CTSB Mouse

Cathepsin-B Mouse Recombinant
Shipped with Ice Packs
In Stock

Description

Behavioral and Cognitive Outcomes

InterventionModel (AD Type)Key FindingsReferences
AAV-mediated CTSB overexpression (muscle)APP/PS1 (familial AD)- Reversed motor coordination deficits (rotarod)
- Improved spatial memory retention (Morris water maze)
- Restored hippocampal neurogenesis
Ctsb knockout (KO)hAβPP-695/Wtβ-Lonγ (sporadic AD)- 85% reduction in Aβ<sub>1–40/42</sub> and amyloid plaques
- 60% decrease in CTFβ (β-secretase marker)
- Improved memory retention
Ctsb KOhAβPP-695/Sweβ-Lonγ (familial AD)No effect on Aβ or memory deficits

Substrate Specificity and Aβ Modulation

  • β-Secretase Activity: CTSB cleaves the wild-type (WT) β-secretase site (Z-Val-Lys-Met-AMC) with 2,735-fold higher efficiency than the Swedish mutant (Z-Val-Asn-Leu-AMC) .

  • Aβ Pathology:

    • Ctsb KO reduced pGlu-Aβ<sub>3–40/42</sub> by 65–90% and amyloid plaques by 85% in sporadic AD models .

    • Overexpression increased pGlu-Aβ and plaque load by 150–200% .

Proteomic and Cellular Mechanisms

Hippocampal and Plasma Proteome Remodeling

  • AD-Ctsb Mice: Normalized dysregulated proteins in APP/PS1 mice to WT levels, including synaptic markers (e.g., synaptophysin) and inflammation mediators (e.g., GFAP) .

  • Muscle-CTSB Interaction: Altered circulating factors (e.g., IGF-1, BDNF) linked to improved neurogenesis and reduced gliosis .

Autophagy and Apoptosis

  • Ctsb KO in pancreatic acinar cells impaired autophagy, leading to chronic pancreatitis via p62/LC3 accumulation .

  • In granulosa cells, Ctsb silencing reduced caspase-3/8 and TNF-α, promoting Bcl-2-mediated survival .

Therapeutic Implications for Human AD

Disease-Modifying Effects

  • Sporadic AD: CTSB inhibitors (e.g., CA-074Me) reduce Aβ<sub>40/42</sub> in neuroinflammatory models .

  • Periodontitis-Associated AD: Ctsb KO blocks PgLPS-induced Aβ<sub>42</sub> elevation, linking oral pathogens to neurodegeneration .

Challenges

  • Dual Role: CTSB overexpression benefits AD mice but impairs spatial memory in WT mice .

  • Mutation Dependency: CTSB modulates Aβ only in models with WT β-secretase sites, not Swedish mutants .

Product Specs

Introduction
Cathepsin B preproprotein (CTSB) is a cysteine protease belonging to the papain family. It is typically found within lysosomes. CTSB possesses the ability to break down various components of the extracellular matrix under both neutral and acidic pH conditions. This protease has been linked to the advancement of certain tumors in humans and rodents, as well as to the progression of arthritis.
Description
Recombinant CTSB Mouse, produced in Sf9 Baculovirus cells, is a single, glycosylated polypeptide chain comprising 330 amino acids (18-339a.a.) with a molecular weight of 36.4kDa. On SDS-PAGE, the apparent molecular size will be approximately 28-40kDa. The protein is expressed with an 8 amino acid His tag located at the C-terminus and purified using proprietary chromatographic techniques.
Physical Appearance
Clear, sterile solution after filtration.
Formulation
CTSB protein solution at a concentration of 0.5mg/ml, prepared in Phosphate Buffered Saline (pH 7.4) and supplemented with 10% glycerol.
Stability
For short-term storage (2-4 weeks), the product can be stored at 4°C. For extended storage, freeze the product at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Repeated freeze-thaw cycles should be avoided.
Purity
Purity exceeds 90% as assessed by SDS-PAGE.
Synonyms
Cathepsin B, Cathepsin B1, CTSB.
Source
Sf9, Baculovirus cells.
Amino Acid Sequence
HDKPSFHPLS DDLINYINKQ NTTWQAGRNF YNVDISYLKK LCGTVLGGPK LPGRVAFGED IDLPETFDAR EQWSNCPTIG QIRDQGSCGS CWAFGAVEAI SDRTCIHTNG RVNVEVSAED LLTCCGIQCG DGCNGGYPSG AWSFWTKKGL VSGGVYNSHV GCLPYTIPPC EHHVNGSRPP CTGEGDTPRC NKSCEAGYSP SYKEDKHFGY TSYSVSNSVK EIMAEIYKNG PVEGAFTVFS DFLTYKSGVY KHEAGDMMGG HAIRILGWGV ENGVPYWLAA NSWNLDWGDN GFFKILRGEN HCGIESEIVA GIPRTDQYWG RFLEHHHHHH.

