May play a role in immune function. Likely involved in the processing of antigenic peptides during MHC class II-mediated antigen presentation.
KEGG: xla:373572
UniGene: Xl.35410
Cathepsin E (CTSE) in Xenopus laevis is an aspartic protease that belongs to the peptidase A1 family. It is expressed as a precursor that undergoes processing to form the mature enzyme. The recombinant form typically contains amino acid residues 53-397 of the native protein sequence. The protein's full sequence includes multiple functional domains, with the active site containing two catalytic aspartic acid residues that are essential for its proteolytic activity . Unlike some other cathepsins that are cysteine proteases, Cathepsin E is distinctly an aspartic protease with specific substrate preferences and cellular localization patterns.
Recombinant Xenopus laevis Cathepsin E can be expressed in multiple heterologous systems, with yeast being particularly efficient for eukaryotic protein expression. The yeast expression system offers an economical and efficient eukaryotic platform for secreted protein production that maintains proper folding and post-translational modifications . Other expression systems include:
Bacterial systems (E. coli) - Generally used for truncated versions (AA 57-363)
Mammalian cell systems (HEK-293 cells) - Used for full-length or near full-length versions
Insect cell systems (baculovirus-infected) - Often used for complex proteins requiring specific modifications
Each system offers different advantages regarding protein yield, post-translational modifications, and biological activity .
For optimal expression of recombinant Xenopus laevis Cathepsin E in yeast systems, researchers should consider:
Media composition: Typically, yeast minimal medium supplemented with appropriate amino acids and carbon source
Induction conditions: For inducible promoters like GAL1, galactose at 0.5-2% concentration
Temperature: 25-30°C, with lower temperatures (25°C) often yielding better folded protein
Duration: 24-72 hours post-induction, monitoring protein expression levels
pH: Maintain between 5.5-6.5 for optimal yeast growth and protein stability
Aeration: Adequate aeration through vigorous shaking (200-250 rpm)
Cell density: Induction at mid-log phase (OD600 of 0.8-1.2)
The yeast expression system provides advantages for secreted proteins like Cathepsin E, allowing for efficient production with appropriate post-translational modifications .
To optimize purification of His-tagged Xenopus laevis Cathepsin E while preserving enzymatic activity:
Lysis conditions: Use gentle lysis methods (for yeast cells, enzymatic digestion of cell wall followed by osmotic shock) in buffers containing 20-50 mM sodium phosphate, pH 7.4, 300 mM NaCl
Protease inhibitors: Include specific protease inhibitors, avoiding those that might affect aspartic proteases
IMAC purification:
Use Ni-NTA or TALON resins
Include 10-20 mM imidazole in binding buffer to reduce non-specific binding
Elute with 250-300 mM imidazole gradient
Maintain pH 7.4-8.0 during binding and elution
Buffer optimization:
After purification, exchange buffer to optimal pH for storage (typically pH 5.5-6.5)
Include stabilizers such as glycerol (10-20%)
Consider adding low concentrations of reducing agents (1-2 mM DTT or β-mercaptoethanol)
Activity preservation:
Avoid freeze-thaw cycles (aliquot before freezing)
Store at -80°C for long-term storage
For working stocks, store at 4°C with appropriate preservatives
Purification protocols should be validated using activity assays to ensure the enzyme maintains its catalytic function throughout the process .
Cathepsin E expression in Xenopus laevis follows specific temporal and spatial patterns during embryonic development, though it has not been as extensively characterized as some other developmental proteins. Based on proteomic studies of Xenopus embryogenesis:
Temporal regulation: Like many developmental proteins, Cathepsin E likely shows stage-specific expression patterns during the transition from fertilized egg to neurula embryo .
Post-MBT expression changes: The midblastula transition (MBT) represents a critical checkpoint when maternal proteins decline and zygotic gene expression increases. Similar to documented changes in DNA replication factors after MBT, Cathepsin E may undergo significant expression changes during this transition .
