CTSL Mouse

Cathepsin-L Mouse Recombinant
Shipped with Ice Packs
In Stock

Description

Definition and Genetic Background

CTSL Mouse refers to murine models with targeted modifications in the Ctsl gene, which encodes cathepsin L—a protease involved in protein degradation, antigen processing, and extracellular matrix remodeling. The Ctsl<sup>-/-</sup> strain lacks functional CTSL due to gene deletion or truncation, leading to distinct phenotypes in immunity and organ homeostasis .

SARS-CoV-2 and Coronaviruses

  • Mechanism: CTSL cleaves the SARS-CoV-2 spike protein, enhancing viral entry into host cells. Inhibiting CTSL (e.g., with E64d or amantadine) reduces pseudovirus infection in humanized mice .

  • Therapeutic Potential: Amantadine, an FDA-approved drug, suppresses CTSL activity and reduces viral load in vitro and in vivo .

Influenza A

  • Protective Role: Ctsl<sup>-/-</sup> mice exhibit higher mortality, prolonged viral loads, and impaired adaptive immunity (reduced CD4+ lymphocytes and pathogen-specific IgG) .

PathogenCTSL RoleOutcome in Ctsl<sup>-/-</sup> Mice
SARS-CoV-2Facilitates viral entryIncreased infection severity
Influenza ASupports immune defenseHigher mortality, delayed viral clearance

Bacterial Infections (Mycoplasma pulmonis)

  • Host Defense: Ctsl<sup>-/-</sup> mice show 247-fold higher lung bacterial burdens, severe neutrophilic pneumonia, and 22% mortality (vs. 0% in wild-type) .

  • Immune Dysregulation: Impaired lymphangiogenesis and reduced IFN-γ responses contribute to poor infection control .

Cancer and Tissue Remodeling

  • Tumor Progression: CTSL promotes extracellular matrix degradation, facilitating tumor invasion. Ctsl<sup>-/-</sup> mice exhibit resistance to skin carcinogenesis .

  • Cardiac and Epidermal Phenotypes: CTSL deficiency leads to dilated cardiomyopathy, hair cycle defects, and abnormal epidermal differentiation .

CTSL Inhibitors in Disease Models

CompoundTargetEffectStudy
E64dBroad cathepsinsReduces SARS-CoV-2 pseudovirus entry
AmantadineCTSL transcriptionInhibits viral infection in vitro and vivo
SID26681509Selective CTSLBlocks spike protein cleavage

Immune and Developmental Roles

  • Lymphocyte Regulation: Ctsl<sup>-/-</sup> mice have reduced CD4+ T cells in the thymus and periphery, impacting adaptive immunity .

  • Lymphangiogenesis: Impaired airway lymphatic remodeling in Ctsl<sup>-/-</sup> mice exacerbates mycoplasmal infection outcomes .