Q&A

What is CTSB and what is its functional significance in mouse models?

Cathepsin B (CTSB) is a lysosomal cysteine protease that has been extensively studied in mouse models for its roles in both physiological and pathological processes. In normal physiology, CTSB contributes to protein turnover within lysosomes. In pathological contexts, CTSB has been linked to cancer progression, particularly in promoting tumor invasion and metastasis, as well as having complex roles in neurodegenerative diseases .

Functionally, CTSB can promote extracellular matrix degradation, which facilitates tumor cell invasion. In breast cancer models such as the PyMT mouse model, CTSB overexpression accelerates tumor growth and increases metastatic burden in lungs . Conversely, in neurological contexts, CTSB has been identified as a myokine (muscle-secreted factor) that can enhance memory function and adult hippocampal neurogenesis in certain conditions .

What are the primary mouse models used to study CTSB function?

Several mouse models have been developed to investigate CTSB function across different contexts:

  • Genetic knockout models: CTSB-deficient mice have been generated to study loss-of-function effects.

  • Transgenic overexpression models: Models with human CTSB overexpression, such as the PyMT+/0;CTSB+/0 model for breast cancer research .

  • Inducible expression systems: DOX-inducible CTSB expression systems have been employed to achieve temporal control over CTSB expression in specific cell types .

  • Tissue-specific expression models: AAV-vector-mediated CTSB overexpression in specific tissues, such as skeletal muscle for neurodegeneration studies in APP/PS1 Alzheimer's Disease mouse models .

  • Orthotopic transplantation models: Primary PyMT breast cancer cells with or without CTSB overexpression transplanted into mammary glands of recipient mice with different CTSB genotypes .

These models provide complementary approaches to investigate both systemic and tissue-specific roles of CTSB in different disease contexts.

How is CTSB expression and activity quantified in mouse tissue samples?

Researchers employ multiple complementary techniques to quantify CTSB expression and activity:

  • Transcriptional analysis:

    • RT-qPCR for CTSB mRNA quantification

    • Microarray or RNA-seq for expression analysis in complex samples

  • Protein expression analysis:

    • Western blotting to detect pro-CTSB (inactive precursor) and mature CTSB forms

    • Immunohistochemistry/immunofluorescence to visualize spatial distribution of CTSB in tissue sections

  • Activity assays:

    • Fluorogenic substrate-based assays using CTSB-specific substrates

    • Activity-based probes that selectively label active CTSB

    • In situ zymography to visualize proteolytic activity in tissue sections

  • Separation of cellular compartments:

    • Fractionation of lysosomes and extracellular media to distinguish between intracellular and secreted CTSB

When interpreting results, it's crucial to consider that CTSB exists in multiple forms (pro-enzyme, mature single-chain, mature double-chain) and that activity may not directly correlate with protein expression levels due to post-translational regulation mechanisms .

How does CTSB overexpression affect tumor progression in PyMT mouse models of breast cancer?

CTSB overexpression in PyMT mouse models has distinct effects on tumor progression that have been experimentally validated:

  • Enhanced tumor growth: Ubiquitous overexpression of human CTSB in the PyMT model results in accelerated tumor growth, as demonstrated by increased tumor volumes over time when comparing PyMT+/0;CTSB+/0 mice to PyMT+/0;wt mice .

  • Increased lung metastasis: CTSB overexpression leads to higher metastatic burden in lungs of PyMT mice .

  • Enhanced invasiveness: CTSB overexpression increases collective cell invasion and extracellular matrix proteolysis in 3D culture models. This is likely mediated by increased proteolytic degradation of extracellular matrix proteins, facilitating invasion into adjacent tissue .

  • No effect on EMT: Interestingly, CTSB overexpression does not significantly alter epithelial-to-mesenchymal transition (EMT) markers or the number of cells undergoing EMT, suggesting it promotes invasion through matrix degradation rather than by inducing EMT .

  • Minimal effect on cell migration: CTSB overexpression has minimal impact on cell migration in Boyden chamber assays without matrix barriers, but enhances invasion through collagen I matrices, further supporting its role in proteolytic matrix degradation rather than intrinsic migratory capacity .

These findings collectively indicate that CTSB promotes tumor progression primarily by enhancing the invasive capacity of cancer cells through increased proteolytic activity.

