CSTA Human, Active

Cystatin-A Human Recombinant, Active
Shipped with Ice Packs
In Stock

Description

Cystatin A Human Recombinant produced in E.Coli is a single, non-glycosylated, Polypeptide chain containing 118 amino acids (1-98a.a.) and having a molecular mass of 13.1 kDa.
The Cystatin A is fused to a 20 amino acid His tag at N-terminus and purified by proprietary chromatographic techniques.

Product Specs

Introduction
Human Cystatin A (CSTA or Stefin A), a member of the cystatin superfamily 1, lacks disulfide bonds and carbohydrates. It functions as an intracellular inhibitor, regulating the activity of cysteine proteases within the papain family, including Cathepsins B, H, and L. CSTA has been linked to various disease states. Due to its role in the altered proteolytic state observed during cancer progression, CSTA may be involved in proteolytic pathways.
Description
Recombinant Human Cystatin A, produced in E. coli, is a single, non-glycosylated polypeptide chain composed of 118 amino acids (1-98a.a.) with a molecular weight of 13.1 kDa. It features a 20 amino acid His tag at the N-terminus and undergoes purification using proprietary chromatographic methods.
Physical Appearance
A clear solution that has been sterilized by filtration.
Formulation
Cystatin-A is supplied at a concentration of 1mg/ml in a buffer consisting of 20mM Tris-HCl (pH 8.0), 1mM DTT, and 10% glycerol.
Stability
For short-term storage (2-4 weeks), the product should be kept at 4°C. For extended storage, it is recommended to freeze the product at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is advised for long-term storage. Repeated freezing and thawing should be avoided.
Purity
The purity is determined to be greater than 95% using reducing SDS-PAGE analysis.
Biological Activity
The IC50 value is less than 1.0 nM. The inhibitory effect of CSTA on the protease activity of papain was assessed using a fluorometric assay with Z-FR-AMC as the substrate. The assay was conducted at a pH of 7.5 and a temperature of 25°C.
Synonyms
Cystatin-A, Cystatin-AS, Stefin-A, CSTA, STF1, STFA.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MIPGGLSEAK PATPEIQEIV DKVKPQLEEK TNETYGKLEA VQYKTQVVAG TNYYIKVRAG DNKYMHLKVF KSLPGQNEDL VLTGYQVDKN KDDELTGF.

Q&A

What is Cystatin A (CSTA) and what are its primary biological functions?

Cystatin A, also known as Stefin A, is an intracellular thiol proteinase inhibitor belonging to the type 1 cystatins (stefins) within the cystatin superfamily. It functions primarily by forming tight complexes with papain and cathepsins B, H, and L to regulate protease activity . CSTA plays a critical role in desmosome-mediated cell-cell adhesion, particularly in the lower levels of the epidermis, making it essential for epidermal development and maintenance . The protein has been identified as one of the precursor proteins of the cornified cell envelope in keratinocytes . As a protease inhibitor, CSTA helps maintain tissue homeostasis by preventing excessive proteolytic activity that could damage cellular structures and disrupt normal physiological processes .

What alternative nomenclature and identifiers are associated with CSTA?

CSTA is known by several synonyms in scientific literature and databases:

  • Cystatin-A/Cystatin-AS

  • Stefin-A/STF1/STFA

  • AREI

  • CystatinAS

  • OTTHUMP00000215569
    In protein databases, CSTA is identified by UniProt accession number P01040 . This standardized identification is essential for consistent retrieval of molecular information across different research platforms and databases.

What is the molecular relationship between CSTA and other proteins in its interaction network?

CSTA exhibits strong functional connections with several cathepsin proteins in its interaction network. According to STRING database analysis, CSTA has particularly high interaction scores with:

  • Cathepsin B (CTSB): 0.998 interaction score

  • Cathepsin L1 (CTSL): 0.992 interaction score

  • Cathepsin H (CTSH): 0.964 interaction score

  • Cathepsin L2/V (CTSV): 0.960 interaction score

  • Cathepsin S (CTSS): 0.921 interaction score
    These high confidence scores indicate that CSTA's primary biological role involves the regulation of these cathepsin proteases. Additionally, CSTA shows a significant interaction with SLC12A8 (Solute carrier family 12 member 8) with a score of 0.958, suggesting potential involvement in keratinocyte proliferation regulation .