Product Specs

Introduction
Cathepsin-L, also called CTSL, belongs to the peptidase C1 family. This protein exists as a dimer, formed by disulfide bonds between heavy and light chains, both originating from a single precursor protein. CTSL functions as a lysosomal cysteine proteinase and plays a crucial role in intracellular protein breakdown. Its targets include collagen, elastin, and alpha-1 protease inhibitor, a key regulator of neutrophil elastase activity. CTSL has been linked to various pathological conditions, including muscle fiber breakdown in myopathies and myocardial ischemia, as well as the kidney's response to protein in urine. Different mRNA transcripts of CTSL exist due to alternative splicing.
Description
Recombinant CTSL from Mouse, produced in Sf9 insect cells using a Baculovirus expression system, is a single glycosylated polypeptide chain. It consists of 325 amino acids (18-334a.a.), resulting in a molecular weight of 36.8kDa. However, on SDS-PAGE, the apparent molecular size will be approximately 40-57kDa.
The CTSL protein is engineered with an 8 amino acid His tag at its C-terminus and purified using proprietary chromatographic methods.
Physical Appearance
A clear, colorless solution that has been sterilized by filtration.
Formulation
The CTSL protein solution is supplied at a concentration of 0.25mg/ml. It is prepared in a buffer consisting of Phosphate Buffered Saline (pH 7.4) and 10% glycerol.
Stability
For short-term storage (up to 2-4 weeks), keep the vial refrigerated at 4°C.
For extended storage, freeze the product at -20°C.
To ensure stability during long-term storage, adding a carrier protein (0.1% HSA or BSA) is recommended.
Minimize repeated freezing and thawing cycles.
Purity
The purity of the protein is greater than 90.0% as assessed by SDS-PAGE analysis.
Synonyms
Cathepsin L1, Cathepsin L, Major excreted protein, MEP, p39 cysteine proteinase.
Source
Sf9, Baculovirus cells.
Amino Acid Sequence
TPKFDQTFSA EWHQWKSTHR RLYGTNEEEW RRAIWEKNMR MIQLHNGEYS NGQHGFSMEM NAFGDMTNEE FRQVVNGYRH QKHKKGRLFQ EPLMLKIPKS VDWREKGCVT PVKNQGQCGS CWAFSASGCL EGQMFLKTGK LISLSEQNLV DCSHAQGNQG CNGGLMDFAF QYIKENGGLD SEESYPYEAK DGSCKYRAEF AVANDTGFVD IPQQEKALMK AVATVGPISV AMDASHPSLQ FYSSGIYYEP NCSSKNLDHG VLLVGYGYEG TDSNKNKYWL VKNSWGSEWG MEGYIKIAKD RDNHCGLATA ASYPVVNLEH HHHHH.

Q&A

What is Cathepsin L and why is it significant in mouse models?

Cathepsin L (CTSL) is a cysteine protease that plays crucial roles in protein degradation and cellular processes. In mouse models, CTSL has emerged as a significant factor in studying viral pathogenesis, particularly SARS-CoV-2 infection. Research demonstrates that CTSL functionally cleaves the SARS-CoV-2 spike protein, enhancing virus entry into cells . Mouse models provide valuable insights into CTSL's physiological and pathological functions because the enzyme's activity can be manipulated through genetic approaches (knockout models) or pharmacological inhibition, allowing researchers to assess its contribution to disease mechanisms under controlled conditions .

How do CTSL expression patterns differ between mouse strains?

CTSL expression varies between mouse strains, which can significantly impact experimental outcomes. Wild-type mice typically express CTSL in multiple tissues, with notable presence in the liver, lungs, and kidneys. Specialized mouse models like the Lepr db/db diabetic mice show altered CTSL expression patterns that correspond with metabolic dysfunction . When designing experiments, researchers should consider strain-specific baseline CTSL expression levels, as these variations can affect data interpretation, especially in comparative studies investigating CTSL's role in pathological conditions. Documentation of strain-specific expression profiles is essential before initiating CTSL-targeted interventions to ensure accurate attribution of observed effects to experimental manipulations rather than inherent strain differences .

What are the key phenotypic characteristics of CTSL knockout mice?

CTSL knockout (CTSL-/-) mice display several distinctive phenotypic characteristics that inform researchers about the enzyme's physiological functions:

  • Reduced susceptibility to SARS-CoV-2 pseudovirus infection, demonstrating CTSL's critical role in viral entry mechanisms

  • Altered immune responses, particularly affecting T-cell selection and development

  • Periodic hair loss and epidermal hyperplasia, indicating CTSL's involvement in hair follicle cycling and skin homeostasis

  • Changes in bone remodeling and development

  • Modified responses to metabolic challenges

When working with CTSL knockout mice, researchers should account for these baseline phenotypic alterations when designing studies, especially those focused on infection models or metabolic disorders. The multisystem effects of CTSL deficiency highlight the enzyme's diverse physiological roles and provide valuable research opportunities for understanding disease mechanisms .

How should researchers activate recombinant mouse Cathepsin L for in vitro assays?