Cell-type specificity: As embryonic cells differentiate, Cathepsin E expression may become restricted to specific tissues or cell lineages.
Epigenetic regulation: Histone modifications, which have been extensively characterized in Xenopus development, likely play a role in regulating Cathepsin E expression across developmental stages. The transitions between "poised" chromatin states and active transcription likely influence Cathepsin E expression patterns .
More specific expression profiling would require targeted studies focusing on Cathepsin E during different developmental stages.
To effectively study Cathepsin E function during Xenopus laevis development, researchers can employ the following techniques:
Quantitative proteomics:
Loss-of-function studies:
Morpholino antisense oligonucleotides for targeted knockdown
CRISPR-Cas9 genome editing for genetic knockout
Small molecule inhibitors specific for aspartic proteases
Gain-of-function studies:
Activity assays:
Fluorogenic peptide substrates to monitor enzymatic activity
In situ zymography to visualize protease activity in tissue sections
Localization studies:
Whole-mount immunohistochemistry
Fluorescent protein tagging for live imaging
In situ hybridization for mRNA localization
Protein interaction studies:
Co-immunoprecipitation
Proximity labeling techniques
Yeast two-hybrid screening
These approaches can be combined to develop a comprehensive understanding of Cathepsin E function in developmental processes .
Advanced mass spectrometry approaches for characterizing post-translational modifications (PTMs) of Xenopus laevis Cathepsin E include:
Bottom-up proteomics:
Enzymatic digestion with trypsin or other proteases
LC-MS/MS analysis with Collision-Induced Dissociation (CID)
Parallel Reaction Monitoring (PRM) for targeted analysis of known modifications
Middle-down approach:
PTM enrichment strategies:
Titanium dioxide chromatography for phosphopeptide enrichment
Lectin affinity chromatography for glycopeptides
Antibody-based enrichment for specific modifications
Quantitative PTM analysis:
Intact protein analysis:
Top-down proteomics using high-resolution instruments
Native MS to preserve protein conformation and some non-covalent interactions
When implementing these approaches, careful sample preparation including protease and phosphatase inhibitors is essential to preserve the native modification state .
Designing experiments to identify physiological substrates of Cathepsin E in Xenopus laevis tissues requires a multi-faceted approach:
Degradomics approaches:
N-terminomics to identify protein cleavage sites (TAILS - Terminal Amine Isotopic Labeling of Substrates)
PICS (Proteomic Identification of protease Cleavage Sites)
Global protein stability profiling in the presence/absence of Cathepsin E
Activity-based protein profiling:
Synthesize activity-based probes specific for Cathepsin E
Apply to tissue lysates or living embryos at different developmental stages
Identify labeled proteins by mass spectrometry
Comparative proteomics:
In vitro validation:
Express and purify candidate substrates
Perform in vitro cleavage assays with recombinant Cathepsin E
Identify cleavage sites by Edman sequencing or mass spectrometry
In vivo validation:
Spatial correlation analysis:
Compare the tissue distribution of Cathepsin E with potential substrates
Employ laser capture microdissection coupled with proteomics
These methods can be applied across different developmental stages to map the dynamic substrate landscape of Cathepsin E throughout Xenopus development .
When encountering low enzymatic activity of recombinant Xenopus laevis Cathepsin E, consider the following troubleshooting strategies:
Expression system optimization:
Protein folding and processing:
Ensure proper pro-enzyme processing (Cathepsin E requires activation from its zymogen form)
Optimize cultivation temperature (lower temperatures often improve folding)
Co-express molecular chaperones if using bacterial systems
Buffer optimization:
Test activity across pH range (typically pH 3.5-5.5 for aspartic proteases)
Evaluate different buffer systems (acetate, citrate, phosphate)
Optimize ionic strength (typically 50-200 mM)
Test different reducing agents (DTT, TCEP) at various concentrations
Substrate considerations:
Ensure using appropriate substrates (peptides containing Phe-Phe or similar hydrophobic residues)
Optimize substrate concentration to prevent inhibition at high concentrations
Consider fluorogenic substrates for greater sensitivity
Activation procedures:
Test acid activation protocols (brief exposure to pH 3.0-4.0)
Evaluate auto-activation kinetics at different temperatures
Consider limited proteolysis to remove pro-peptides if auto-activation is inefficient
Activity assay optimization:
Increase sensitivity by using optimal fluorophore/quencher pairs
Extend incubation times for slow-acting enzymes
Control for background proteolysis with specific inhibitors
By systematically addressing these factors, researchers can significantly improve the activity yield of recombinant Cathepsin E .