How can researchers differentiate between cancer cell-derived and stromal cell-derived CTSB effects?

Distinguishing between cancer cell-derived and stromal CTSB effects is methodologically challenging but can be achieved through various experimental approaches:

  • Orthotopic transplantation with genetic manipulation:

    • Transplanting PyMT+/0;CTSB+/0 or PyMT+/0;wt cancer cells into mammary glands of either CTSB+/0 or wt recipient mice creates four experimental conditions that allow separation of cancer cell vs. stromal CTSB effects .

    • This approach revealed that cancer cell CTSB expression, rather than stromal CTSB, drives tumor growth in the PyMT model .

  • Cell type-specific inducible expression systems:

    • DOX-inducible CTSB expression systems can be used to selectively express CTSB in cancer cells or in specific stromal cells like macrophages .

    • This approach demonstrated that CTSB expression in tumor cells, but not in macrophages, enhanced invasive sprouting in 3D coculture models .

  • 3D coculture systems:

    • Coculturing tumor cells with bone marrow-derived macrophages of defined CTSB genotypes allows investigation of cellular crosstalk .

    • These systems revealed that while macrophages enhance tumor spheroid invasiveness, this effect was not further promoted by CTSB overexpression in macrophages .

  • Cell-selective CTSB inhibition:

    • Using cell-type specific genetic ablation of CTSB through conditional knockout approaches.

    • Employing cell-selective delivery of CTSB inhibitors.

These approaches have collectively demonstrated that in the PyMT breast cancer model, the tumor-promoting effects of CTSB are primarily cancer cell-autonomous rather than mediated by stromal cells .

What molecular mechanisms underlie CTSB-mediated invasion in breast cancer mouse models?

CTSB promotes invasion through several interconnected molecular mechanisms:

These mechanisms emphasize CTSB's primary role as a direct mediator of matrix degradation supporting invasive processes, rather than as a regulator of broader phenotypic changes in cancer cells.

How does muscle-specific CTSB overexpression affect neurodegenerative disease progression in mouse models?

Skeletal muscle-specific CTSB overexpression has shown remarkable neuroprotective effects in mouse models of Alzheimer's Disease (AD):

  • Cognitive improvement:

    • AAV-vector-mediated CTSB overexpression in skeletal muscle of APP/PS1 mice (AD model) significantly improves memory retention in Morris water maze and fear conditioning tasks .

    • AD mice treated with muscle CTSB spent more time in the target quadrant during probe trials, comparable to wildtype control mice .

  • Motor function enhancement:

    • CTSB treatment prevents motor coordination deficits typically observed in AD mice .

    • This suggests broad neurological benefits beyond memory function .

  • Enhanced adult hippocampal neurogenesis:

    • Muscle CTSB expression increases the generation of new neurons in the adult hippocampus of AD mice .

    • This neurogenic effect may contribute to the observed cognitive improvements .

  • Tissue-specific proteome remodeling:

    • CTSB treatment modifies hippocampal, muscle, and plasma proteomic profiles in AD mice to resemble those of wildtype controls .

    • In the hippocampus, CTSB treatment increases abundance of proteins involved in mRNA metabolism and protein synthesis, including those relevant to adult neurogenesis and memory function .

    • In plasma, CTSB enhances metabolic and mitochondrial processes while reducing inflammatory responses .

    • In muscle, CTSB elevates protein translation in AD mice .

  • No effect on amyloid pathology:

    • Interestingly, CTSB treatment does not affect amyloid plaque pathology or neuroinflammation directly .

    • This suggests CTSB acts through mechanisms independent of classical AD pathology .

These findings indicate that muscle-derived CTSB may serve as a potential therapeutic approach for AD, working through mechanisms that improve neuroplasticity rather than directly targeting disease pathology.

What are the contrasting effects of CTSB in wildtype versus AD mouse models?

CTSB overexpression produces strikingly opposite effects in wildtype compared to AD mouse models, revealing an important context-dependent function:

  • Cognitive function:

    • In AD mice: CTSB improves spatial memory retention in Morris water maze and fear conditioning .

    • In wildtype mice: CTSB impairs spatial memory retention, with mice performing at chance levels in probe trials .

  • Fear conditioning:

    • In AD mice: CTSB treatment normalizes fear learning and memory to levels similar to wildtype controls .

    • In wildtype mice: CTSB reduces conditioned fear responses during both acquisition and testing phases .

  • Proteomic changes:

    • In AD mice: CTSB shifts proteome profiles toward those of healthy controls .

    • In wildtype mice: CTSB alters protein profiles across tissues to resemble those of untreated AD mice .

