The Ctsz gene is located on chromosome 2 (20q13.32 in humans) and spans six exons .
Two transcript variants exist, but only one is fully characterized .
Unique structural motifs: A three-amino acid insertion in the conserved catalytic region distinguishes CTSZ from other cysteine cathepsins .
Enzymatic activity: Functions as a carboxypeptidase with mono-/di-peptidase activity but lacks endopeptidase function .
Expression: Ubiquitous in cancer cell lines, immune cells (monocytes, macrophages, dendritic cells), and primary tumors .
Feature | Description |
---|---|
Gene Location | Chromosome 2 (Mus musculus) |
Protein Size | 33–38 kDa (glycosylated) |
Catalytic Activity | Exopeptidase (C-terminal cleavage) |
Substrates | Integrins, γ-enolase, PRLP0 |
Breast cancer: In PyMT-driven mammary tumor models, Ctsz deficiency delayed tumor onset and reduced lung metastasis by 70–80% .
Pancreatic neuroendocrine tumors (PanNETs): Ctsz knockout (Ctsz−/−) mice exhibited a 63% reduction in tumor volume and improved survival .
Hepatocellular carcinoma (HCC): Overexpression of CTSZ increased metastasis via epithelial-mesenchymal transition (EMT), downregulating E-cadherin and upregulating vimentin .
Combined Ctsb and Ctsz deficiencies in breast cancer models reduced tumor burden additively, suggesting compensatory mechanisms between these proteases .
Ctsz−/− mice exposed to silica crystals showed reduced pulmonary inflammation and IL-1β secretion compared to wild-type mice .
Mechanistically, CTSZ facilitates NLRP3 inflammasome activation extracellularly, independent of transcriptional regulation .
CTSZ exhibits a protective role in gastric mucosa during Helicobacter pylori infection, likely through non-proteolytic mechanisms .
CTSZ interacts with proteins critical for cellular adhesion, signaling, and cytoskeletal organization:
Integrins: Binds via an RGD motif, modulating cell migration .
Kinesins (KIFC1) and kinases (PLK2): Implicated in mitotic regulation .
Inflammasome components: Regulates IL-1β maturation in macrophages .
Protein | Function | Citation |
---|---|---|
Integrin α5β1 | Cell adhesion and migration | |
γ-Enolase | Glycolytic enzyme and plasminogen receptor | |
PRLP0 | Ribosomal protein synthesis |
Ctsz−/− mice are viable and fertile but show attenuated tumor growth and inflammation .
Double knockouts (Ctsb−/− Ctsz−/−) exhibit additive tumor suppression, underscoring functional overlap .
Active recombinant mouse CTSZ (His-tagged) is used to study enzymatic activity, with >1,200 pmol/min/μg specificity for fluorogenic substrates .
CTSZ is widely expressed across multiple tissues in mice, with particularly high expression in immune cells, especially macrophages. Following mycobacterial infection, CTSZ expression is significantly upregulated in macrophages, positioning it as a key player in the immune response . Transcriptional analyses across diverse species (mice, humans, macaques, and zebrafish) consistently demonstrate that cathepsin Z expression is highest in macrophages post-infection, suggesting evolutionary conservation of this response pattern . This expression pattern places CTSZ at critical host-pathogen interfaces, particularly within pulmonary microenvironments during infectious challenges.
Unlike other cathepsins that primarily function as proteases, CTSZ possesses unique dual functionality. It maintains proteolytic activity while also containing an Arg-Gly-Asp (RGD) motif that enables direct interaction with integrins . This distinguishing feature allows CTSZ to participate in cellular adhesion, migration, and signaling pathways independent of its proteolytic activity. In mouse models, this dual functionality translates to context-dependent roles in disease processes, with different mechanisms predominating depending on the pathological setting and cellular source of CTSZ expression.
