexts

  • Western blotting confirms the identity of co-precipitated proteins

Surface Plasmon Resonance (SPR):

  • Provides real-time binding kinetics (kon and koff rates)

  • Quantifies binding affinity (KD values)

  • Experimental setup:

    • Immobilize purified recombinant ppiC on a sensor chip

    • Flow potential binding partners over the surface

    • Measure association and dissociation phases

    • Analyze data to determine binding constants

Isothermal Titration Calorimetry (ITC):

  • Measures thermodynamic parameters of binding

  • Provides KD, stoichiometry, enthalpy (ΔH), and entropy (ΔS)

  • No labeling required, performed in solution

Fluorescence-based Assays:

  • Förster Resonance Energy Transfer (FRET)

  • Fluorescence polarization for studying smaller ligands

  • Microscale Thermophoresis (MST) for determining binding affinities

Pull-down Assays:

  • Using tagged recombinant ppiC (His-tag, GST, etc.)

  • Coupled with mass spectrometry for unbiased identification of binding partners

Yeast Two-Hybrid Screening:

  • For discovering novel protein interactions

  • Can be followed by validation using the methods above

These methods can reveal how ppiC interacts with substrates, inhibitors, or other cellular components, providing insights into its biological roles and regulatory mechanisms.

How does recombinant ppiC affect protein folding in experimental systems?

Recombinant ppiC can be used to study protein folding dynamics in several experimental paradigms:

In vitro Protein Refolding Assays:

  • Basic Protocol:

    • Denature model proteins containing proline residues (e.g., ribonuclease T1, staphylococcal nuclease)

    • Initiate refolding by dilution into native buffer conditions

    • Monitor refolding kinetics with and without recombinant ppiC

    • Detect folding using spectroscopic methods (fluorescence, CD, NMR)

  • Quantitative Analysis:

    • Measure folding rate acceleration

    • Determine concentration-dependent effects

    • Calculate catalytic efficiency (kcat/KM)

Pulse-Chase Experiments with Cell Extracts:

  • Label newly synthesized proteins

  • Add recombinant ppiC to cell extracts

  • Monitor acquisition of native structure over time

  • Compare folding rates and native protein yields

Single-Molecule Studies:

  • Using optical tweezers or atomic force microscopy

  • Directly observe the effect of ppiC on individual protein folding events

  • Measure force-extension curves with and without ppiC

Recombinant ppiC typically accelerates the refolding of proline-containing proteins, with the most pronounced effects observed for proteins with critical prolines in cis conformation in their native state. The degree of acceleration depends on:

  • The number and position of proline residues

  • The intrinsic isomerization rates of specific prolyl bonds

  • The structural context surrounding the proline residues

  • Concentration and activity of the recombinant ppiC

These studies help elucidate the role of prolyl isomerization as a rate-limiting step in protein folding and how ppiC can overcome this kinetic barrier.

How can site-directed mutagenesis be used to investigate the catalytic mechanism of ppiC?

Site-directed mutagenesis provides powerful insights into the catalytic mechanism of ppiC by allowing systematic modification of key residues. The following methodological approach has proven effective:

Key Residues for Mutagenesis:
Based on structural studies of PPIases, several conserved residues are typically targeted:

  • Catalytic site residues that directly participate in isomerization

  • Substrate-binding pocket residues that determine specificity

  • Residues involved in maintaining structural integrity

Experimental Workflow:

  • Design of Mutations:

    • Conservative substitutions (maintaining charge/polarity)

    • Non-conservative substitutions (altering properties)

    • Creation of catalytically inactive variants as controls

  • Generation of Mutants:

    • PCR-based site-directed mutagenesis

    • Verification by DNA sequencing

    • Expression and purification using identical conditions as wild-type

  • Functional Characterization:

    • Enzyme kinetics (kcat, KM, kcat/KM) using standard PPIase assays

    • Substrate specificity profiles using peptide libraries

    • Structural integrity confirmation by CD and fluorescence

  • Structure-Function Correlation:

    • Molecular dynamics simulations to predict effects of mutations

    • X-ray crystallography or NMR of mutant proteins

    • Correlation of structural changes with altered catalytic parameters

Table 1: Example Mutation Effects on PPIase Activity

MutationRegionActivity (% of WT)KM (μM)kcat (s⁻¹)kcat/KM (M⁻¹s⁻¹)
WT-10086.59401.09 × 10⁷
R55ACatalytic<10NDNDND
F60YBinding pocket65120.37105.9 × 10⁶
H94QSecondary shell8592.18309.0 × 10⁶

ND = Not determined due to very low activity
Note: Values in this table are representative examples based on similar PPIases

Through this approach, researchers can identify residues essential for catalysis versus those involved in substrate binding or structural stability, ultimately elucidating the catalytic mechanism of ppiC at the molecular level.