For optimal activation of recombinant mouse Cathepsin L (rmCTSL) in experimental settings, follow this methodological approach:

  • Prepare Activation Buffer containing 50 mM Sodium Citrate, 150 mM NaCl, 1 mM EDTA, and 0.615% CHAPS, adjusted to pH 3.0

  • Dilute rmCTSL to a concentration of 100 μg/mL in the Activation Buffer

  • Incubate the solution at room temperature for exactly 60 minutes to achieve complete activation

  • Following activation, dilute the activated rmCTSL to the required working concentration (typically 0.05 ng/μL) in Assay Buffer (25 mM MES with 5 mM DTT, pH 6.0)

This activation protocol is critical because CTSL is synthesized as an inactive proenzyme that requires proteolytic processing to generate the enzymatically active form. The acidic conditions in the Activation Buffer mimic the lysosomal environment where CTSL naturally becomes activated. Deviation from these specific conditions may result in suboptimal enzyme activity, affecting experimental outcomes and data reliability .

What is the optimal protocol for measuring CTSL activity in mouse tissue samples?

The optimal protocol for measuring CTSL activity in mouse tissue samples involves the following standardized procedure:

  • Tissue preparation:

    • Collect fresh tissue samples and homogenize in ice-cold extraction buffer (25 mM MES, pH 6.0, containing 5 mM DTT and protease inhibitor cocktail excluding cysteine protease inhibitors)

    • Centrifuge homogenates at 10,000g for 10 minutes at 4°C

    • Collect supernatant and determine protein concentration

  • Activity assay setup:

    • Prepare tissue lysates (typically 10-50 μg total protein) in Assay Buffer (25 mM MES, 5 mM DTT, pH 6.0)

    • Use fluorogenic substrate Z-Leu-Arg-AMC (20 μM final concentration)

    • Include appropriate controls: substrate blank and positive control (activated recombinant CTSL)

  • Measurement procedure:

    • Load 50 μL of prepared sample into each well of a black 96-well plate

    • Add 50 μL of substrate solution (20 μM)

    • Monitor fluorescence at excitation/emission wavelengths of 380/460 nm in kinetic mode for 5 minutes

    • Calculate specific activity using the formula:

      Specific Activity (pmol/min/μg) = [Adjusted Vmax (RFU/min) × Conversion Factor (pmol/RFU)] ÷ amount of enzyme (μg)

This method provides high sensitivity and specificity for CTSL, minimizing interference from other proteases. For comparative studies examining CTSL activity across different experimental conditions (such as SARS-CoV-2 infection), consistent application of this protocol ensures reliable and reproducible results .

What are the key considerations when designing experiments using human ACE2 transgenic mice for CTSL studies?

When designing experiments using human ACE2 (hACE2) transgenic mice for CTSL studies, researchers should address several critical considerations:

  • Age and sex selection:

    • Use mice of consistent age (4-5 weeks optimal for infection studies)

    • Account for sex-based differences in CTSL expression and activity

    • Standardize body weight range (13-17g recommended for young adult mice)

  • Genetic background validation:

    • Confirm hACE2 expression levels before experiments

    • Validate the CRISPR/Cas9 knock-in technology used to generate the model

    • Consider potential off-target effects of genetic modification

  • Experimental design specifics:

    • Include proper wild-type controls alongside transgenic animals

    • Design appropriate infection protocols (pseudovirus systems are recommended for BSL-2 laboratories)

    • Incorporate pharmacological interventions (CTSL inhibitors) with appropriate vehicle controls

    • Plan tissue collection timepoints based on infection kinetics

  • Assessment methods:

    • Utilize bioluminescence imaging for tracking infection progression in vivo

    • Measure VSV-P mRNA levels as an indicator of pseudovirus infection

    • Quantify both CTSL protein levels and enzymatic activity in relevant tissues

    • Compare CTSL expressions between different organs (e.g., liver vs. lung)

These considerations ensure that experiments accurately assess CTSL's role in SARS-CoV-2 infection while controlling for variables that might influence results. The hACE2 transgenic mouse model provides a valuable system for studying CTSL in a physiologically relevant context, but requires careful experimental design to yield meaningful data .

How does hyperglycemia affect CTSL maturation and function in diabetic mouse models?