  • Mitochondrial proteins:

    • In AD mice: CTSB enhances mitochondrial processes in plasma .

    • In wildtype mice: CTSB decreases mitochondrial proteins in muscle tissue .

  • Protein translation:

    • In AD mice: CTSB elevates protein translation in muscle .

    • In wildtype mice: Different effects on translational machinery are observed .

This bidirectional effect emphasizes the importance of disease context in determining CTSB outcomes. The beneficial effects in AD mice versus detrimental effects in wildtype mice suggest CTSB may function as a compensatory mechanism that is only beneficial in the context of neurodegeneration, potentially acting to restore disrupted homeostasis rather than enhancing normal function .

What methodological approaches are optimal for studying muscle-brain crosstalk involving CTSB?

Investigating muscle-brain crosstalk mediated by CTSB requires sophisticated methodological approaches:

  • Tissue-specific genetic manipulation:

    • AAV-vector-mediated expression systems for targeted CTSB overexpression in skeletal muscle .

    • Muscle-specific promoters (e.g., MCK promoter) to drive transgene expression exclusively in muscle tissue.

    • Inducible systems (e.g., tetracycline-controlled) to regulate timing of expression.

  • Multi-tissue analysis:

    • Parallel assessment of muscle, plasma, and brain tissue to track CTSB expression, secretion, and downstream effects .

    • Proteomic profiling across tissues to identify molecular changes associated with muscle-brain communication .

  • Behavioral testing battery:

    • Comprehensive behavioral assessment including tests for motor function (rotarod), spatial memory (Morris water maze), and associative learning (fear conditioning) .

    • Multiple probe trials with varying retention intervals to assess short and long-term memory effects.

  • Adult neurogenesis quantification:

    • BrdU labeling combined with neuronal markers to quantify newborn neurons in the hippocampus.

    • Doublecortin staining to assess levels of immature neurons.

    • Retroviral labeling for morphological analysis of newly generated neurons.

  • Secretome analysis:

    • Cultured muscle tissue to analyze the composition of secreted factors.

    • Mass spectrometry-based approaches to identify and quantify secreted proteins.

    • Tracking labeled CTSB to determine tissue distribution after release from muscle.

  • In vivo microdialysis:

    • To measure changes in extracellular CTSB and associated factors in the brain following muscle manipulation.

  • Blood-brain barrier studies:

    • Assessing whether muscle-derived CTSB crosses the blood-brain barrier or acts through intermediate factors.

    • Two-photon microscopy with fluorescently labeled CTSB to track potential BBB crossing.

These methodological approaches collectively enable comprehensive investigation of how muscle-derived CTSB communicates with the brain and influences neurological outcomes in both healthy and disease contexts.

How can researchers design experiments to resolve contradictory findings regarding CTSB's role in different disease models?

Resolving contradictory findings about CTSB requires carefully designed experiments:

  • Context-dependent experimental design:

    • Direct comparison of CTSB effects in multiple disease models under identical experimental conditions.

    • Use of matched genetic backgrounds and standardized methodologies across studies.

    • Systematic evaluation of CTSB in both gain-of-function and loss-of-function approaches within the same disease model .

  • Temporal considerations:

    • Time-course experiments using inducible systems to determine whether CTSB effects vary with disease stage.

    • DOX-inducible CTSB expression systems can provide temporal control over when CTSB is expressed, allowing assessment of stage-specific effects .

  • Cell/tissue type specificity:

    • Utilize tissue-specific CTSB manipulation to distinguish between effects in different compartments.

    • Compare cancer cell-specific versus stromal cell-specific CTSB overexpression in the same tumor model .

    • Contrast muscle-specific versus neuronal CTSB expression in neurodegeneration models .

  • Dosage considerations:

    • Titration experiments with varying levels of CTSB expression to determine whether effects are dose-dependent.

    • Assessment of physiological versus supra-physiological CTSB levels.

  • Compensatory mechanism evaluation:

    • Analysis of other cathepsin family members to identify potential compensatory changes.

    • Broad proteomic profiling to detect system-wide adaptations to altered CTSB levels .

  • Genetic and environmental interaction:

    • Factorial experimental designs combining CTSB manipulation with other genetic factors.

    • Testing CTSB effects across different environmental conditions (e.g., exercise, diet).

By implementing these experimental design principles, researchers can better contextualize seemingly contradictory findings and develop a more nuanced understanding of CTSB's multifaceted roles across different disease contexts.

What challenges arise in interpreting proteomics data from CTSB-modified mouse tissues?