CTSZ deficiency substantially impacts TB progression in mouse models, with Ctsz−/− mice demonstrating significantly increased susceptibility. Following aerosol infection with Mycobacterium tuberculosis (Mtb), Ctsz−/− mice exhibit higher bacterial burdens in the lungs at both 2 weeks (4.09 log10 CFU vs. 3.41 in wild-type; p<0.05) and 4 weeks (5.17 log10 CFU vs. 4.09 in wild-type; p<0.05) post-infection . This compromised bacterial control correlates with earlier dissemination to the spleen and increased mortality. The data demonstrate that CTSZ plays a protective role during TB infection, with its absence leading to dysregulated immune responses and failure to contain bacterial replication.
The inflammatory profile of Ctsz−/− mice during TB infection reveals significant dysregulation of several key cytokines. At 4 weeks post-infection, these mice show elevated levels of TH1-associated cytokines including TNF-α (p=0.019) and IL-1β (p=0.016), coupled with reduced levels of GM-CSF (p=3.8e-06), IL-6 (p=5.9e-04), LIF (p=6.6e-07), and VEGF (p=6.6e-07) compared to wild-type controls . Most notably, CXCL1 levels are consistently elevated in Ctsz−/− mice throughout infection, with this chemokine emerging as a dominant feature in distinguishing Ctsz−/− from wild-type responses in unsupervised analysis (sPLS-DA). This CXCL1 overproduction is particularly significant as it serves as a biomarker of active TB disease severity in both mice and humans .
Current evidence suggests CTSZ mediates TB susceptibility through a conserved CTSZ-CXCL1 axis. In the absence of CTSZ, there is dysregulated production of CXCL1, a neutrophil chemoattractant associated with severe TB disease . This dysregulation occurs in a cell-autonomous manner within macrophages, as demonstrated by elevated CXCL1 production in Ctsz−/− bone marrow-derived macrophages following mycobacterial infection. Additionally, CTSZ's interaction with cell surface integrins, including lymphocyte function-associated antigen-1 (LFA-1) and macrophage-1 (Mac-1) antigen, likely influences phagocytosis and immune cell migration during infection . The presence of CTSZ in CD68+ macrophages within patient-derived pulmonary granulomas further supports its role at the host-pathogen interface, where it appears to modulate critical immune containment mechanisms.
Mouse model findings show remarkable concordance with human TB studies. Research from a Ugandan household contact study identified significant associations between CTSZ variants and TB disease severity, with 4 SNPs and 1 INDEL in the CTSZ gene significantly correlating with clinical outcomes . Immunohistochemistry of patient-derived TB granulomas confirms CTSZ localization within CD68+ macrophages, positioning CTSZ directly at the host-pathogen interface in human disease . The consistency of the CTSZ-CXCL1 relationship across species supports a conserved mechanism, where CTSZ deficiency or dysfunction leads to CXCL1 overproduction and increased disease severity. This cross-species validation strengthens the translational relevance of mouse model findings to human TB pathogenesis.
The cellular source of CTSZ significantly impacts its function in tumor progression, revealing context-dependent roles. Through elegant bone marrow transplantation experiments in the RIP1-Tag2 PanNET model, researchers demonstrated that tumor cell-derived CTSZ primarily drives tumor growth, while tumor-associated macrophage (TAM)-derived CTSZ predominantly promotes tumor invasion . Specifically, transplantation of wild-type bone marrow into Ctsz−/− tumor-bearing mice could not rescue tumor growth deficits but did enhance tumor invasiveness . These findings highlight the importance of considering cellular source when studying CTSZ functions, as the same protein can serve distinct roles depending on whether it originates from tumor cells or from stromal components such as TAMs.
CTSZ influences tumor cell behavior through at least two distinct molecular mechanisms:
RGD motif-integrin interactions: CTSZ contains a unique Arg-Gly-Asp (RGD) motif that mediates binding to integrins, facilitating cellular adhesion, migration, and signaling . This interaction appears critical for many of CTSZ's tumor-promoting functions and represents a mechanism independent of its proteolytic activity.
