CTSZ Antibody

Cathepsin-Z, Mouse Anti Human
Shipped with Ice Packs
In Stock

Description

Structure and Function of CTSZ

CTSZ, encoded by the CTSZ gene, is a 34 kDa enzyme with carboxypeptidase activity, acting on peptide bonds at the C-terminal end of proteins. Key features include:

  • Unique Pro-Region: A short, 41-amino-acid pro-region lacking conserved motifs (e.g., ERFNIN) found in other cysteine cathepsins .

  • Dual Roles:

    • Immune Function: Facilitates T-cell adhesion and migration through endothelial barriers .

    • Tumor Progression: Overexpression in cancers (e.g., prostate, breast, liver) promotes metastasis via epithelial-mesenchymal transition (EMT) and extracellular matrix remodeling .

Types and Applications of CTSZ Antibodies

CTSZ Antibodies are primarily used in molecular biology for detecting CTSZ protein expression. Below is a comparative overview of available products:

CharacteristicMonoclonal (Mouse) Polyclonal (Rabbit)
Clone/SourcePAT6G11AT (Mouse)Recombinant fragment (Human aa 150-250)
ApplicationsELISA, Western BlotWB, IHC, ICC/IF, IP
ReactivityHumanHuman, Mouse, Rat
Key UsesCancer cell line analysisTissue localization in tumors

Table 1: CTSZ Antibody Applications and Reactivity

ApplicationRecommended DilutionTested Samples
Western Blot1:500–1:3000 HeLa cells, mouse heart
Immunohistochemistry1:50–1:500 Human pancreas, breast tumor
Immunoprecipitation0.5–4.0 µg/mg lysate HeLa cells

Diagnostic Potential in Prostate Cancer

  • Biomarker Utility: Decreased CTSZ mRNA in blood cells correlates with prostate cancer (PCa) diagnosis and biochemical relapse .

    • Sensitivity: 63%

    • Specificity: 93.3%

    • AUC: 0.832 (ROC curve analysis) .

  • Mechanistic Insight: Low CTSZ in immune cells reduces T-cell infiltration into tumors, impairing anti-tumor immunity .

Role in Tumor Metastasis

CTSZ overexpression promotes metastasis by:

  • Inducing EMT: Upregulating mesenchymal markers (fibronectin, vimentin) and downregulating epithelial markers (E-cadherin, α-catenin) .

  • Remodeling ECM: Activating matrix metalloproteinases (MMP2, MMP3, MMP9) .

Diagnostic and Therapeutic Implications

  • Prognostic Value: High CTSZ expression in tumors (e.g., lung, breast) correlates with poor survival .

  • Biomarker Complementarity: CTSZ mRNA in blood complements PSA testing for PCa, reducing unnecessary biopsies .

Challenges and Future Directions

  • Heterogeneity: CTSZ’s dual role (tumor-promoting vs immune-protective) requires context-specific targeting .

  • Therapeutic Targeting: Inhibiting CTSZ in tumors while preserving its immune function remains a challenge.

Product Specs

Introduction
Cathepsin-Z (CTSZ), also known as cathepsin X or cathepsin P, is a lysosomal cysteine proteinase belonging to the peptidase C1 family. It exhibits both carboxy-monopeptidase and carboxy-dipeptidase activities. CTSZ is widely expressed in cancer cell lines and primary tumors, and like other members of its family, plays a role in tumor development.
Physical Appearance
A clear, colorless solution that has been sterilized by filtration.
Formulation
The solution contains 1mg/ml of CTSZ antibody in a buffer consisting of PBS at pH 7.4, 10% glycerol, and 0.02% sodium azide.
Storage Procedures
For storage up to 1 month, keep at 4°C. For longer storage, store at -20°C. Avoid repeated freeze-thaw cycles.
Stability / Shelf Life
The product is stable for 12 months when stored at -20°C and for 1 month at 4°C.
Applications
This antibody has been validated for ELISA and Western blot analysis to ensure its specificity and reactivity. However, optimal working dilutions should be determined empirically for each application. A starting dilution of 1:1000 is recommended.
Synonyms
Cathepsin Z preproprotein, Cathepsin Z, CTSX, Cathepsin P, Cathepsin X, CTSZ, Cathepsin-Z.
Purification Method
CTSZ antibody was purified from mouse ascitic fluids by protein-A affinity chromatography.
Type
Mouse Anti Human Monoclonal.
Clone
PAT6G11AT.
Immunogen
Anti-human CTSZ mAb, clone PAT6G11AT, is derived from hybridization of mouse F0 myeloma cells with spleen cells from BALB/c mice immunized with a recombinant human CTSZ protein 62-303 amino acids purified from E. coli.
Ig Subclass
Mouse IgG2b heavy chain and k light chain.

