Knockout Models:
Transgenic Overexpression Models:
CTSC mice are pivotal in studying diseases linked to protease dysregulation.
CTSC mice enable preclinical evaluation of inhibitors:
AZD7986: A CTSC inhibitor tested in asthma models reduces airway inflammation and remodeling .
N-Acetyldopamine Dimers: Suppress CTSC activity, attenuating LPS-induced inflammation .
Research using CTSC mice often involves:
Activity Assays: Fluorogenic substrates (e.g., Gly-Arg-AMC) quantify CTSC activity in tissues .
Phenotyping: Histopathology for granuloma formation, immune cell infiltration, or organ damage .
Cathepsin C (CTSC) is a key enzyme involved in the activation of serine proteases found in granules within inflammatory cells. It specifically cleaves dipeptides from the N-terminal end of protein and peptide substrates. The activation of these proteases by CTSC enables them to break down components of the extracellular matrix. This process, while important for normal physiological functions, can contribute to tissue damage and chronic inflammation if not properly regulated. CTSC is a tetrameric enzyme, meaning it is composed of four subunits. Each subunit consists of three parts: an N-terminal proregion fragment, a heavy chain, and a light chain. Defects in the CTSC gene can lead to Papillon-Lefevre syndrome, an autosomal recessive disorder characterized by severe periodontal disease.
Recombinant CTSC protein derived from mouse has been produced in HEK293 cells. This protein is a single polypeptide chain consisting of 444 amino acids (amino acids 25-462), resulting in a molecular weight of 50.5 kDa. A 6-amino acid Histidine tag is present at the C-terminus to facilitate purification, which is carried out using proprietary chromatographic techniques.
The CTSC protein is supplied in a solution with a concentration of 0.5 mg/ml. The solution contains Phosphate Buffered Saline at a pH of 7.4 and 10% glycerol.
The purity of the protein is greater than 95% as determined by SDS-PAGE analysis.
The enzyme exhibits significant biological activity, exceeding 50,000 pmol/min/ug. This activity is measured as the amount of enzyme required to hydrolyze 1 picomole of GlyArg-AMC per minute at a pH of 6.0 and a temperature of 37°C.
DPPI, Cathepsin C, CTSC
HEK293 cells.
DTPANCTYPD LLGTWVFQVG PRSSRSDINC SVMEATEEKV VVHLKKLDTA YDELGNSGHF TLIYNQGFEI VLNDYKWFAF FKYEVRGHTA ISYCHETMTG WVHDVLGRNW ACFVGKKVES HIEKVNMNAA HLGGLQERYS ERLYTHNHNF VKAINTVQKS WTATAYKEYE KMSLRDLIRR SGHSQRIPRP KPAPMTDEIQ QQILNLPESW DWRNVQGVNY VSPVRNQESC GSCYSFASMG MLEARIRILT NNSQTPILSP QEVVSCSPYA QGCDGGFPYL IAGKYAQDFG VVEESCFPYT AKDSPCKPRE NCLRYYSSDY YYVGGFYGGC NEALMKLELV KHGPMAVAFE VHDDFLHYHS GIYHHTGLSD PFNPFELTNH AVLLVGYGRD PVTGIEYWII KNSWGSNWGE SGYFRIRRGT DECAIESIAV AAIPIPKL-HHHHHH
CTSC (Cathepsin C) is a widely expressed exo-cysteine protease involved in the proteolytic processing of various lysosomal enzymes. CTSC mouse models, particularly knockout models, are valuable for investigating the functional roles of this enzyme in both normal physiology and disease states.
CTSC knockout mice have proven essential for understanding how this protease contributes to inflammatory conditions, as CTSC activates neutrophil serine proteases including neutrophil elastase, cathepsin G, and proteinase 3 . These models allow researchers to examine the direct consequences of CTSC deficiency in vivo, providing insights that cannot be obtained through in vitro studies alone.
The importance of these models is highlighted in acute pancreatitis research, where CTSC deletion has been shown to reduce disease severity primarily by affecting neutrophil infiltration rather than direct intracellular protease activation .
CTSC is expressed in both mouse and human pancreatic tissue as well as in inflammatory cells, particularly neutrophils . The expression patterns show considerable homology between species, making mouse models relevant for translational research.
In mouse models, CTSC has demonstrated higher activity in metastatic tumors compared to primary tumors, mirroring patterns observed in human cancer progression . This conservation of function makes CTSC mouse models particularly valuable for cancer research, especially studies focused on metastasis.
While expression patterns are similar, some tissue-specific differences may exist between mouse and human CTSC expression. Researchers should validate findings in human samples when possible to confirm translational relevance.