Proteomics analysis of CTSB-modified tissues presents several interpretive challenges:

  • Direct versus indirect effects:

    • Distinguishing protein changes directly caused by CTSB proteolytic activity versus secondary signaling cascade effects.

    • Identifying which proteome alterations are responsible for phenotypic changes versus being consequences of those changes .

  • Tissue heterogeneity:

    • Brain tissue contains multiple cell types (neurons, glia, vascular cells), making it difficult to assign proteome changes to specific cellular populations.

    • Single-cell or cell type-specific proteomics approaches may be necessary for proper interpretation.

  • Temporal dynamics:

    • Proteome changes may be transient or evolve over time, requiring time-course analyses.

    • Early versus late effects of CTSB modification may reflect different biological processes.

  • Post-translational modifications:

    • Standard proteomics may miss critical post-translational modifications affected by CTSB.

    • Specialized approaches like phosphoproteomics or glycoproteomics may be required for complete understanding.

  • Proteoform complexity:

    • CTSB itself exists in multiple forms (pro-enzyme, mature single-chain, mature double-chain) that may not be distinguished in standard proteomics .

    • Other proteins may similarly exist in multiple processed forms with distinct functions.

  • Context-dependent interpretation:

    • The same proteome changes may have opposite functional implications in different disease contexts.

    • As observed with CTSB treatment, changes beneficial in AD mice may be detrimental in wildtype mice .

  • Integration across tissues:

    • Correlating proteome changes in muscle, plasma, and brain requires sophisticated bioinformatic approaches.

    • Identifying key mediators in muscle-brain communication pathways from complex proteomics datasets .

What are the optimal 3D culture systems for studying CTSB functions in tumor microenvironments?

Three-dimensional culture systems offer powerful platforms for dissecting CTSB functions in tumor microenvironments:

  • Tumor spheroid models:

    • Single-cell-derived tumor spheroids enable assessment of CTSB effects on collective invasive behavior.

    • CTSB overexpression in PyMT tumor cells enhances invasive sprouting in spheroid models, mimicking in vivo invasion patterns .

  • Tumor-stroma coculture systems:

    • Cocultures of tumor cells with bone marrow-derived macrophages allow investigation of tumor-stroma interactions.

    • These systems have revealed that macrophages enhance tumor spheroid invasiveness regardless of their CTSB status, while CTSB overexpression in tumor cells further promotes invasion .

  • Extracellular matrix composition:

    • Varying matrix composition (collagen I, Matrigel, or defined matrix mixtures) to assess substrate-specific CTSB effects.

    • DQ-collagen IV fluorogenic substrates incorporated into matrices enable direct visualization of proteolytic activity .

  • Inducible expression systems in 3D cultures:

    • DOX-inducible CTSB expression allows temporal control over CTSB activity during specific phases of spheroid formation or invasion.

    • This approach has demonstrated that CTSB induction in tumor cells, but not in macrophages, increases spheroid invasiveness .

  • Live imaging capabilities:

    • Time-lapse microscopy of 3D cultures permits dynamic assessment of invasion processes.

    • Fluorescently labeled cells or matrices enable tracking of cell movement and matrix degradation in real-time.

  • Microfluidic systems:

    • Microfluidic devices with tumor spheroids and defined gradients of soluble factors can model invasion in response to chemotactic stimuli.

    • These systems allow precise control over the spatial arrangement of different cell types and matrix components.

These 3D culture approaches offer significant advantages over traditional 2D systems by better recapitulating the complex cellular interactions and physical constraints of in vivo tumor microenvironments, while still allowing precise experimental manipulation and detailed observation of CTSB-mediated processes.

How do findings from CTSB mouse models inform potential therapeutic strategies for human diseases?

Findings from CTSB mouse models suggest several promising therapeutic strategies:

  • Cancer therapy approaches:

    • CTSB inhibition as an anti-cancer strategy, particularly for invasive cancers like breast cancer.

    • Cell type-specific targeting of CTSB inhibition to cancer cells rather than stromal cells, based on findings that cancer cell CTSB is primarily responsible for promoting invasion .

    • Combining CTSB inhibitors with other therapies targeting complementary pathways in tumor progression.

  • Neurodegenerative disease strategies:

    • Muscle-specific CTSB enhancement as a potential therapy for Alzheimer's Disease, based on findings that muscle CTSB expression prevents cognitive decline in AD mouse models .

    • Exercise-based interventions to naturally increase CTSB levels, as exercise upregulates circulating CTSB in humans .

    • Development of CTSB mimetics or delivery systems that could replicate beneficial effects of muscle-derived CTSB.