Q&A

What is CTSZ and why are antibodies against it important in research?

CTSZ (cathepsin Z) is a member of the cysteine cathepsins, primarily localized in lysosomes, with distinctive exopeptidase activity. This protein regulates various cellular physiological functions, including adhesion, migration, invasion, and maturation of immune cells . CTSZ antibodies are essential research tools that enable detection, quantification, and functional analysis of this protein across multiple experimental platforms. These antibodies have become increasingly significant as CTSZ has been implicated in neurodegenerative disorders, multiple sclerosis, and various cancers including gastric and prostate cancer. Additionally, variants in the CTSZ gene have been associated with susceptibility to tuberculosis and progression of primary biliary cholangitis (PBC) .

What are the most common applications for CTSZ antibodies in laboratory research?

CTSZ antibodies are utilized across a wide range of experimental applications in research settings. The primary applications include:

  • Western Blot (WB): For detecting and quantifying CTSZ protein in cell or tissue lysates, allowing researchers to assess expression levels and processing of the protein.

  • Enzyme-Linked Immunosorbent Assay (ELISA): Enabling quantitative measurement of CTSZ in biological samples such as serum, plasma, or cell culture supernatants.

  • Immunohistochemistry (IHC): For visualizing CTSZ expression patterns in tissue sections, providing insights into its localization and potential role in normal and pathological contexts.

  • Immunocytochemistry (ICC) and Immunofluorescence (IF): For examining CTSZ distribution within cells.

  • Immunoprecipitation (IP): To isolate CTSZ from complex biological samples for further analysis .

The selection of the appropriate application depends on the specific research question, sample type, and experimental design. For optimal results, researchers should verify the validated applications for each specific CTSZ antibody product, as performance can vary between manufacturers and clones.

How should researchers select the appropriate CTSZ antibody for their specific experimental needs?

Selecting the optimal CTSZ antibody requires careful consideration of multiple factors to ensure experimental success:

  • Species reactivity: Ensure the antibody recognizes CTSZ in your species of interest. Available antibodies offer reactivity with human, mouse, rat, monkey, and other species samples .

  • Antibody type: Choose between:

    • Monoclonal antibodies: Provide high specificity and consistency between lots

    • Polyclonal antibodies: Often offer higher sensitivity but potential batch-to-batch variation

  • Validated applications: Verify that the antibody has been validated for your intended application (WB, ELISA, IHC, etc.) .

  • Target epitope: Select antibodies that target different regions of CTSZ based on your research needs:

    • N-terminal region antibodies

    • C-terminal region antibodies

    • Cleaved-form specific antibodies (e.g., Cleaved-Cathepsin Z at L62)

  • Conjugation needs: Determine if you require unconjugated antibodies or those conjugated with biotin, FITC, HRP, or other tags for specific detection methods .

  • Validation data: Request and review validation data from manufacturers, including positive and negative controls, to ensure the antibody performs as expected in your experimental system.

  • Literature precedent: Search for published studies using specific CTSZ antibodies for similar applications to inform your selection.

A methodical approach to antibody selection helps minimize experimental variables and increases the likelihood of obtaining reliable, reproducible results.

What are the optimal methods for validating CTSZ antibody specificity in experimental systems?

Validating antibody specificity is critical for ensuring reliable experimental results when working with CTSZ. A comprehensive validation approach should include:

  • Knockout/knockdown controls:

    • Use CTSZ knockout cell lines or tissues as negative controls

    • Employ siRNA or shRNA-mediated CTSZ knockdown samples for comparative analysis

    • Perform side-by-side testing of samples with known CTSZ expression levels

  • Peptide competition assays:

    • Pre-incubate the antibody with excess purified CTSZ protein or immunizing peptide

    • Compare blocked antibody signal with unblocked antibody signal

    • A specific antibody will show significantly reduced signal when pre-blocked

  • Multiple antibody verification:

    • Use at least two different antibodies targeting distinct epitopes of CTSZ

    • Concordant results between different antibodies increase confidence in specificity

  • Cross-reactivity testing:

    • Test antibody against related cathepsin family members (e.g., cathepsin B, L, S)

    • Evaluate potential cross-reactivity with other proteins of similar molecular weight

  • Mass spectrometry validation:

    • Perform immunoprecipitation using the CTSZ antibody

    • Analyze precipitated proteins by mass spectrometry

    • Confirm the presence of CTSZ peptides and assess for non-specific proteins

  • Positive and negative tissue controls:

    • Include tissues with known high CTSZ expression (e.g., immune cells) as positive controls

    • Use tissues with minimal CTSZ expression as negative controls

A methodical validation strategy significantly enhances the reliability of subsequent experiments and provides confidence when interpreting results in complex biological systems.