What are the recommended methods for detecting and quantifying CSTA in human samples?

For accurate detection and quantification of CSTA in human samples, enzyme-linked immunosorbent assay (ELISA) is the gold standard methodology. Specialized Human Cystatin A ELISA kits offer high sensitivity (as low as 0.11 ng/mL) and specificity for analyzing CSTA in various sample types including:

  • Serum and plasma

  • Tissue homogenates

  • Cell culture supernatants

  • Other biological fluids
    These sandwich ELISA assays typically provide a detection range of 0.47-30 ng/mL with good reproducibility (intra-assay CV: 5.3%, inter-assay CV: 9.9%) . For research requiring visualization of CSTA in tissues or cells, immunohistochemistry and immunofluorescence using specific anti-CSTA antibodies are recommended approaches. Western blotting can be employed for semi-quantitative analysis of CSTA expression in cell or tissue lysates.

How should researchers design experiments to study CSTA's inhibitory activity against proteases?

When investigating CSTA's inhibitory activity against proteases, researchers should consider the following experimental design principles:

  • Enzyme kinetics approach: Design experiments to measure the inhibition constants (Ki) of CSTA against target proteases (cathepsins B, H, L) using fluorogenic or chromogenic substrates.

  • Protein-protein interaction assays: Employ co-immunoprecipitation, surface plasmon resonance, or isothermal titration calorimetry to characterize the binding dynamics between CSTA and target proteases.

  • Functional cellular assays: Develop cell-based assays where CSTA expression is manipulated (overexpression, knockdown, or knockout) to observe changes in protease activity and downstream cellular processes.

  • Controls: Include appropriate controls such as:

    • Positive controls with known inhibitors of target proteases

    • Negative controls with inactive CSTA mutants

    • Dose-response experiments to establish concentration-dependent effects

  • Physiological relevance: Conduct experiments under conditions that mimic the physiological environment where CSTA naturally functions, particularly considering pH and redox conditions relevant to cytoplasmic and epidermal environments .

What approaches can address data inconsistencies in CSTA expression analysis?

Researchers studying CSTA expression may encounter data inconsistencies due to various factors. To address these challenges, consider implementing the following approaches:

  • Standardized notation for contradiction patterns: Adopt a structured notation system like the (α, β, θ) parameters proposed for biomedical data quality assessment, where:

    • α represents the number of interdependent items

    • β represents the number of contradictory dependencies defined by domain experts

    • θ represents the minimal number of required Boolean rules to assess contradictions

  • Boolean minimization techniques: Apply Boolean logic to minimize the number of rules needed to identify contradictions in complex datasets, which can significantly reduce the complexity of data quality assessment frameworks .

  • Cross-validation with multiple detection methods: Compare results from different methodological approaches (ELISA, western blot, qPCR, mass spectrometry) to identify and resolve inconsistencies.

  • Metadata documentation: Thoroughly document experimental conditions, sample processing methods, and analytical procedures to track potential sources of variation.

  • Reference standards: Include validated reference standards and calibrators across experiments to normalize data and enable more accurate cross-study comparisons .

How is CSTA being investigated as a potential biomarker in disease research?

CSTA has been proposed as a prognostic and diagnostic tool, particularly in cancer research . The protein's role in epithelial cell adhesion and protease regulation makes it a candidate biomarker for various pathological conditions. Current research directions include:

  • Cancer biomarker development: CSTA expression patterns are being investigated in various cancer types to determine their correlation with disease progression, metastatic potential, and treatment response .

  • Inflammatory disorders: Given CSTA's role in protease regulation, alterations in its expression or activity may serve as biomarkers for inflammatory conditions characterized by dysregulated proteolytic activity.