  • Context-specific approaches:

    • Careful consideration of disease context, as CTSB appears to have opposing effects in healthy versus pathological states .

    • Personalized medicine approaches that consider individual disease contexts and potential response to CTSB modulation.

  • Biomarker applications:

    • Using circulating CTSB levels as biomarkers for therapeutic efficacy of exercise or other interventions in neurodegenerative diseases.

    • Monitoring CTSB activity in tumors as a potential prognostic indicator.

  • Combination therapies:

    • In cancer, combining CTSB inhibitors with anti-metastatic or anti-angiogenic agents.

    • In neurodegeneration, combining CTSB-enhancing approaches with traditional AD therapeutics.

The translation of these findings requires careful consideration of species differences in CTSB biology and disease pathophysiology, as well as development of appropriate delivery systems and clinical trial designs that account for the context-dependent effects of CTSB modulation.

What methodological considerations are essential when comparing CTSB function between mouse models and human samples?

Comparing CTSB function between mouse models and human samples requires careful methodological considerations:

  • Species-specific differences:

    • Human and mouse CTSB have structural and functional differences that must be accounted for in comparative studies.

    • Studies involving human CTSB overexpression in mouse models, as in the PyMT breast cancer model, provide valuable insights into human CTSB function in vivo .

  • Disease model fidelity:

    • Assessment of how accurately mouse models recapitulate human disease pathophysiology.

    • For example, APP/PS1 mice model certain aspects of AD but may not fully replicate the human disease complexity .

  • Tissue-specific expression patterns:

    • Comparative analysis of CTSB expression patterns across tissues in mice versus humans.

    • Consideration of potential differences in regulatory mechanisms controlling CTSB expression.

  • Standardized measurement techniques:

    • Use of consistent methodologies for measuring CTSB expression and activity across species.

    • Development of species-neutral activity assays that are equally sensitive to mouse and human CTSB.

  • Patient-derived xenograft models:

    • Use of human tumor samples in immunocompromised mice to better model human CTSB function in cancer.

    • Assessment of CTSB in patient-derived organoids as an intermediate between cell lines and in vivo models.

  • Humanized mouse models:

    • Engineering mice with humanized CTSB genomic loci to better model human CTSB regulation.

    • Using CRISPR-based approaches to introduce human CTSB variants into mouse models.

  • Translational validation pipeline:

    • Systematic validation of mouse model findings in human samples.

    • Parallel analysis of mouse model tissues and human patient samples using identical methodologies.

These methodological considerations are essential for accurately translating findings from mouse models to human applications and for developing effective therapeutic strategies based on CTSB biology.

What are the optimal conditions for measuring CTSB enzymatic activity in mouse tissue samples?

Measuring CTSB enzymatic activity in mouse tissues requires careful attention to experimental conditions:

Sample Preparation Protocol:

  • Tissue collection and storage:

    • Rapid tissue harvesting and flash-freezing in liquid nitrogen to preserve enzymatic activity.

    • Storage at -80°C until analysis to prevent degradation.

  • Homogenization buffer composition:

    • Buffer containing 50 mM sodium acetate (pH 5.5), 1 mM EDTA, 0.1% Triton X-100.

    • Addition of protease inhibitor cocktail lacking cysteine protease inhibitors.

    • Maintenance of acidic pH (5.0-5.5) to preserve CTSB activity.

  • Homogenization technique:

    • Gentle mechanical disruption using a Dounce homogenizer or tissue grinder.

    • Keeping samples on ice during processing to minimize protein degradation.

Activity Assay Protocol:

  • Fluorogenic substrate selection:

    • Z-Arg-Arg-AMC is highly specific for CTSB.

    • Z-Phe-Arg-AMC can detect CTSB but also reacts with other cathepsins.

  • Assay conditions:

    • Buffer: 100 mM sodium acetate (pH 5.5), 1 mM EDTA, 2 mM DTT.

    • Temperature: 37°C for reaction.

    • Substrate concentration: 20-50 μM (determine optimal concentration for each tissue type).

  • Specificity controls:

    • Include CA-074 or CA-074Me (specific CTSB inhibitors) in parallel reactions.

    • The difference between total activity and activity in the presence of inhibitor represents CTSB-specific activity.

  • Calibration:

    • Use AMC standards for accurate quantification.

    • Generate standard curves under identical buffer conditions.

  • Normalization:

    • Normalize activity to total protein content determined by Bradford or BCA assay.

    • Consider alternative normalization to DNA content or tissue weight as appropriate.

Troubleshooting:

  • Low activity levels:

    • Check pH of buffers (CTSB optimal activity at pH 5.0-5.5).