When designing experiments with CTSC knockout mice, researchers should consider:
Strain background effects: The genetic background of your CTSC knockout mice can significantly influence phenotypic outcomes. Always use littermate controls to minimize variability .
Sample size calculations: Proper statistical power analysis is crucial. CTSC knockout effects may vary in magnitude across different disease models (e.g., more prominent in milder models of pancreatitis than severe ones) .
Environmental standardization: CTSC knockout phenotypes can be sensitive to environmental conditions. Control for housing conditions, diet, and handling procedures .
Time-dependent effects: In models like acute pancreatitis, CTSC knockout effects may vary at different time points. For example, neutrophil infiltration differences between wild-type and CTSC knockout mice were significant at 8 hours but not at 1 hour post-induction .
Cell-specific considerations: CTSC functions differently in various cell types. For instance, CTSC deletion affects neutrophil infiltration but not macrophage infiltration in pancreatitis models .
Following the 3Rs principles (Replacement, Refinement, and Reduction) will promote not only ethical animal use but also enhance reproducibility, the "4th R" in animal research .
Proper validation of CTSC knockout in mouse models requires a multi-faceted approach:
Genotyping: Confirm genetic deletion using PCR with appropriate primers that span the targeted region.
Protein expression analysis: Verify absence of CTSC protein using Western blotting or immunohistochemistry in relevant tissues.
Enzymatic activity assays: Measure CTSC activity using specific substrates. Recombinant mouse active CTSC can serve as a positive control for activity assays .
Functional validation: Assess downstream effects on CTSC-dependent processes, such as reduced activity of neutrophil serine proteases (elastase, cathepsin G, and proteinase 3) .
Phenotypic characterization: Document any baseline phenotypic differences between CTSC knockout and wild-type littermates before experimental intervention.
It's important to note that commercial CTSC knockout kits use CRISPR technology and typically involve knocking in a cassette downstream of the target gene . Researchers should verify the specific molecular details of their knockout model to ensure appropriate interpretation of results.
CTSC deficiency significantly modulates inflammatory responses in mouse models through several mechanisms:
Reduced neutrophil infiltration: In acute pancreatitis models, CTSC knockout mice show decreased neutrophil granulocyte infiltration into both the pancreas and lungs, as evidenced by reduced myeloperoxidase (MPO) activity and fewer Ly6g-positive cells in pancreatic tissue .
Impaired neutrophil protease activity: CTSC knockout reduces the activity of neutrophil serine proteases including elastase, cathepsin G, and proteinase 3, which are critical mediators of inflammatory tissue damage .
Attenuated E-cadherin cleavage: CTSC-deficient neutrophils show reduced capacity to cleave E-cadherin in adherens junctions between acinar cells, potentially preserving tissue integrity during inflammation .
Cell-specific effects: Importantly, while CTSC deletion decreases neutrophil infiltration, it does not affect macrophage infiltration, as demonstrated by unchanged CD68 and F4/80 antigen staining in pancreatic tissue .
These findings highlight CTSC's role as a key regulator of neutrophil-mediated inflammatory responses, with its absence conferring partial protection against inflammatory tissue damage.
CTSC plays significant roles in cancer progression and metastasis as demonstrated in mouse models:
Increased activity in metastatic tumors: Studies have shown that CTSC has higher activity in metastatic tumors compared to primary tumors, suggesting its involvement in the metastatic process .
Promotion of lung metastasis: CTSC promotes cancer cell proliferation in the lungs at early stages of breast cancer metastasis. This has been demonstrated in mouse models where CTSC activity is upregulated during breast cancer metastasis to the lungs .
Biomarker potential: In mouse models of breast cancer metastasis, serum CTSC activity increases progressively. By days 3, 7, 10, and 14 post-injection of cancer cells, the signal ratios between metastatic mice and normal mice were 1.74-fold, 2.26-fold, 2.61-fold, and 2.50-fold, respectively .
Imaging applications: Fluorescent CTSC substrates like YF-2 can identify metastatic tumors in vivo, with the lungs of mice bearing metastatic tumors showing 3.78-fold higher fluorescence signals compared to normal mice .
These findings suggest CTSC as both a potential therapeutic target and diagnostic marker for metastatic progression.
Advanced monitoring of CTSC activity in vivo can be achieved through several approaches:
Fluorescent substrate probes: Substrate-based probes like YF-2 that are activated by CTSC can be used for real-time visualization of enzyme activity. In metastatic breast cancer models, YF-2 showed progressively enhanced fluorescence signals in lungs with metastatic tumors, with peak intensity at 90 minutes post-injection .