EMT induction: CTSZ overexpression drives epithelial-mesenchymal transition by altering the expression of key markers. It decreases epithelial markers (E-cadherin, α-catenin, and β-catenin) while increasing mesenchymal markers (fibronectin and N-cadherin) . This reprogramming enhances cellular motility and invasive capacity, contributing to metastatic potential.
These mechanisms appear to operate simultaneously but may have different relative contributions depending on the cancer type, stage, and microenvironmental context.
Several experimental approaches have proven particularly valuable in elucidating CTSZ's role in cancer metastasis:
Genetic modification models: Both knockout (Ctsz−/−) and overexpression models have been instrumental. Crossing RIP1-Tag2 mice with Ctsz−/− mice demonstrated CTSZ's importance in tumor growth and invasion , while stable overexpression in cell lines like QGY-7703 revealed its metastasis-promoting properties .
Bone marrow transplantation: This technique has been crucial for distinguishing between tumor cell-derived and stromal cell-derived CTSZ functions. By transplanting wild-type or Ctsz−/− bone marrow into tumor-bearing mice of either genotype, researchers identified cell source-specific roles .
In vitro migration and invasion assays: Wound healing assays and invasion chamber experiments have consistently demonstrated CTSZ's ability to enhance cellular motility and matrix invasion .
In vivo metastasis models: Tail vein injection of CTSZ-overexpressing cells into SCID mice, followed by quantification of liver metastases, has provided definitive evidence of CTSZ's pro-metastatic function .
These complementary approaches provide robust evidence for CTSZ's multifaceted roles in cancer progression.
For comprehensive CTSZ characterization in mouse tissues, a multi-modal approach yields the most reliable results:
Expression Analysis:
RT-qPCR: Provides sensitive quantification of Ctsz mRNA expression across tissues, with particularly reliable results when normalized to appropriate housekeeping genes.
Western blotting: Enables protein-level quantification using specific anti-CTSZ antibodies (commercially available from vendors like Santa Cruz Biotechnology) .
Immunohistochemistry/Immunofluorescence: Allows visualization of CTSZ localization within tissue microenvironments, particularly valuable for identifying cell type-specific expression patterns in complex tissues such as granulomas or tumors .
Activity Assessment:
Enzymatic activity assays: Using specific substrates that release detectable products when cleaved by CTSZ.
Functional assays: Including adhesion, migration, and invasion assays that capture CTSZ's functional impact beyond enzymatic activity .
The combination of these approaches provides comprehensive profiling of both expression levels and functional activity across different experimental conditions.
When designing experiments with Ctsz knockout mice, several critical considerations must be addressed:
Genetic background effects: Phenotypes may vary depending on the background strain. The studies cited primarily used C57BL/6 background , and phenotypes might differ in other genetic backgrounds.
Compensation by other cathepsins: Potential compensatory upregulation of functionally related cathepsins should be assessed, as this may mask or modify knockout phenotypes.
Tissue-specific vs. global knockout: Global Ctsz knockout affects all tissues, potentially confounding interpretation of tissue-specific effects. Consider tissue-specific or inducible knockout systems for more targeted analysis.
Age-dependent effects: CTSZ has been implicated in aging processes , suggesting that phenotypes may vary with mouse age. Age-matched controls are essential, and analyzing multiple age points may reveal temporal dynamics.
Challenge model selection: Ctsz−/− phenotypes become most apparent under challenge conditions (infection, cancer, etc.), necessitating careful selection of appropriate disease models and control conditions.
Cell-autonomous vs. non-cell-autonomous effects: As demonstrated by the distinct roles of tumor-derived versus TAM-derived CTSZ , determining the cellular source of relevant CTSZ activity is crucial for interpretation.