    • Ensure reducing conditions with fresh DTT.

    • Verify sample storage conditions haven't compromised activity.

  • High background:

    • Increase specificity by using more selective substrates or inhibitors.

    • Optimize substrate concentration to minimize non-specific hydrolysis.

  • Variable results between replicates:

    • Standardize tissue collection time and processing procedures.

    • Account for potential diurnal variations in CTSB activity.

These optimized conditions ensure reliable and reproducible measurement of CTSB activity in mouse tissue samples.

What are the key considerations for generating and validating inducible CTSB expression systems in mice?

Generating and validating inducible CTSB expression systems involves several critical considerations:

System Design:

  • Vector selection:

    • Lentiviral vectors like pTRIPZ provide efficient delivery and stable integration for long-term expression .

    • AAV vectors enable tissue-specific targeting without genomic integration .

  • Promoter choice:

    • Cell-type specific promoters for targeted expression.

    • Tetracycline-responsive elements (TRE) for temporal control with doxycycline.

    • Consider the specific variant of CTSB cDNA, as human cancers predominantly express variant 1 .

  • Induction mechanism:

    • Tet-On systems provide tighter control than Tet-Off systems.

    • rtTA3 transactivator offers improved sensitivity to doxycycline.

Validation Protocol:

  • Expression kinetics:

    • Determine time course of induction (mRNA detection within 6h, pro-CTSB protein by 6h, mature CTSB forms by 10h after DOX addition) .

    • Assess persistence after DOX removal (mature enzyme stable for >4 days) .

    • Optimize DOX dosage for desired expression levels.

  • Expression level verification:

    • qRT-PCR for transcript quantification.

    • Western blotting to confirm protein expression and processing of pro-CTSB to mature forms .

    • Aim for physiologically relevant expression levels comparable to endogenously expressed CTSB in disease states.

  • Functional validation:

    • Enzymatic activity assays to confirm that induced CTSB is catalytically active.

    • Substrate degradation assays (e.g., DQ-collagen IV) to verify extracellular proteolytic activity .

    • Phenotypic assays relevant to the research context (e.g., invasion assays for cancer studies) .

  • Specificity controls:

    • Empty vector controls.

    • Expression of catalytically inactive CTSB mutants.

    • Parallel induction of other proteases to control for non-specific effects.

Potential Challenges:

  • Leaky expression:

    • Background expression in the absence of inducer.

    • Use second-generation Tet-On systems with reduced leakiness.

    • Include uninduced controls in all experiments.

  • Variable expression levels:

    • Clone selection or cell sorting may be necessary for homogeneous expression.

    • For in vivo studies, consider reporter genes to identify cells expressing the transgene.

  • Doxycycline delivery issues:

    • Optimize doxycycline delivery method (drinking water vs. food vs. injection).

    • Monitor doxycycline levels in blood to ensure consistent exposure.

  • Long-term expression stability:

    • Test for potential silencing of transgene over multiple passages or time points.

    • Consider site-specific integration systems for stable long-term expression.

These considerations ensure the generation of reliable inducible CTSB expression systems that enable precise temporal control of CTSB activity for mechanistic studies.

What emerging technologies could advance our understanding of CTSB biology in mouse models?

Several cutting-edge technologies hold promise for advancing CTSB research:

  • Single-cell multi-omics approaches:

    • Single-cell RNA/protein co-detection to correlate CTSB expression with cellular phenotypes.

    • Single-cell proteomics to profile CTSB-dependent changes in protein expression at cellular resolution.

    • Spatial transcriptomics to map CTSB expression patterns in complex tissues with spatial context.

  • Advanced imaging technologies:

    • Activity-based probes for live imaging of CTSB activity in vivo.

    • Intravital microscopy to visualize CTSB-dependent processes in living animals.

    • Super-resolution microscopy to study subcellular localization and trafficking of CTSB.

    • Light-sheet microscopy for 3D visualization of CTSB distribution in intact tissues.

  • CRISPR-based approaches:

    • CRISPRa/CRISPRi for titratable and reversible modulation of CTSB expression.

    • Base editing or prime editing for precise introduction of disease-associated CTSB variants.

    • CRISPR screens to identify genetic modifiers of CTSB function.

  • Protein engineering and synthetic biology:

    • Engineered CTSB variants with altered substrate specificity or cellular localization.

    • Optogenetic control of CTSB activity for spatiotemporal precision.

    • Split-CTSB systems for induced proximity applications.

  • Organoid technologies:

    • Patient-derived organoids for translational studies of CTSB biology.

    • Brain organoids to study CTSB effects on neural development and function.