How can researchers effectively use CTSZ antibodies to investigate its role in disease progression?

Investigating CTSZ's role in disease progression requires sophisticated experimental approaches utilizing CTSZ antibodies:

  • Tissue expression profiling:

    • Perform IHC analysis on disease-relevant tissue microarrays

    • Quantify expression differences between normal, early-stage, and advanced-stage disease tissues

    • Correlate CTSZ expression with clinical parameters and disease outcomes

  • Genetic association studies:

    • Examine CTSZ expression in tissues with different CTSZ SNP genotypes

    • For example, in PBC patients, rs163800 SNP was identified as a significant risk factor (OR = 2.22, 95% CI = 1.63–3.02, P = 8.77 × 10⁻⁷) for jaundice-stage progression

    • Use CTSZ antibodies to establish protein-level correlations with genotypes

  • Functional mechanism studies:

    • Employ proximity ligation assays to identify CTSZ interaction partners in disease contexts

    • Use phospho-specific or modified-form specific antibodies to track CTSZ activation states

    • Perform immunofluorescence co-localization studies to track CTSZ trafficking in disease models

  • Therapeutic intervention monitoring:

    • Track changes in CTSZ expression or localization following treatment

    • Develop ELISA protocols using CTSZ antibodies to monitor circulating CTSZ as a biomarker

  • Multi-parameter analysis:

    • Combine CTSZ antibody staining with other disease markers for comprehensive assessment

    • For example, in PBC, analyze CTSZ expression alongside anti-gp210 antibody status

FactorP valueOdds Ratio95% CI
CTSZ rs6138008.77 × 10⁻⁷2.221.63–3.02
Anti-gp210 antibody positive1.01 × 10⁻⁹3.042.15–4.31
Anti-centromere antibody positive0.070.660.41–1.05

Table: Multivariate analysis of risk factors for jaundice-stage progression in PBC

These approaches enable researchers to establish not only correlative but also mechanistic links between CTSZ and disease processes, potentially identifying new therapeutic targets.

What are the critical considerations for optimizing CTSZ antibody performance in Western blot applications?

Optimizing Western blot protocols for CTSZ detection requires careful attention to several key parameters:

  • Sample preparation:

    • Use appropriate lysis buffers containing protease inhibitors to prevent CTSZ degradation

    • Consider that CTSZ is primarily localized in lysosomes, so ensure thorough cell lysis

    • Include both reducing and non-reducing conditions to compare results, as disulfide bonds may affect epitope accessibility

  • Protein loading and separation:

    • Load 15-30 μg total protein for cell lysates; higher amounts may be required for tissue samples

    • Use 10-12% SDS-PAGE gels for optimal resolution around the 33.9 kDa range (CTSZ molecular weight)

    • Include molecular weight markers spanning 10-50 kDa range for accurate size determination

  • Transfer optimization:

    • Use PVDF membranes for higher protein binding capacity

    • Transfer at lower voltage (30V) overnight at 4°C for more efficient transfer of proteins in this size range

  • Blocking and antibody dilution:

    • Test both BSA and non-fat dry milk blocking agents, as CTSZ antibodies may perform differently with each

    • Optimize primary antibody concentration; typical starting dilutions range from 1:500 to 1:2000

    • Extend primary antibody incubation to overnight at 4°C for improved signal-to-noise ratio

  • Detection and troubleshooting:

    • Be aware of potential detection of multiple bands:

      • 33.9 kDa (full-length CTSZ)

      • Lower molecular weight bands (cleaved/processed forms)

      • Higher molecular weight bands (glycosylated forms or complexes)

    • Use positive control lysates from cells known to express CTSZ (e.g., immune cells)

    • If signal is weak, consider signal enhancement systems or increased antibody concentration

  • Quantification considerations:

    • Use appropriate housekeeping controls for normalization

    • When comparing samples, load on the same gel to minimize technical variations

    • For densitometry, capture images within the linear range of detection

Following these methodological guidelines can significantly improve the quality and reproducibility of CTSZ Western blot results, enabling more reliable protein quantification and comparisons between experimental conditions.