  • Dermatological conditions: Since CSTA plays a crucial role in epidermal development and maintenance, its levels are being studied as potential biomarkers for skin disorders involving disrupted epithelial integrity .

  • Methodology evolution: The development of highly sensitive assays like the Human CSTA ELISA Kit, with detection sensitivity down to 0.11 ng/mL, has facilitated more precise quantification of CSTA in clinical samples, enhancing its potential utility as a biomarker .

What are the current understanding and research gaps regarding CSTA's role in desmosome-mediated cell adhesion?

CSTA has been identified as having "an important role in desmosome-mediated cell-cell adhesion in the lower levels of the epidermis" . Current research understanding includes:

  • Structural contribution: CSTA contributes to the stability of desmosomes, which are specialized cell junctions that provide strong adhesion between epithelial cells, particularly in tissues subjected to mechanical stress.

  • Regulatory function: Beyond its protease inhibitory activity, CSTA appears to have a regulatory role in desmosome assembly or maintenance, potentially by protecting desmosomal components from proteolytic degradation.
    Significant research gaps remain, including:

  • Molecular mechanisms: The precise molecular mechanisms by which CSTA influences desmosome structure and function require further elucidation.

  • Interaction partners: While CSTA's interactions with cathepsins are well-characterized, its potential interactions with desmosomal proteins or regulatory factors involved in desmosome assembly are less understood.

  • Tissue-specific variations: How CSTA's role in desmosome-mediated adhesion varies across different epithelial tissues beyond the epidermis remains to be fully investigated.

  • Pathological implications: The consequences of CSTA dysfunction on desmosomal integrity in disease states warrant further research, particularly in conditions characterized by compromised epithelial adhesion.

What experimental models are most appropriate for studying CSTA function?

Researchers investigating CSTA function should consider the following experimental models, each with specific advantages for addressing different research questions:

  • In vitro cellular models:

    • Keratinocyte cell lines (HaCaT, NHEK) for studying epidermal functions

    • 3D organotypic skin models that recapitulate the stratified structure of epidermis

    • Co-culture systems to examine cell-cell adhesion dynamics

  • Recombinant protein systems:

    • Purified recombinant CSTA protein (available with various tags such as His-tag) for biochemical and structural studies

    • Reconstituted systems with purified cathepsins for protease inhibition studies

  • Genetic manipulation models:

    • CRISPR/Cas9-mediated knockout or knockin cell lines

    • siRNA or shRNA for transient knockdown of CSTA expression

    • Overexpression systems using lentiviral or plasmid vectors

  • Animal models:

    • Transgenic mouse models with CSTA modifications

    • Skin-specific conditional knockout models to avoid developmental effects

    • Xenograft models for studying CSTA's role in cancer progression

  • Patient-derived samples:

    • Primary cells isolated from patient biopsies

    • Patient tissue samples for ex vivo analysis

    • Patient-derived organoids for functional studies
      The choice of model should be guided by the specific research question, with consideration given to the physiological relevance and technical limitations of each approach.

What are the critical quality control parameters for recombinant CSTA protein studies?

When working with recombinant human CSTA protein, researchers should implement the following quality control parameters to ensure reliable experimental outcomes:

  • Purity assessment:

    • SDS-PAGE analysis with Coomassie staining (>95% purity recommended)

    • Mass spectrometry verification of intact protein mass

    • Absence of endotoxin contamination (<1 EU/μg protein)

  • Activity verification:

    • Functional assays measuring inhibitory activity against cathepsins B, H, and L

    • Determination of the inhibition constant (Ki) compared to reference standards

    • Dose-dependent inhibition curves to confirm specific activity

  • Structural integrity:

    • Circular dichroism spectroscopy to confirm proper secondary structure

    • Size exclusion chromatography to verify monomeric state and absence of aggregates

    • Thermal stability analysis to ensure protein stability at experimental temperatures

  • Tag interference assessment:

    • For tagged versions (His, GST, etc.), verification that the tag does not interfere with activity

    • Comparison with tag-cleaved versions when possible

    • Controls with tag-only proteins to identify tag-specific effects

  • Storage stability:

    • Activity retention after freeze-thaw cycles

    • Long-term stability monitoring at recommended storage conditions

    • Avoidance of repeated freeze-thaw cycles that may compromise protein activity

How can researchers effectively design experiments to investigate CSTA's role in cancer?