Dissecting CTSZ's dual functionality requires experimental approaches that can distinguish between proteolytic activity and integrin binding:
Mutant constructs:
Protease-dead mutants (with catalytic site mutations) that retain the RGD motif
RGD motif mutants (e.g., RGD→RGE) that preserve proteolytic activity
Double mutants affecting both functions
Selective inhibitors:
Protease inhibitors targeting CTSZ's catalytic activity
RGD-mimetic peptides that compete for integrin binding without affecting proteolytic function
Rescue experiments:
Complementing Ctsz−/− cells or mice with wild-type or function-specific mutants to determine which activities restore specific phenotypes
Domain-specific antibodies:
Antibodies targeting either the catalytic domain or the RGD-containing region can selectively block specific functions
Integrin knockout/knockdown:
Using cells/mice lacking specific integrins known to interact with CTSZ's RGD motif to isolate effects dependent on this interaction
These approaches, particularly when used in combination, can effectively separate CTSZ's proteolytic functions from its adhesion/signaling roles mediated by the RGD motif.
CTSZ likely exhibits disease-specific functions across inflammatory conditions due to contextual differences in immune response patterns, affected tissues, and triggering agents. In tuberculosis models, CTSZ appears protective by regulating CXCL1 production and subsequent neutrophil recruitment . This contrasts with its potentially detrimental role in certain autoimmune disease models where excessive inflammatory regulation might be harmful. The cellular composition of inflammatory infiltrates likely influences CTSZ's impact, as its prominent expression in macrophages positions it to modulate myeloid cell responses differentially depending on the inflammatory stimulus . Additionally, CTSZ's integrin-binding capacity through its RGD motif may have varying significance depending on which integrins predominate in specific disease microenvironments . Future comparative studies across multiple inflammatory disease models could reveal condition-specific regulatory mechanisms and identify commonalities in CTSZ's immunomodulatory functions.
Targeting CTSZ presents distinct therapeutic opportunities across disease contexts:
Tuberculosis: Enhancing CTSZ function might improve bacterial control and reduce disease severity by normalizing CXCL1 levels and improving granuloma integrity . Small molecules or biologics that boost CTSZ expression or activity specifically in macrophages could represent novel host-directed therapeutics.
Cancer: Inhibiting CTSZ could potentially reduce tumor growth and metastasis . Dual-targeting approaches addressing both proteolytic activity and RGD-integrin interactions might prove most effective, potentially via:
Catalytic site inhibitors
RGD-mimetic peptides blocking integrin interactions
Antibodies targeting CTSZ's functional domains
Cell type-specific delivery systems to target tumor cells versus TAMs
Temporal considerations given CTSZ's varying roles at different disease stages
Combination with conventional therapies (antibiotics for TB; chemotherapy for cancer)
Given CTSZ's involvement in normal physiological processes, careful evaluation of potential side effects will be essential during therapeutic development.
Comparative analysis of mouse and human CTSZ genetic variations reveals important parallels in disease susceptibility patterns:
Five variants in CTSZ (4 SNPs and 1 INDEL) show significant association with TB severity in Ugandan populations
These variations may influence CTSZ expression levels or function, potentially affecting the CTSZ-CXCL1 regulatory axis
The TB susceptibility locus Tip5 in mice contains the Ctsz gene, with susceptible strains producing lower CTSZ protein levels
This mirrors human findings where CTSZ variants associate with disease severity
Both species demonstrate consistent CTSZ localization in macrophages within granulomas
Mouse models carrying human CTSZ variants could help validate causality and explore mechanisms
Humanized mouse models expressing human CTSZ variants might better recapitulate patient-specific disease phenotypes
Cross-species conservation of the CTSZ-CXCL1 relationship supports the translational relevance of mouse findings to human disease
These comparisons strengthen the value of mouse models for understanding CTSZ's role in human diseases while highlighting the importance of validating findings across species.