Serum activity measurements: CTSC activity in serum can be assessed after incubation with activatable substrates. This approach has successfully detected increased CTSC activity in the serum of mice with breast cancer metastasis compared to normal mice .
Correlation with bioluminescence: In models using luciferase-expressing cancer cells, CTSC activity (measured by substrate fluorescence) can be correlated with tumor burden (measured by bioluminescence), allowing for multimodal validation of metastatic sites .
Tissue-specific activity analysis: Different organs show varying levels of CTSC activity during disease progression. For example, orthotopic breast tumors exhibited approximately fourfold higher fluorescence intensity than normal mammary glands 4 hours after YF-2 injection .
Ex vivo validation: Extracted tissues can be analyzed for CTSC activity and correlated with immunohistochemical staining for CTSC protein to confirm specificity .
These methods provide researchers with versatile tools to monitor CTSC activity longitudinally in disease models, enhancing our understanding of its dynamic roles in pathological processes.
Establishing CTSC conditional knockout mouse models presents several technical challenges that researchers should consider:
Design of targeting constructs: Creating cell-type specific CTSC knockout requires careful design of floxed alleles that don't interfere with normal expression before Cre-mediated recombination. The location of loxP sites must avoid disrupting regulatory elements while ensuring efficient excision .
Validation of conditional deletion efficiency: Unlike global knockouts, conditional models require verification of deletion efficiency in specific cell types, which may vary depending on the Cre driver used. Quantitative assessment of remaining CTSC expression is essential .
Temporal control considerations: When using inducible Cre systems (e.g., tamoxifen-inducible CreERT2), researchers must optimize induction protocols to achieve consistent deletion while minimizing off-target effects of the inducer.
Background leakiness: Some Cre driver lines may exhibit leaky expression in unintended tissues or developmental stages, requiring thorough characterization of the deletion pattern beyond the target tissue.
Compensatory mechanisms: Acute CTSC deletion may trigger compensatory upregulation of other cathepsins or related proteases that might not occur in global knockout models where adaptation happens developmentally.
Phenotypic analysis complexity: With tissue-specific deletion, phenotypes may be subtler or more complex than in global knockouts, requiring more sophisticated analysis techniques to detect and characterize effects.
Addressing these challenges requires careful experimental design, rigorous validation protocols, and consideration of appropriate controls, including Cre-only and floxed-only controls to account for potential Cre toxicity or insertional effects.
CTSC knockout mice exhibit variable phenotypes across different disease models, highlighting the context-specific roles of this protease:
These variable phenotypes underscore the importance of studying CTSC function across multiple disease contexts and timepoints to fully understand its biological roles.
Isolation and analysis of CTSC-expressing cells from mouse tissues require specialized protocols:
Tissue-specific isolation techniques:
For pancreatic acinar cells: Use collagenase digestion followed by gradient centrifugation to obtain pure populations
For neutrophils: Isolate from bone marrow using Percoll gradient separation or commercially available neutrophil isolation kits
For macrophages: Harvest from peritoneal cavity after thioglycollate stimulation or isolate from spleen using CD11b magnetic beads
Validation of cell purity:
Flow cytometry using cell-specific markers (e.g., Ly6G for neutrophils, F4/80 for macrophages)
Morphological assessment using cytospin preparations and Wright-Giemsa staining
CTSC activity measurement in isolated cells:
Analysis workflow:
Lyse cells in appropriate buffers with protease inhibitors (excluding cysteine protease inhibitors if measuring CTSC activity)
Adjust protein concentration for standardized comparison
Perform activity assays under optimal pH conditions (pH 5.5-6.0 for CTSC)
Follow enzyme kinetics over time rather than single endpoint measurements
Comparative analysis between cell types:
Normalize CTSC activity to cell number or total protein content
Account for cell type-specific baseline CTSC expression levels when comparing responses to stimuli
This methodological approach ensures reliable isolation and accurate assessment of CTSC expression and activity in different cell populations from mouse tissues.
Designing effective combination studies with CTSC inhibitors and knockout mice requires careful consideration of several factors:
Complementary experimental approaches:
Use CTSC knockout mice to establish baseline phenotypes and identify CTSC-dependent processes
Employ pharmacological inhibitors to assess acute inhibition effects and dose-response relationships
Compare outcomes to identify potential compensatory mechanisms in knockout models
Validation strategies:
Confirm inhibitor specificity by testing in both wild-type and CTSC knockout mice
Measure residual CTSC activity in tissues after inhibitor treatment
Assess inhibitor effects on downstream targets (e.g., neutrophil elastase activity)
Study design considerations:
Include appropriate control groups: wild-type + vehicle, wild-type + inhibitor, knockout + vehicle, knockout + inhibitor
Time inhibitor administration to target specific disease phases
Consider age and sex as variables that may affect inhibitor pharmacokinetics
Dosing optimization:
Establish inhibitor dose-response curves in wild-type mice before combination studies
Monitor for potential off-target effects at higher doses
Consider pharmacokinetic/pharmacodynamic modeling to optimize dosing regimens
Advanced applications:
Use tissue-specific or inducible CTSC knockout models with inhibitors to distinguish systemic from local effects
Consider genetic knockdown approaches (siRNA, shRNA) as an intermediate between pharmacological inhibition and genetic knockout
This integrated approach allows researchers to distinguish between developmental adaptations to CTSC absence (in knockout models) and acute CTSC inhibition effects (with pharmacological agents), providing more comprehensive insights into CTSC biology.