To effectively investigate CSTA's role in cancer research, scientists should design experiments that address multiple aspects of cancer biology while maintaining methodological rigor:

  • Expression analysis in clinical samples:

    • Compare CSTA expression levels between tumor and adjacent normal tissues

    • Correlate expression with clinical parameters (stage, grade, survival)

    • Use multiple detection methods (IHC, qPCR, ELISA) for robust validation

  • Functional studies in cancer cell lines:

    • Manipulate CSTA expression (overexpression/knockdown) in relevant cancer cell models

    • Assess effects on hallmark cancer behaviors (proliferation, migration, invasion)

    • Investigate changes in cathepsin activity and downstream proteolytic events

  • Mechanistic investigations:

    • Identify signaling pathways affected by CSTA modulation

    • Characterize protein-protein interactions in cancer cell contexts

    • Explore potential non-canonical functions beyond protease inhibition

  • In vivo models:

    • Develop xenograft models with CSTA-modified cancer cells

    • Use orthotopic implantation when possible for physiological relevance

    • Consider genetically engineered mouse models for tissue-specific studies

  • Translational approaches:

    • Evaluate CSTA as a biomarker for diagnosis, prognosis, or treatment response

    • Investigate the potential of targeting CSTA-protease interactions therapeutically

    • Develop companion diagnostics for stratifying patients based on CSTA status
      Researchers should incorporate appropriate controls, statistical power calculations, and blinded analyses when possible to enhance the robustness of findings .

What strategies can optimize the detection of low-abundance CSTA in complex biological samples?

Detecting low-abundance CSTA in complex biological samples presents technical challenges that can be addressed through several optimization strategies:

  • Sample enrichment techniques:

    • Immunoprecipitation using anti-CSTA antibodies prior to analysis

    • Fractionation of samples to reduce complexity and concentrate CSTA

    • Affinity purification using immobilized cathepsins as bait proteins

  • Enhanced detection systems:

    • Ultrasensitive ELISA platforms with amplification steps (detection limit 0.11 ng/mL)

    • Proximity ligation assays for improved sensitivity and specificity

    • Mass spectrometry with multiple reaction monitoring (MRM) for targeted detection

  • Signal amplification methods:

    • Tyramide signal amplification for immunohistochemistry/immunofluorescence

    • Rolling circle amplification for in situ protein detection

    • Poly-HRP conjugated detection antibodies in immunoassays

  • Reduction of background interference:

    • Optimization of blocking conditions to minimize non-specific binding

    • Sample pre-treatment to remove common interfering compounds

    • Use of specialized buffers to maintain CSTA stability while reducing background

  • Computational approaches:

    • Application of advanced data analysis algorithms for signal extraction from noise

    • Machine learning methods for pattern recognition in complex datasets

    • Implementation of standardized contradiction detection frameworks for identifying data inconsistencies
      By combining these approaches based on the specific sample type and research question, researchers can substantially improve the detection of low-abundance CSTA even in challenging biological matrices.

What are promising areas for future CSTA research beyond established applications?