Several technological advances would significantly enhance CTSZ research in mouse models:
CRISPR-based temporal and spatial control systems:
Inducible, cell type-specific CTSZ knockout or overexpression
Precise editing of specific domains (RGD motif, catalytic site) to dissect functions
Base editing to introduce specific disease-associated variants
Advanced imaging technologies:
Intravital microscopy to track CTSZ-expressing cells in real-time during disease progression
Activity-based probes to visualize active CTSZ within living tissues
Multiplexed imaging to simultaneously visualize CTSZ, CXCL1, and cellular markers
Single-cell multi-omics approaches:
Single-cell RNA/protein profiling to characterize CTSZ expression heterogeneity
Spatial transcriptomics to map CTSZ expression relative to microenvironmental features
Integrated multi-omic analyses to correlate CTSZ with downstream signaling pathways
Improved mouse models:
Humanized models expressing human CTSZ variants
Reporter mice with fluorescently tagged CTSZ for live tracking
Conditional alleles allowing separation of CTSZ's proteolytic and RGD-motif functions
Computational approaches:
Machine learning to identify patterns in CTSZ-dependent disease progression
Systems biology modeling of CTSZ-CXCL1 regulatory networks
Predictive modeling for therapeutic targeting strategies
These technologies would collectively enable more precise dissection of CTSZ's complex roles across different pathological contexts.
This comparative analysis highlights the context-dependent nature of CTSZ functions, with protective roles in infectious disease contrasting with detrimental effects in cancer progression. The consistent involvement of macrophage-derived CTSZ across models underscores its importance in modulating immune responses in disease microenvironments.
Research Context | Key Experimental Readouts | Technical Approaches | Significance |
---|---|---|---|
TB Infection Models | Bacterial burden (CFU) | Lung/spleen homogenization and plating | Quantifies impact on bacterial control |
Cytokine/chemokine levels (especially CXCL1) | Multiplex cytokine assays, ELISA | Reveals inflammatory dysregulation | |
Granuloma organization | Histopathology, immunofluorescence | Assesses structural containment | |
Survival | Mortality analysis | Ultimate measure of disease outcome | |
Cancer Models | Tumor volume/number | Caliper measurement, nodule counting | Quantifies growth impact |
Invasion assessment | Histological grading of invasive fronts | Evaluates local spread | |
Metastatic burden | Distant organ nodule quantification | Measures systemic spread | |
EMT marker expression | IF/IHC, Western blot | Mechanisms of invasion/metastasis | |
Cell-Based Assays | Migration capacity | Wound healing, transwell migration | Cell motility assessment |
Invasion ability | Matrigel invasion assays | Matrix penetration capacity | |
CXCL1 production | ELISA of culture supernatants | Cell-autonomous cytokine regulation | |
Protein-protein interactions | Co-IP, proximity ligation | Molecular mechanism insights |
This comprehensive panel of readouts enables thorough characterization of CTSZ functions across different experimental systems, facilitating integration of findings across disease contexts and research groups.
Cathepsin-Z exhibits mono- and di-peptidase activity at its C-terminus, unlike cathepsin B, which acts as an endopeptidase . This enzyme is restricted to the cells of the immune system, predominantly monocytes, macrophages, and dendritic cells . It is widely expressed in human tissues, suggesting its involvement in normal intracellular protein degradation across various cell types .
The recombinant form of Cathepsin-Z (Mouse) is typically expressed in HEK293 cells and is purified to a high degree of purity (>95%) as determined by SDS-PAGE . The biological activity of recombinant Cathepsin-Z is measured by its ability to cleave the fluorogenic peptide substrate, Mca-RPPGFSAFK(Dnp)-OH . This specific activity is greater than 1,200 pmoles/min/μg .
Cathepsin-Z is involved in various physiological and pathological processes. It is capable of cleaving regulatory motifs at the C-terminus, affecting the function of targeted molecules . This enzyme may also regulate the maturation of dendritic cells, which is crucial in the initiation of adaptive immunity . Higher levels of Cathepsin-Z are found in tumor and immune cells of prostate and gastric carcinomas, as well as in macrophages of gastric mucosa, especially after infection by Helicobacter pylori . This suggests that Cathepsin-Z may play a role in tumor progression and immune response regulation.