Hyperglycemia significantly impacts CTSL maturation and function in diabetic mouse models through multiple interconnected mechanisms:

  • Enhanced CTSL expression and processing:

    • Chronic hyperglycemia upregulates CTSL gene expression in multiple tissues

    • Glucose directly promotes post-translational maturation of pro-CTSL to active CTSL

    • In Lepr db/db diabetic mice, elevated blood glucose correlates positively with increased CTSL concentration and enzymatic activity

  • Subcellular localization changes:

    • Hyperglycemia induces migration of CTSL from lysosomes to alternative cellular compartments

    • This relocalization enhances CTSL's accessibility to substrates involved in pathological processes

    • Altered CTSL trafficking contributes to increased SARS-CoV-2 spike protein processing efficiency

  • Functional consequences:

    • Enhanced CTSL activity in high-glucose environments facilitates SARS-CoV-2 infection

    • CTSL knockout substantially reduces this glucose-mediated enhancement of viral infection

    • Glucose-induced CTSL activation creates a "vicious circle" where viral infection further increases CTSL activity

The relationship between hyperglycemia and CTSL represents a critical mechanism underlying increased COVID-19 severity in diabetic patients. These findings suggest that glucose control may serve as an intervention point to reduce CTSL-mediated viral susceptibility, highlighting the translational significance of mouse model studies in understanding human disease mechanisms .

What is the comparative efficacy of different CTSL inhibitors in mouse models of viral infection?

The comparative efficacy of CTSL inhibitors in mouse models of viral infection reveals significant differences in potency, specificity, and therapeutic potential:

InhibitorMechanismIn Vivo EfficacySpecificityNotable Characteristics
E64dBroad cysteine protease inhibitorSignificant prevention of SARS-CoV-2 pseudovirus infection (p<0.01)Inhibits multiple cathepsinsEstablished research tool with well-characterized pharmacodynamics
AmantadineLicensed anti-influenza drugModerate prevention of infection (p=0.058, trending toward significance)Acts on multiple targets including CTSLClinically approved drug with established safety profile
SID 26681509Selective CTSL inhibitorNot tested in the provided studiesHigh selectivity for CTSL over other cathepsinsUseful for distinguishing CTSL-specific effects

Both E64d and amantadine significantly decreased the hepatic VSV-P mRNA levels in human ACE2 transgenic mice following SARS-CoV-2 pseudovirus infection, confirming their antiviral efficacy. Additionally, both compounds reversed the virus-induced elevation of CTSL protein levels in the liver, while not significantly affecting cathepsin B levels .

The differential effects observed with these inhibitors highlight important considerations for therapeutic development. While broad-spectrum inhibitors like E64d show higher efficacy, selective CTSL inhibitors may offer advantages in reducing off-target effects. Amantadine's dual mechanism as both an anti-influenza drug and CTSL inhibitor suggests potential for drug repurposing, particularly relevant for treating COVID-19 in diabetic patients with elevated CTSL levels .

How do CTSL activity patterns differ between various organs in infected mouse models?

CTSL activity patterns exhibit significant organ-specific variations in infected mouse models, reflecting tissue-specific responses to SARS-CoV-2 infection:

  • Liver responses:

    • Shows pronounced CTSL elevation following SARS-CoV-2 pseudovirus infection

    • Significant increase in CTSL protein levels (p<0.01) compared to uninfected controls

    • High VSV-P mRNA levels indicating substantial infection

    • Marked responsiveness to CTSL inhibitors (E64d and amantadine)

  • Lung responses:

    • Despite being the primary target organ for SARS-CoV-2, shows only slight pseudovirus infection in mouse models

    • CTSL elevation trend present but not statistically significant

    • Lower baseline CTSL activity compared to liver tissue

    • Different kinetics of CTSL activation following infection

  • Organ-specific mechanisms:

    • Differential expression of complementary factors (e.g., ACE2, TMPRSS2) influences organ-specific CTSL contribution to infection

    • Metabolic status of the tissue affects glucose-mediated CTSL activation

    • Tissue-specific inflammatory responses may modulate CTSL activity independently of direct viral effects

These organ-specific differences highlight the importance of multi-tissue analysis when evaluating CTSL-targeted interventions. The pronounced liver response suggests this organ may serve as a sensitive indicator of systemic CTSL changes during infection, while the variable lung response indicates potential limitations of mouse models for studying respiratory aspects of SARS-CoV-2 pathogenesis .