    • Multi-organ-on-chip approaches to study systemic effects of muscle-derived CTSB.

  • Artificial intelligence approaches:

    • Machine learning for analysis of complex phenotypic data from CTSB mouse models.

    • AI-driven prediction of CTSB substrates and interaction partners.

    • Deep learning for image analysis of CTSB localization and activity patterns.

  • Ex vivo tissue platforms:

    • Precision-cut tissue slices for maintaining native tissue architecture while allowing experimental manipulation.

    • Skeletal muscle-brain co-culture systems to study myokine effects directly.

These emerging technologies will enable more precise, comprehensive, and physiologically relevant investigation of CTSB biology across different disease models and cellular contexts.

What are the most promising directions for understanding muscle-brain CTSB signaling mechanisms?

Several promising research directions could advance our understanding of muscle-brain CTSB signaling:

  • Defining the muscle secretome:

    • Comprehensive characterization of factors co-secreted with CTSB from skeletal muscle.

    • Identification of potential carrier proteins that facilitate CTSB transport in circulation.

    • Investigation of exercise-dependent changes in CTSB post-translational modifications that might affect its secretion or activity.

  • Blood-brain barrier (BBB) interactions:

    • Determining whether CTSB directly crosses the BBB or acts through intermediary signaling molecules.

    • Investigating potential CTSB receptors on BBB endothelial cells or transport mechanisms.

    • Exploring whether CTSB alters BBB permeability to other factors relevant to neurological function.

  • Cellular targets in the brain:

    • Identification of brain cell types (neurons, astrocytes, microglia) that respond to muscle-derived CTSB.

    • Characterization of neuronal subtypes particularly sensitive to CTSB signaling.

    • Investigation of whether CTSB acts directly on neural stem/progenitor cells to enhance neurogenesis .

  • Molecular signaling pathways:

    • Elucidation of downstream signaling pathways activated by CTSB in target cells.

    • Investigation of potential proteolytic targets of CTSB in the brain that mediate its effects.

    • Exploration of transcriptional programs induced by CTSB signaling in neural cells.

  • Context-dependent mechanisms:

    • Systematic comparison of CTSB effects in healthy versus diseased brain states.

    • Investigation of age-dependent differences in responsiveness to muscle-derived CTSB.

    • Exploration of how other factors (inflammation, metabolic state) modify CTSB signaling efficacy.

  • Translation to human biology:

    • Correlation of circulating CTSB levels with cognitive performance in humans with neurodegenerative conditions.

    • Exploration of genetic variants in CTSB or its signaling pathway that might affect cognitive resilience in humans.

    • Development of blood-based biomarkers to monitor muscle-brain CTSB signaling in humans.

  • Therapeutic development:

    • Design of CTSB mimetics that could replicate beneficial effects without requiring muscle expression.

    • Development of targeted delivery systems to enhance CTSB activation specifically in muscle tissue.

    • Identification of exercise regimens optimized for enhancing CTSB secretion in humans.

These research directions would significantly advance our understanding of how muscle-derived CTSB contributes to brain health and could lead to novel therapeutic strategies for neurodegenerative diseases.

Product Science Overview

Structure and Function

Cathepsin B is a papain-family cysteine protease that is normally located in lysosomes . It possesses both endopeptidase and exopeptidase activities, meaning it can act on peptide bonds within a protein as well as at the ends of peptide chains . This dual functionality allows Cathepsin B to process a variety of proteins, including pro and active caspases, prorenin, and secretory leucoprotease inhibitor (SLPI) .

Biological Roles

Cathepsin B plays several critical roles in maintaining normal cellular metabolism. It is involved in the turnover of proteins within lysosomes, contributing to the degradation and recycling of cellular components . Additionally, Cathepsin B is implicated in several pathological conditions, such as tumor progression and arthritis .

Recombinant Mouse Cathepsin B

Recombinant Mouse Cathepsin B is produced using various expression systems, such as HEK293 cells or mouse myeloma cell lines . The recombinant protein is typically tagged with a His-tag for purification purposes and is available in both carrier-free and carrier-containing formulations . The purity of these recombinant proteins is generally greater than 95%, as determined by SDS-PAGE .

Applications

Recombinant Mouse Cathepsin B is widely used in research to study its enzymatic activity and role in various biological processes. It is also used in assays to measure the cleavage of specific substrates, providing insights into its function and regulation .

Storage and Stability

Lyophilized recombinant Cathepsin B proteins are stable for up to 12 months when stored at -20 to -80°C . Once reconstituted, the protein solution can be stored at 4-8°C for 2-7 days or at < -20°C for up to 3 months .

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.