Translating findings from CTSC mouse models to human disease research involves several considerations:
Comparative biology assessment:
CTSC shows conserved expression patterns between mice and humans in many tissues, particularly in inflammatory cells
Neutrophil serine proteases activated by CTSC have similar functions across species, supporting translational relevance
Higher CTSC activity in metastatic tumors compared to primary tumors is observed in both mouse models and human cancers
Disease-specific translatability:
Translational research applications:
CTSC activity biomarkers developed in mouse models (e.g., serum activity measurements, imaging probes) show potential for clinical diagnostic applications
Therapeutic targeting strategies validated in mouse models provide rationale for human clinical trials
YF-2 substrate activation patterns in mouse breast cancer lung metastasis models suggest potential for human diagnostic imaging applications
Limitations in translation:
Mouse models may not fully recapitulate the complexity of human disease progression
Species differences in immune system composition and function may affect CTSC-dependent inflammatory processes
Metabolic and physiological differences between mice and humans necessitate careful interpretation of pharmacological studies
Despite these limitations, CTSC mouse models have provided valuable insights that have accelerated understanding of human disease mechanisms and identified potential therapeutic targets.
CTSC mouse research has revealed several promising therapeutic applications:
Cancer metastasis intervention:
Anti-inflammatory applications:
CTSC knockout mice show reduced severity in inflammatory conditions like pancreatitis
Selective CTSC inhibitors could modulate neutrophil-mediated tissue damage without broadly suppressing immune function
Particularly promising for acute inflammatory conditions where neutrophil elastase contributes to pathology
Diagnostic imaging development:
Activatable CTSC substrates like YF-2 effectively visualize metastatic tumors in mouse models
These agents show potential for translation into clinical diagnostic tools
The fourfold higher signal in orthotopic tumors versus normal tissue demonstrates favorable signal-to-noise ratio for imaging applications
Biomarker applications:
Combination therapy approaches:
CTSC inhibition could sensitize tumors to existing therapies by altering the tumor microenvironment
Combined targeting of multiple cathepsins informed by mouse studies may provide synergistic therapeutic effects
Cathepsin C (CTSC), also known as dipeptidyl peptidase I (DPPI), is a lysosomal cysteine protease belonging to the papain-like cysteine peptidases family . It plays a crucial role in the immune system by activating various serine proteases, which are essential for immune defenses, antimicrobial activity, inflammation, and programmed cell death (apoptosis) . Recombinant mouse Cathepsin C is a valuable tool for studying these processes in a controlled laboratory setting.
Cathepsin C is a tetrameric enzyme composed of four identical subunits, each containing a heavy chain, a light chain, and an exclusion domain . The enzyme sequentially removes dipeptides from the free N-termini of proteins and peptides, except when a basic amino acid (Arg or Lys) is in the N-terminal position or Pro is on either side of the scissile bond . This broad specificity allows Cathepsin C to participate in various physiological processes.
Cathepsin C is constitutively expressed at high levels in the lung, kidney, liver, and spleen . It is synthesized as an inactive proenzyme (zymogen) and undergoes proteolytic cleavage to become active . In the laboratory, recombinant mouse Cathepsin C is typically produced using a mouse myeloma cell line (NS0) and activated by recombinant human Cathepsin L .
Cathepsin C is pivotal in activating serine proteases in cytotoxic T cells, natural killer cells, mast cells, and neutrophils . These proteases include granzymes A and B, chymase, tryptase, cathepsin G, neutrophil elastase, and proteinase 3 . Dysregulation of Cathepsin C is linked to various inflammatory diseases, such as chronic obstructive pulmonary disease (COPD), asthma, cystic fibrosis, and COVID-19-related acute respiratory distress syndrome (ARDS) . Additionally, mutations in the CTSC gene are associated with genetic disorders like Papillon–Lefevre syndrome (PLS), Haim–Munk syndrome (HMS), and aggressive periodontitis (AP) .