How can CTSZ antibodies be utilized to investigate its role in Primary Biliary Cholangitis (PBC)?

CTSZ antibodies provide valuable tools for investigating the association between CTSZ and Primary Biliary Cholangitis (PBC) progression:

  • Genetic-protein correlation studies:

    • Stratify PBC patient samples based on CTSZ SNP genotypes (particularly rs163800)

    • Use CTSZ antibodies in Western blot and IHC to quantify protein expression differences between genotypes

    • Correlate protein levels with disease stage and progression rates

  • Liver biopsy immunoprofiling:

    • Perform IHC using CTSZ antibodies on liver biopsies from PBC patients at different disease stages

    • Analyze CTSZ expression patterns in relation to histopathological features

    • Compare CTSZ expression between jaundice-stage and early-stage PBC patients

  • Multiplex immunofluorescence analysis:

    • Co-stain liver sections with CTSZ antibodies and markers for immune cells, cholangiocytes, and fibrosis

    • Evaluate cellular sources of CTSZ in the PBC microenvironment

    • Assess spatial relationships between CTSZ-expressing cells and areas of bile duct damage

  • Functional studies in experimental models:

    • Utilize CTSZ antibodies to monitor protein expression in animal or cellular models of PBC

    • Track changes in CTSZ expression following experimental interventions

    • Correlate with biochemical markers of cholestasis and disease progression

  • Biomarker development:

    • Develop ELISA protocols using CTSZ antibodies to quantify circulating CTSZ in PBC patient sera

    • Assess potential for CTSZ as a prognostic biomarker alongside established markers

  • Integrated multiparameter analysis:

    • Combine CTSZ antibody-based detection with anti-nuclear antibody status

    • Multivariate analysis has shown that both CTSZ SNP status and anti-gp210 antibody positivity independently contribute to jaundice-stage progression risk :

Risk Factor CombinationRelative RiskObservation
CTSZ rs163800 risk allele + anti-gp210 positiveHighest riskSuggests potential synergistic effect
CTSZ rs163800 risk allele onlyIntermediate riskOR = 2.22, independent risk factor
Anti-gp210 positive onlyIntermediate riskOR = 3.04, independent risk factor
Neither factorBaseline riskReference group

These research approaches can provide deeper insights into how CTSZ contributes to PBC pathogenesis and potentially identify novel therapeutic targets or prognostic markers.

What methodologies are recommended for studying CTSZ in neurodegenerative disorders using specific antibodies?

Investigating CTSZ in neurodegenerative contexts requires specialized approaches utilizing CTSZ antibodies:

  • Brain region-specific expression analysis:

    • Perform immunohistochemistry on post-mortem brain tissues from patients with neurodegenerative disorders and healthy controls

    • Map CTSZ expression across different brain regions with specialized attention to areas affected in specific disorders:

      • Huntington's disease: striatum and cortex

      • Multiple sclerosis: white matter lesions and normal-appearing white matter

      • Other polyglutamine diseases: cerebellum and affected neurons

  • Cellular localization studies:

    • Use fluorescent double-labeling with CTSZ antibodies and neural cell type markers:

      • NeuN for neurons

      • GFAP for astrocytes

      • Iba1 for microglia

      • MBP for oligodendrocytes

    • Assess changes in CTSZ distribution in disease states compared to controls

  • Activity-based probes combined with antibody detection:

    • Employ activity-based probes that bind active CTSZ

    • Follow with antibody detection to distinguish between active and inactive forms

    • Compare activity profiles between diseased and healthy tissues

  • Lysosomal dysfunction assessment:

    • Co-stain with CTSZ antibodies and lysosomal markers (LAMP1, LAMP2)

    • Evaluate lysosomal morphology, distribution, and CTSZ content in disease models

    • Assess lysosomal membrane permeabilization and CTSZ translocation to cytosol

  • Protein aggregation interaction studies:

    • Investigate co-localization of CTSZ with disease-specific protein aggregates:

      • Huntingtin in Huntington's disease

      • Amyloid-β and tau in Alzheimer's disease

      • α-synuclein in Parkinson's disease

    • Assess potential proteolytic processing of aggregation-prone proteins by CTSZ

  • Experimental therapeutic monitoring:

    • Track changes in CTSZ expression, activity, and localization following experimental treatments

    • Correlate with behavioral or neuropathological outcomes in animal models

  • Functional studies in primary neural cultures:

    • Use CTSZ antibodies to monitor protein levels following genetic manipulation

    • Correlate CTSZ expression with neuronal survival, morphology, and function

These methodological approaches can reveal the specific contributions of CTSZ to neurodegenerative pathogenesis and potentially identify targetable pathways for therapeutic intervention.