When facing non-specific binding issues with anti-Cathepsin E antibodies in Xenopus laevis samples, implement these troubleshooting strategies:
Antibody selection:
Use antibodies specifically validated in Xenopus laevis systems
Consider generating custom antibodies against Xenopus-specific epitopes
Evaluate multiple antibodies targeting different epitopes
Blocking optimization:
Test different blocking agents (BSA, casein, non-fat dry milk, fish gelatin)
Increase blocking time (overnight at 4°C may reduce background)
Include detergents (0.1-0.3% Triton X-100 or Tween-20) in blocking solutions
Sample preparation refinements:
Optimize fixation protocols (duration, fixative concentration, temperature)
Perform antigen retrieval if using fixed tissues
Pre-absorb antibodies with Xenopus tissue lysates lacking Cathepsin E
Antibody incubation conditions:
Reduce antibody concentration
Extend incubation time at lower temperatures (4°C)
Add non-ionic detergents and carrier proteins to antibody dilutions
Validation controls:
Signal amplification alternatives:
Test different detection systems (direct fluorescence vs. enzymatic)
Consider tyramide signal amplification for weak signals
Use monovalent Fab fragments instead of complete IgGs
Western blot specific strategies:
These strategies should be tested systematically, changing one variable at a time to identify optimal conditions .
When analyzing Cathepsin E expression changes across Xenopus laevis developmental stages, consider these statistical approaches:
Normalization strategies:
Time-series analysis methods:
ANOVA with post-hoc tests for multi-stage comparisons
Linear mixed-effects models to account for biological variability
Functional data analysis for continuous developmental trajectories
Principal component analysis to identify major patterns of variation
Clustering approaches:
Differential expression analysis:
Limma-based approaches for protein expression data
Bayesian methods to handle missing values common in proteomics
Multiple testing correction (FDR or Bonferroni) to control false positives
Visualization techniques:
Heat maps with hierarchical clustering
Line plots for temporal profiles
Volcano plots for significance assessment
Interactive visualization tools for exploring complex datasets
Integration with other data types:
Correlation with transcriptomic data
Pathway enrichment analysis to identify biological processes
Network analysis to identify regulatory relationships
These approaches should be adapted based on experimental design, sample size, and the specific hypotheses being tested .
When facing contradictory results between Cathepsin E activity assays and expression levels in developmental studies, consider these interpretation frameworks:
Post-translational regulation mechanisms:
Inhibitor dynamics:
Endogenous inhibitors might be stage-specifically expressed
Quantify inhibitor levels using proteomics approaches
Test tissue extracts for inhibitory activity against recombinant enzyme
Compartmentalization effects:
Subcellular localization changes can alter substrate accessibility
Perform fractionation studies to track Cathepsin E distribution
Use immunolocalization to confirm compartmentalization in intact tissues
Substrate availability:
Varying substrate levels across developmental stages
Perform targeted metabolomics or proteomics to track substrate availability
Consider competitive interactions with other proteases
Technical considerations:
Assay pH conditions may not reflect in vivo microenvironment
Activity assays may detect other proteases with overlapping specificity
Expression quantification may be affected by extraction efficiency
Integrative analysis approaches:
Single-embryo analysis:
Understanding these potential mechanisms can help reconcile seemingly contradictory results and provide deeper insights into Cathepsin E regulation during development .