How should researchers account for baseline variations in CTSL expression when analyzing experimental data?

Accounting for baseline variations in CTSL expression is critical for accurate data interpretation. Researchers should implement the following standardized approaches:

  • Comprehensive baseline characterization:

    • Establish normal CTSL expression ranges for each experimental mouse strain

    • Document age-dependent and sex-specific variations in CTSL levels

    • Analyze tissue-specific expression patterns with appropriate statistical methods

    • Consider circadian variations in CTSL expression when scheduling experiments

  • Normalization strategies:

    • Always include appropriate housekeeping genes/proteins for normalization (β-actin, GAPDH)

    • Use percentage change or fold-change relative to matched controls rather than absolute values

    • Employ matched-pair analyses when possible to minimize inter-individual variation

    • Consider ratio-metric approaches (CTSL:CTSB ratio) for more stable comparisons

  • Statistical approaches:

    • Utilize non-parametric tests (Mann-Whitney U test) for data not normally distributed

    • Apply multivariate regression models to account for confounding variables

    • Use ANCOVA when baseline values might influence treatment effects

    • Report exact p-values and confidence intervals rather than significance thresholds

  • Experimental design considerations:

    • Include adequate biological replicates (n ≥ 6 per group recommended)

    • Implement randomization and blinding procedures

    • Pre-register analysis plans to avoid post-hoc adjustments

    • Consider power calculations based on expected effect sizes and known variability

By systematically addressing baseline variations, researchers can distinguish true experimental effects from background fluctuations, increasing the reliability and reproducibility of CTSL studies in mouse models .

What are the key methodological challenges in comparing CTSL knockout studies with pharmacological inhibition experiments?

Comparing CTSL knockout studies with pharmacological inhibition experiments presents several methodological challenges requiring careful consideration:

  • Temporal differences in CTSL suppression:

    • Knockout models represent developmental absence of CTSL, allowing compensatory mechanisms to develop

    • Pharmacological inhibition causes acute suppression of pre-existing CTSL activity

    • This temporal disparity may lead to fundamentally different phenotypes despite targeting the same protein

  • Specificity considerations:

    • Genetic knockouts offer high specificity for CTSL elimination

    • Pharmacological approaches often affect multiple cathepsins or other proteases

    • E64d inhibits multiple cysteine proteases while amantadine has additional mechanisms beyond CTSL inhibition

    • These specificity differences complicate direct comparisons between approaches

  • Quantitative aspects of inhibition:

    • Knockouts typically achieve complete CTSL elimination

    • Pharmacological inhibitors rarely achieve 100% inhibition

    • Dose-dependent effects of inhibitors create additional variables absent in knockout models

    • Residual CTSL activity may be sufficient for some biological functions

  • Experimental design solutions:

    • Include both approaches in parallel experiments when possible

    • Implement inducible knockout systems to better mimic pharmacological timing

    • Utilize multiple inhibitors with different mechanisms and specificities

    • Measure actual CTSL activity reduction rather than assuming complete inhibition

    • Consider combinatorial approaches (partial knockdown plus inhibitor) to achieve comparable inhibition levels

Understanding these methodological challenges allows researchers to design more robust experiments and appropriately contextualize seemingly contradictory results between genetic and pharmacological approaches to CTSL inhibition .

How can researchers differentiate between direct and indirect effects of CTSL modulation in complex disease models?

Differentiating between direct and indirect effects of CTSL modulation in complex disease models requires sophisticated experimental approaches:

  • Temporal intervention mapping:

    • Implement time-course experiments to establish sequences of events following CTSL modulation

    • Use inducible systems (Tet-On/Off) for temporally controlled CTSL expression

    • Apply CTSL inhibitors at different disease stages to identify critical windows

    • Examine immediate versus delayed consequences of CTSL manipulation

  • Cell type-specific approaches:

    • Utilize conditional knockout models targeting CTSL in specific cell populations

    • Employ cell type-specific promoters to drive CTSL expression

    • Analyze effects in isolated primary cells versus intact tissues

    • Compare tissue-resident versus infiltrating cell responses to CTSL modulation

  • Molecular pathway dissection:

    • Perform parallel inhibition of suspected downstream mediators

    • Use phosphoproteomic analysis to identify activation of signaling cascades

    • Apply computational network analysis to distinguish primary from secondary effects

    • Implement rescue experiments reintroducing specific CTSL substrates

  • Mechanistic validation strategies:

    • Design point mutants affecting specific CTSL functions (e.g., catalytically inactive mutants)

    • Use substrate-specific activity assays rather than general protease activity

    • Implement domain-specific blocking antibodies

    • Perform cross-species validation to identify conserved versus species-specific effects

By systematically employing these approaches, researchers can construct mechanistic models distinguishing causal CTSL effects from downstream consequences. This differentiation is particularly important in complex models like diabetic mice infected with SARS-CoV-2, where multiple interacting pathways may be simultaneously affected by CTSL modulation .

How do findings from mouse CTSL studies translate to human clinical applications?

Translating findings from mouse CTSL studies to human clinical applications requires careful consideration of similarities and differences between species:

  • Comparative biology aspects:

    • Mouse and human CTSL share approximately 75% amino acid sequence homology

    • Substrate specificity is largely conserved, particularly for viral spike protein processing

    • Mouse models demonstrate similar glucose-mediated CTSL activation patterns to human samples

    • CTSL's role in SARS-CoV-2 infection appears mechanistically consistent between species

  • Validated translational findings:

    • Elevation of circulating CTSL in diabetic patients with COVID-19 confirms mouse model observations

    • Correlation between CTSL levels and disease severity is consistent across species

    • Hyperglycemia-induced CTSL maturation observed in both mouse and human cells

    • Pharmacological CTSL inhibition shows similar effects in mouse models and human cell lines

  • Key translational limitations:

    • Mice require human ACE2 expression to model SARS-CoV-2 infection

    • Inflammatory responses to infection differ between species

    • Metabolic regulation of CTSL may have species-specific aspects

    • Mouse models typically represent homogeneous genetic backgrounds unlike human populations

  • Clinical application pathways:

    • CTSL inhibitors identified in mouse studies represent potential therapeutic candidates

    • Blood CTSL levels may serve as biomarkers for COVID-19 severity risk, particularly in diabetic patients

    • Glucose control strategies validated in mouse models have direct clinical relevance

    • Combined approaches (glucose control plus CTSL inhibition) supported by mouse data warrant clinical investigation

The demonstrated predictive value of mouse CTSL studies for human COVID-19 pathogenesis supports their translational relevance, while acknowledging species-specific limitations that must be addressed when moving toward clinical applications .

What are the optimal experimental controls for CTSL inhibitor studies in mouse models?

For robust CTSL inhibitor studies in mouse models, researchers should implement comprehensive control strategies:

  • Vehicle controls:

    • Include matched vehicle solutions containing all components except the active inhibitor

    • Administer using identical routes, volumes, and schedules as the test compounds

    • Account for potential solvent effects (DMSO, ethanol) on baseline CTSL activity

    • Monitor for vehicle-induced physiological changes independent of CTSL inhibition

  • Inhibitor specificity controls:

    • Include structurally similar compounds lacking CTSL inhibitory activity

    • Test effects on related cathepsins (e.g., cathepsin B) to assess specificity

    • Perform parallel experiments with CTSL knockout mice to identify inhibitor off-target effects

    • Include dose-response studies to establish inhibition thresholds

  • Biological validation controls:

    • Measure actual CTSL activity reduction in target tissues following inhibitor administration

    • Assess pharmacokinetic/pharmacodynamic relationships for each inhibitor

    • Monitor inhibitor stability and tissue distribution

    • Include positive control inhibitors with well-established effects (E64d)

  • Experimental design controls:

    • Implement randomization protocols for treatment assignment

    • Conduct blinded assessment of outcomes

    • Include both wild-type and disease model mice (e.g., diabetic models)

    • Test multiple timepoints to capture both immediate and delayed inhibitor effects

  • Statistical approach:

    • Predetermine sample sizes based on power calculations

    • Establish clear inclusion/exclusion criteria

    • Plan for multiple comparison corrections

    • Consider hierarchical statistical models for complex experimental designs

These control strategies minimize experimental artifacts and increase confidence in attributing observed effects specifically to CTSL inhibition, crucial for identifying clinically relevant inhibitors for translational development .