How can researchers effectively investigate CTSZ's role in immune cell function using specific antibodies?

Investigating CTSZ's role in immune cell function requires sophisticated methodological approaches utilizing CTSZ antibodies:

  • Immune cell subset profiling:

    • Use flow cytometry with CTSZ antibodies to quantify expression across immune cell populations

    • Perform intracellular staining protocols to detect total CTSZ content

    • Consider membrane permeabilization techniques to distinguish between lysosomal and cytosolic CTSZ

  • Activation-dependent expression analysis:

    • Track CTSZ expression changes following immune cell activation:

      • T cells: anti-CD3/CD28 stimulation

      • B cells: anti-IgM, CD40L, cytokine stimulation

      • Monocytes/macrophages: LPS, IFN-γ activation

      • Dendritic cells: TLR ligand stimulation

    • Use Western blot and flow cytometry with CTSZ antibodies to monitor protein levels over activation time course

  • Functional inhibition studies:

    • Combine CTSZ-specific inhibitors with antibody detection to correlate inhibition with functional outcomes

    • Use function-blocking CTSZ antibodies to investigate specific activities

    • Monitor effects on:

      • Cell migration and adhesion

      • Antigen processing and presentation

      • Cytokine production

      • Cell-cell interactions

  • Cellular localization in immune synapses:

    • Perform immunofluorescence microscopy using CTSZ antibodies during:

      • T cell-APC interactions

      • Natural killer cell cytotoxicity events

      • Phagocytosis by macrophages

    • Track CTSZ redistribution during immune cell functional activities

  • Extracellular CTSZ assessment:

    • Use CTSZ antibodies to detect secreted forms in culture supernatants

    • Investigate potential extracellular functions and receptor interactions

  • In situ tissue immune analysis:

    • Perform multiplex immunofluorescence with CTSZ and immune cell markers in tissue sections

    • Focus on lymphoid tissues, inflammatory sites, and tumor microenvironments

    • Assess relationships between CTSZ expression and immune cell functional states

  • Cytotoxicity and proliferation assays:

    • Correlate CTSZ expression with functional readouts including:

      • Antigen-specific cytotoxicity against target cells

      • Proliferative capacity following antigen presentation

      • HLA-dependent cytotoxic responses

These methodological approaches provide a comprehensive framework for understanding CTSZ's multifaceted roles in immune cell biology, with potential implications for immunotherapeutic interventions and biomarker development.

What are common challenges in CTSZ immunodetection and how can researchers overcome them?

Researchers frequently encounter several technical challenges when working with CTSZ antibodies. Here are methodological solutions to these common problems:

  • Multiple band detection in Western blot:

    • Challenge: Observing multiple bands rather than a single 33.9 kDa band

    • Solutions:

      • Verify if bands represent different glycosylation states or processing forms of CTSZ

      • Run positive control lysates alongside experimental samples for comparison

      • Use more stringent washing conditions to reduce non-specific binding

      • Consider testing antibodies targeting different epitopes of CTSZ

      • Include protease inhibitors in sample preparation to prevent degradation

  • Low signal intensity:

    • Challenge: Weak CTSZ detection despite confirmed expression

    • Solutions:

      • Optimize protein extraction from lysosomal compartments using specialized lysis buffers

      • Increase antibody concentration (using titration to determine optimal concentration)

      • Extend primary antibody incubation time (overnight at 4°C)

      • Use signal amplification systems (e.g., biotin-streptavidin)

      • Try alternative detection methods (chemiluminescence vs. fluorescence)

      • Ensure samples are not overloaded, which can inhibit proper separation

  • High background in immunohistochemistry:

    • Challenge: Non-specific staining obscuring specific CTSZ signals

    • Solutions:

      • Optimize blocking conditions (test different blocking agents: BSA, serum, commercial blockers)

      • Include additional blocking steps for endogenous biotin or peroxidase

      • Increase washing duration and frequency between steps

      • Optimize antibody dilution through systematic titration

      • Use antigen retrieval methods appropriate for lysosomal proteins

  • Low purity in immunoprecipitation experiments:

    • Challenge: Co-precipitation of non-specific proteins with CTSZ

    • Solutions:

      • Use more stringent washing buffers with increased salt concentration

      • Consider crosslinking antibody to beads to prevent antibody co-elution

      • Perform pre-clearing of lysates before immunoprecipitation

      • Use tandem purification approaches for higher specificity

  • Inconsistent results across different tissue types:

    • Challenge: Variable CTSZ detection in different tissues

    • Solutions:

      • Optimize fixation and processing protocols for each tissue type

      • Adjust antigen retrieval conditions based on tissue characteristics

      • Consider tissue-specific blocking agents to reduce background

      • Validate with multiple CTSZ antibodies targeting different epitopes

By implementing these methodological approaches, researchers can significantly improve the specificity, sensitivity, and reproducibility of CTSZ detection across various experimental platforms.