What is the role of ppiC in cellular stress responses and how can this be experimentally investigated?

PPIases including ppiC play crucial roles in cellular stress responses, particularly in protein quality control and adaptation to environmental challenges. Several experimental approaches can elucidate these functions:

Stress Response Induction Experiments:

  • Subject cells (with normal or altered ppiC levels) to various stressors:

    • Heat shock (37-42°C)

    • Oxidative stress (H₂O₂, paraquat)

    • ER stress (tunicamycin, thapsigargin)

    • Nutrient deprivation

  • Monitor survival, growth rates, and recovery kinetics

  • Measure stress marker expression through RT-qPCR or western blotting

Protein Aggregation and Misfolding Assays:

  • Use reporter proteins prone to misfolding (e.g., luciferase, GFP variants)

  • Analyze aggregation patterns with and without functional ppiC

  • Apply techniques such as:

    • Fluorescence microscopy for aggregate visualization

    • Filter trap assays for quantification

    • FRET-based folding sensors

Proteomics Approaches:

  • Comparative proteomics of wild-type vs. ppiC-deficient cells under stress

  • Pulse-chase experiments to track protein turnover rates

  • Identification of stress-responsive ppiC substrates through crosslinking and mass spectrometry

Transcriptional Response Analysis:

  • RNA-seq to identify genes differentially expressed in ppiC-deficient cells

  • ChIP-seq to detect changes in transcription factor binding

  • Analysis of stress-responsive promoter activities using reporter constructs

Dynamic Intracellular Localization:

  • Track ppiC localization during stress using fluorescent protein fusions

  • Co-localization studies with stress granules, processing bodies, or chaperone complexes

  • Live-cell imaging to monitor real-time responses

Research has shown that PPIases like ppiC may contribute to stress responses through:

  • Preventing aggregation of misfolded proteins

  • Facilitating assembly/disassembly of stress-responsive complexes

  • Modulating activity of transcription factors through conformational changes

  • Participating in DNA repair processes and cell cycle regulation under stress conditions

These experimental approaches provide complementary insights into the multifaceted roles of ppiC in cellular adaptation to stress.

How can researchers study the potential role of ppiC in modulating microtubule dynamics?

Investigating the role of ppiC in microtubule dynamics requires specialized approaches that connect enzymatic activity with cytoskeletal function:

In Vitro Microtubule Assembly Systems:

  • Purified Tubulin Polymerization Assays:

    • Measure polymerization kinetics of purified tubulin with/without recombinant ppiC

    • Monitor assembly by light scattering or fluorescence

    • Quantify parameters: nucleation rate, elongation rate, steady-state polymer mass

    • Experimental setup:

      • Incubate purified tubulin (10-20 μM) with GTP

      • Add varying concentrations of recombinant ppiC (0.1-10 μM)

      • Monitor turbidity at 350 nm over time

      • Calculate polymerization rates from the slopes of assembly curves

  • Microscopy-Based Analysis:

    • Total Internal Reflection Fluorescence (TIRF) microscopy of fluorescently-labeled microtubules

    • Measure:

      • Growth/shrinkage rates (μm/min)

      • Catastrophe frequency (transitions from growth to shrinkage)

      • Rescue frequency (transitions from shrinkage to growth)

      • Pause duration

Cellular Systems:

  • Live Cell Imaging:

    • Express fluorescent tubulin markers (e.g., GFP-α-tubulin)

    • Manipulate ppiC levels through:

      • Overexpression of wild-type or mutant ppiC

      • siRNA/shRNA-mediated knockdown

      • CRISPR/Cas9 gene editing

    • Track individual microtubule dynamics using spinning disk confocal microscopy

    • Quantify parameters using tracking software (e.g., plusTipTracker)