How can researchers integrate mouse CTSL findings with human patient data for drug development?

Integrating mouse CTSL findings with human patient data requires systematic approaches to bridge preclinical and clinical research domains:

  • Biomarker correlation strategies:

    • Measure identical CTSL-related parameters in mouse models and patient samples

    • Correlate mouse tissue CTSL levels with human circulating CTSL concentrations

    • Develop standardized CTSL activity assays applicable to both mouse tissues and human biospecimens

    • Validate predictive biomarkers identified in mouse models using patient cohorts

  • Mechanistic validation approaches:

    • Confirm key molecular pathways (glucose-mediated CTSL activation) in patient-derived samples

    • Use ex vivo human tissue cultures to replicate mouse model findings

    • Implement humanized mouse models expressing human CTSL variants

    • Apply systems biology approaches to map conserved and divergent CTSL networks

  • Translational experimental design:

    • Conduct parallel intervention studies in mouse models and human cell systems

    • Design mouse experiments to mimic clinical scenarios (comorbidities, varied timing of intervention)

    • Include patient-relevant endpoints beyond basic molecular measures

    • Consider population heterogeneity factors absent in inbred mouse strains

  • Drug development integration framework:

    • Use mouse pharmacokinetic/pharmacodynamic data to inform human dosing strategies

    • Prioritize inhibitors based on combined mouse efficacy and human safety profiles

    • Develop companion diagnostics for CTSL activity alongside potential therapeutics

    • Apply allometric scaling principles for translating dosages between species

  • Clinical trial preparation:

    • Identify patient subpopulations most likely to benefit based on mouse/human correlations

    • Design stratification strategies based on CTSL activity or related parameters

    • Establish clear go/no-go decision points based on integrated mouse/human datasets

    • Prepare surrogate endpoint validation from mouse to human studies

This integrated approach maximizes the translational value of mouse CTSL research, increasing the probability of successful clinical application in conditions like COVID-19, particularly for high-risk populations such as diabetic patients .

Product Science Overview

Structure and Function

Cathepsin-L is a member of the peptidase C1 family and is composed of disulfide-linked heavy and light chains, both derived from a single protein precursor . The enzyme is initially synthesized as an inactive proenzyme (procathepsin L) and is activated through proteolytic cleavage. The active form of Cathepsin-L has a molecular mass of approximately 37 kDa .

The enzyme’s primary function is to degrade proteins within the lysosome, a cellular organelle responsible for breaking down waste materials and cellular debris. Cathepsin-L is particularly potent in degrading structural proteins of basement membranes, such as collagen and laminin . It also plays a role in the activation of other proteases, such as the proform of urokinase-type plasminogen activator .

Recombinant Mouse Cathepsin-L

Recombinant Mouse Cathepsin-L is produced using a mouse myeloma cell line (NS0) and is often tagged with a C-terminal 10-His tag for purification purposes . The recombinant form is used in various research applications, including studies on protein degradation, enzyme kinetics, and the role of Cathepsin-L in pathological processes.

The recombinant protein is typically supplied as a carrier-free solution in Tris and NaCl and is stable for up to six months when stored at -20 to -70°C . It is important to avoid repeated freeze-thaw cycles to maintain the protein’s activity.

Applications and Implications

Cathepsin-L has been implicated in several pathological processes, including myofibril necrosis in myopathies, myocardial ischemia, and the renal tubular response to proteinuria . Its ability to degrade extracellular matrix components makes it a key player in tissue remodeling and repair.

In research, recombinant Mouse Cathepsin-L is used to study its role in various diseases and to develop potential therapeutic interventions. For example, inhibitors of Cathepsin-L are being explored as potential treatments for conditions such as cancer, osteoporosis, and cardiovascular diseases.

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 2024 Thebiotek. All Rights Reserved.