Several emerging areas present opportunities for expanding CSTA research beyond its established roles:

  • CSTA in immune regulation:

    • Investigation of CSTA's potential role in modulating immune cell function

    • Exploration of connections between CSTA and inflammatory pathways

    • Study of CSTA's impact on antigen processing and presentation via cathepsin regulation

  • Therapeutic targeting strategies:

    • Development of CSTA mimetics as potential therapeutics

    • Design of small molecule modulators of CSTA-cathepsin interactions

    • Exploration of CSTA as a delivery vehicle for targeting cancer cells

  • Cross-talk with other protease inhibitor systems:

    • Investigation of functional interplay between CSTA and other protease inhibitor families

    • Systems biology approaches to model protease-inhibitor networks

    • Identification of compensatory mechanisms in CSTA-deficient conditions

  • Role in cellular stress responses:

    • Examination of CSTA regulation under various cellular stresses

    • Investigation of potential protective functions during oxidative stress

    • Study of CSTA in proteostasis and protein quality control mechanisms

  • Development of advanced research tools:

    • Creation of biosensors for real-time monitoring of CSTA activity in living cells

    • Generation of conditional knockout models with tissue-specific and temporal control

    • Application of proteomics approaches to identify novel CSTA substrates and interactors
      These directions expand our understanding of CSTA biology while potentially opening new therapeutic avenues and research applications.

How can contradictions in CSTA research data be systematically addressed?

The systematic addressing of contradictions in CSTA research data can be approached through:

  • Implementation of structured contradiction analysis:

    • Apply the (α, β, θ) parameter system to classify contradiction patterns

    • Utilize Boolean minimization techniques to reduce complex interdependencies to manageable rule sets

    • Develop domain-specific contradiction libraries for CSTA research

  • Cross-domain validation frameworks:

    • Establish standardized protocols for data quality assessment across different experimental platforms

    • Create shared repositories of verified contradictions and their resolutions

    • Implement automated contradiction detection in data analysis pipelines

  • Metadata enrichment and standardization:

    • Enhance experimental reporting with detailed metadata about methods, reagents, and conditions

    • Adopt standardized ontologies for describing CSTA-related experiments

    • Incorporate contradiction pattern information in published datasets

  • Collaborative validation initiatives:

    • Establish multi-laboratory validation studies for controversial findings

    • Develop community standards for CSTA research methodologies

    • Create frameworks for resolution of apparently contradictory results By systematically addressing contradictions through these approaches, researchers can improve data quality and accelerate progress in understanding CSTA biology and its applications.

Product Science Overview

Introduction

Cystatin-A, also known as stefin A, is a member of the cystatin superfamily, which encompasses proteins containing multiple cystatin-like sequences. These proteins are primarily known for their role as cysteine protease inhibitors. Cystatin-A is particularly significant due to its function in inhibiting cysteine proteases such as papain and the cathepsins B, H, and L .

Structure and Expression

Recombinant human Cystatin-A is typically produced using an E. coli expression system. The protein is expressed with a sequence (Met1-Phe98) of human Cystatin-A, often fused with a 6×His tag at the C-terminus to facilitate purification . The molecular mass of Cystatin-A is approximately 12 kDa .

Function and Activity

Cystatin-A functions as a cysteine protease inhibitor, forming tight complexes with target proteases. This inhibitory activity is crucial for regulating the proteolytic activity within cells. The protein’s ability to inhibit papain cleavage of a fluorogenic peptide substrate is often used to measure its bioactivity, with an IC50 value of less than 0.7 nM .

Biological Role

Cystatin-A plays a vital role in epidermal development and maintenance. It is one of the precursor proteins of the cornified cell envelope in keratinocytes, contributing to the formation of the skin barrier . Additionally, stefins, including Cystatin-A, have been proposed as prognostic and diagnostic tools for cancer due to their regulatory functions in protease activity .

Applications

Recombinant human Cystatin-A is used extensively in research to study its inhibitory effects on cysteine proteases. It is also utilized in various assays to understand its role in cellular processes and its potential implications in diseases such as cancer .

Storage and Handling

For long-term storage, the lyophilized protein should be kept at -20°C to -80°C. After reconstitution, the protein solution is stable at -20°C for up to three months and at 2-8°C for up to one week. It is recommended to add a carrier protein or stabilizer to the reconstituted solution to minimize freeze-thaw cycles .

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.