What are the emerging research directions for CTSZ antibody applications in biomedical research?

The utilization of CTSZ antibodies in biomedical research continues to evolve, with several promising future directions emerging from recent findings:

  • Precision medicine applications:

    • Development of companion diagnostics using CTSZ antibodies for stratifying patients with conditions linked to CTSZ variants

    • Implementation of CTSZ expression profiling in personalized treatment approaches for PBC, where CTSZ SNP status has been identified as an independent risk factor for disease progression

    • Integration of CTSZ antibody-based assays into multi-parameter prognostic algorithms for neurodegenerative and autoimmune conditions

  • Advanced imaging technologies:

    • Application of super-resolution microscopy with CTSZ antibodies to visualize subcellular localization with unprecedented detail

    • Development of intravital imaging approaches to track CTSZ dynamics in living tissues

    • Implementation of multiplexed imaging systems allowing simultaneous detection of CTSZ alongside dozens of other proteins in tissue sections

  • Single-cell analysis integration:

    • Combining CTSZ antibody-based detection with single-cell transcriptomics to correlate protein expression with gene expression patterns

    • Development of single-cell Western blotting techniques for CTSZ quantification in rare cell populations

    • Integration with spatial transcriptomics to map CTSZ protein expression in tissue architectural contexts

  • Therapeutic monitoring applications:

    • Utilization of CTSZ antibodies to evaluate the efficacy of CTSZ-targeting therapeutics

    • Development of circulating CTSZ detection methods as liquid biopsy approaches

    • Monitoring changes in CTSZ expression as pharmacodynamic markers in clinical trials

  • Systems biology approaches:

    • Integration of CTSZ antibody-based proteomics with other omics data for comprehensive understanding of CTSZ in biological networks

    • Development of computational models predicting CTSZ behavior based on antibody-derived quantitative data

    • Implementation of machine learning algorithms to identify patterns in CTSZ expression across disease states

Product Science Overview

Introduction

Cathepsin-Z, also known as Cathepsin-X or Cathepsin-P, is a unique lysosomal cysteine protease. It is primarily expressed by antigen-presenting cells and has been associated with various physiological and pathological processes, including neuroinflammation . The antibody “Mouse Anti Human Cathepsin-Z” is specifically designed to detect human Cathepsin-Z in various applications, such as Western blotting, immunohistochemistry, and immunoprecipitation .

Structure and Function

Cathepsin-Z is a member of the papain-like cysteine protease family. It is synthesized as an inactive proenzyme and activated in the acidic environment of lysosomes. The enzyme plays a crucial role in protein degradation and processing within the lysosome. Unlike other cathepsins, Cathepsin-Z has a unique C-terminal extension that may contribute to its specific functions .

Expression and Localization

Cathepsin-Z is predominantly expressed in antigen-presenting cells, such as macrophages and dendritic cells. It is also found in various tissues, including the liver, spleen, and lungs. The enzyme is localized in the lysosomes, where it participates in the degradation of intracellular and extracellular proteins .

Role in Disease

Cathepsin-Z has been implicated in several diseases, particularly those involving inflammation and immune responses. For instance, its expression is upregulated in neuroinflammatory disorders, suggesting a role in mediating neuroinflammation . Additionally, Cathepsin-Z has been associated with cancer progression, where it may contribute to tumor invasion and metastasis.

Applications of Mouse Anti Human Cathepsin-Z Antibody

The Mouse Anti Human Cathepsin-Z antibody is widely used in research to study the expression and function of Cathepsin-Z. Some of the key applications include:

  • Western Blotting: Detects Cathepsin-Z in cell lysates and tissue extracts .
  • Immunohistochemistry: Localizes Cathepsin-Z in tissue sections, providing insights into its distribution and role in various tissues .
  • Immunoprecipitation: Isolates Cathepsin-Z from complex protein mixtures, facilitating further analysis of its interactions and functions .

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.