  • Fixed Cell Analysis:

    • Immunofluorescence staining of tubulin and associated proteins

    • Evaluate microtubule organization, density, and post-translational modifications

    • Analyze co-localization of ppiC with microtubules or regulatory factors

Biochemical Interaction Studies:

  • Co-sedimentation Assays:

    • Mix preformed microtubules with recombinant ppiC

    • Centrifuge to pellet microtubules and associated proteins

    • Analyze supernatant and pellet fractions by SDS-PAGE

    • Determine binding affinity from saturation curves

  • Identification of Tubulin or MAP Substrates:

    • Screen for proline-containing regions in tubulins and MAPs

    • Perform in vitro isomerization assays with synthetic peptides

    • Use mass spectrometry to detect cis/trans isomer ratios

This comprehensive approach can reveal whether ppiC influences microtubule dynamics directly through isomerization of tubulin subunits or indirectly via microtubule-associated proteins (MAPs) .

What are common challenges in obtaining active recombinant ppiC and how can they be overcome?

Researchers often encounter several challenges when producing active recombinant ppiC. Here are the most common issues and their solutions:

Low Expression Yields:

ChallengeSolution ApproachImplementation Details
Codon biasCodon optimizationAdapt codons to match host preference; commercially synthesize optimized gene
Toxicity to hostTightly regulated expressionUse tunable promoters; lower induction temperatures (16-25°C); reduce inducer concentration
Protein degradationProtease inhibitionAdd protease inhibitor cocktail; use protease-deficient host strains
Poor solubilitySolubility enhancementFuse with solubility tags (MBP, SUMO, TrxA); optimize buffer conditions

Improper Folding:

ChallengeSolution ApproachImplementation Details
Inclusion body formationRefolding protocolsSolubilize inclusions in urea/guanidine; perform gradual dialysis with redox buffers
Misfolded soluble proteinChaperone co-expressionCo-express with GroEL/GroES or trigger factor; add chemical chaperones to media
Disulfide bond issuesOxidative environmentExpress in E. coli Origami or SHuffle strains; add glutathione to refolding buffer

Loss of Activity During Purification:

ChallengeSolution ApproachImplementation Details
Protein instabilityBuffer optimizationScreen different pH values (6.5-8.0); add stabilizing agents (glycerol 10-20%, low concentrations of detergents)
Metal-induced inactivationChelator additionInclude EDTA (0.1-1 mM) in buffers; avoid metal-containing resins if problematic
Oxidative damageReducing agentsAdd DTT or β-mercaptoethanol (1-5 mM); handle under nitrogen atmosphere
Activity loss during storageCryoprotectionAdd glycerol (25-50%); flash-freeze in liquid nitrogen; avoid freeze-thaw cycles

Quality Control Strategies:

Implementing robust quality control at each stage can prevent downstream issues:

  • Verify gene sequence before expression

  • Use analytical SEC to confirm monodispersity

  • Perform activity assays immediately after purification and after storage

  • Validate proper folding using spectroscopic methods as described in section 2.3

By systematically addressing these challenges, researchers can significantly improve the yield, purity, and activity of recombinant ppiC preparations, ensuring reliable results in downstream applications.

How can researchers address contradictory results when studying ppiC functions in different experimental systems?

When confronted with contradictory results across different experimental systems, researchers should implement a systematic troubleshooting approach:

Analytical Framework for Resolving Contradictions:

  • System-Specific Variables Analysis:

    • Document all experimental conditions precisely

    • Create a comparative table of variables across systems:

      • Protein concentrations

      • Buffer compositions

      • Temperature and pH

      • Presence of cofactors or inhibitors

      • Cell types or strains used

      • Assay detection methods

  • Protein Quality Assessment:

    • Verify ppiC activity in each system using standardized assays

    • Confirm structural integrity through biophysical methods

    • Assess batch-to-batch variation with reference standards

    • Check for post-translational modifications across systems

  • Methodological Validation:

    • Implement positive and negative controls in each experiment

    • Use orthogonal methods to confirm key findings

    • Perform spike-in experiments to detect potential inhibitors

    • Cross-validate with different detection technologies

  • Reconciliation Strategies:

Contradiction TypeInvestigation ApproachResolution Strategy
Activity differencesEnzyme kinetics under varied conditionsIdentify system-specific factors affecting catalysis
Binding partner discrepanciesCross-validation with multiple interaction assaysMap condition-dependent interaction networks
Functional impact variationsDose-response studies across systemsDetermine threshold concentrations for effects
Subcellular localization conflictsLive-cell imaging with controlled expression levelsIdentify dynamic localization patterns
  • Statistical and Quantitative Analysis:

    • Apply rigorous statistical methods appropriate for each dataset

    • Consider Bayesian approaches to integrate conflicting data

    • Perform meta-analysis when multiple studies are available

    • Develop mathematical models to explain system-dependent behaviors

  • Biological Context Interpretation:

    • Consider that contradictions may reflect genuine biological complexity

    • Investigate tissue-specific or condition-dependent regulation

    • Examine potential redundancy with other PPIases

    • Explore context-dependent protein-protein interactions

By systematically analyzing contradictions, researchers can often transform apparent discrepancies into deeper insights about the context-dependent functions of ppiC, leading to more nuanced understanding of its biological roles.

What are the limitations of current methods for studying ppiC activity and how might these be overcome in future research?

Current methodologies for studying ppiC activity present several limitations that affect data interpretation and application. Understanding these constraints and emerging solutions is critical for advancing the field:

Current Limitations and Future Directions:

  • Spectroscopic Assay Limitations:

LimitationImpactEmerging Solutions
Artificial peptide substratesMay not reflect native substrate specificityDevelopment of protein-based substrates with fluorescent reporters at conformationally sensitive positions
Indirect measurement via coupled enzymesPotential for false positives/negatives due to interaction with coupling enzymeDirect measurement using NMR or mass spectrometry to detect cis/trans isomers without coupling enzymes
Limited throughputRestricts comprehensive substrate profilingAdaptation to microplate formats; development of continuous fluorescence-based assays
Poor performance in complex environmentsDifficult to measure activity in cellular contextsCell-penetrating fluorogenic substrates; genetically encoded sensors for intracellular activity
  • Structural and Mechanistic Understanding:

LimitationImpactEmerging Solutions
Limited structural data on enzyme-substrate complexesIncomplete understanding of catalytic mechanismTime-resolved crystallography; cryo-EM studies of conformational states
Difficulty capturing transition statesMissing critical mechanistic detailsComputational approaches (QM/MM); transition state analogs as probes
Challenges in distinguishing catalysis from bindingConfounding interpretation of mutational studiesSingle-molecule studies to directly observe catalytic events
  • Cellular and Physiological Context:

LimitationImpactEmerging Solutions
Redundancy among PPIase familiesCompensation masks phenotypes in knockout studiesDevelopment of highly specific inhibitors; CRISPR-based acute depletion strategies
Limited temporal resolutionMissing dynamic aspects of ppiC functionOptogenetic control of ppiC activity; degron-based rapid protein depletion
Difficulty identifying physiological substratesIncomplete understanding of biological rolesProximity labeling approaches (BioID, APEX); covalent trapping of enzyme-substrate complexes
Challenges in measuring ECM-related activitiesMissing important extracellular functionsDevelopment of ECM-specific reporters; advanced rheological techniques with molecular readouts
  • Integration with Systems Biology:

LimitationImpactEmerging Solutions
Isolation from broader cellular networksMissing regulatory contextMulti-omics approaches to connect ppiC function with proteome, transcriptome, and metabolome
Limited predictive modelsDifficulty extrapolating from in vitro to in vivoMachine learning approaches to integrate diverse datasets; network models of PPIase function

Transformative Methodologies on the Horizon:

  • Single-molecule FRET-based assays: Direct visualization of individual isomerization events in real-time

  • CRISPR-based screening: Systematic identification of genetic interactions and functional networks

  • Advanced imaging technologies: Super-resolution microscopy combined with specific probes for ppiC activity

  • In-cell NMR spectroscopy: Direct measurement of protein conformational changes in cellular environments

  • AI-driven experimental design: Optimization of experimental conditions based on machine learning predictions

These emerging approaches promise to overcome current limitations and provide deeper insights into the multifaceted roles of ppiC in biological systems, ultimately enabling more precise targeting for therapeutic applications .

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.