CST7 Human

Cystatin 7 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction and Basic Characteristics

CST7 Human (Cystatin F) is a glycosylated cysteine protease inhibitor encoded by the CST7 gene located on human chromosome 20 . It belongs to the type 2 cystatin family, which regulates protease activity in immune and inflammatory pathways.

Key Molecular Features

PropertyValueSource
Molecular Mass15.3 kDa
Amino Acid Sequence132 residues (20–145) with a His-tag at the C-terminus
Purity>95% (SDS-PAGE)
Expression SystemRecombinant E. coli with proprietary chromatographic purification

The protein’s amino acid sequence includes motifs characteristic of cystatins, such as conserved cysteine residues and a C-terminal His-tag for affinity chromatography .

Biological Function and Pathways

CST7 inhibits cysteine proteases like cathepsins L and C, modulating immune responses and cellular homeostasis . Its expression is linked to:

  • Immune regulation: Suppresses protease activity in hematopoietic cells .

  • Neurodegeneration: Upregulated in microglia during amyloid β (Aβ)-driven Alzheimer’s disease (AD) .

  • Inflammation: Neutrophil-specific upregulation in sepsis, viral infections, and sterile inflammation .

Tissue Expression

TissueCST7 ExpressionSource
Blood (Neutrophils)High upregulation during acute inflammation
MicrogliaRobustly induced in AD models, localized to amyloid plaques
Cancer Cell LinesObserved in malignant tumors

Neurodegenerative Diseases

In App NL-G-F mouse models of AD:

SexCST7 Deletion EffectAβ BurdenLysosomal Activity
Female↑ Endolysosomal genes, ↑ LAMP2 staining, ↑ Microglial Aβ uptakeIncreasedEnhanced
Male↓ Inflammatory genes (Il1b, Tnf), ↓ LAMP2 stainingUnchangedReduced

CST7 knockout in females exacerbates Aβ burden via increased phagocytosis, while males show reduced inflammation .

Inflammatory and Infectious Diseases

CST7 is upregulated in neutrophils during:

  • Bacterial infections: Tuberculosis (effect size = 0.84)

  • Viral infections: HIV (1.54), IAV (1.47), RSV (1.18)

  • Sterile inflammation: Systemic juvenile idiopathic arthritis (0.6)

This upregulation is neutrophil-specific and regulated by host factors .

Cancer

CST7 is identified as a survival biomarker in pancreatic ductal adenocarcinoma (PDAC), with higher expression correlating with poorer prognosis .

Neuroprotective vs. Pathogenic Roles

CST7’s dual role in AD highlights a sex-dependent therapeutic challenge:

  • Females: Targeting CST7 may reduce Aβ phagocytosis but risk lysosomal dysfunction .

  • Males: CST7 inhibition could mitigate inflammation without affecting Aβ clearance .

Biomarker Potential

CST7’s expression in PDAC and inflammatory diseases positions it as a candidate for diagnostic or prognostic markers .

Table 1: CST7 Expression Across Diseases

DiseaseCST7 UpregulationCell TypeMechanismSource
Alzheimer’s DiseaseHighMicrogliaEndolysosomal regulation, Aβ phagocytosis
SepsisHighNeutrophilsInflammatory response
Pancreatic CancerHighTumor cellsPoor survival prognosis

Product Specs

Introduction
Cystatin 7 (CST7) is a glycosylated cysteine protease inhibitor believed to play a role in immune regulation by inhibiting a specific target within the hematopoietic system. CST7 possesses multiple cystatin-like sequences and belongs to the cystatin superfamily, which consists of three inhibitory families: type 1 cystatins (stefins), type 2 cystatins, and kininogens. While some members actively inhibit cysteine proteases, others lack or may have never possessed this inhibitory function. Type 2 cystatin proteins, a class of cysteine proteinase inhibitors, are present in various human fluids and secretions. CST7 protein expression has been detected in numerous human cancer cell lines derived from malignant tumors.
Description
Recombinant human CST7, produced in E. coli, is a single polypeptide chain comprising 132 amino acids (residues 20-145) with a molecular weight of 15.3 kDa. A 7-amino acid His-tag is fused to the C-terminus of CST7. The protein is purified using proprietary chromatographic techniques.
Physical Appearance
A clear, sterile-filtered solution.
Formulation
The CST7 solution (0.5 mg/mL) is prepared in phosphate-buffered saline (pH 7.4).
Stability
For short-term storage (up to 2-4 weeks), keep at 4°C. For extended storage, freeze at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
Purity exceeds 95.0% as determined by SDS-PAGE analysis.
Synonyms
Cystatin-F, Cystatin-7, Cystatin-like metastasis-associated protein, CMAP, Leukocystatin, Cystatin 7, CST7, CMAP.
Source
Escherichia Coli.
Amino Acid Sequence
GPSPDTCSQD LNSRVKPGFP KTIKTNDPGV LQAARYSVEK FNNCTNDMFL FKESRITRAL VQIVKGLKYM LEVEIGRTTC KKNQHLRLDD CDFQTNHTLK QTLSCYSEVW VVPWLQHFEV PVLRCHHHHH HH.

Q&A

What is CST7 and what is its basic function in human cells?

CST7 encodes cystatin F (CF), an atypical cystatin protein that is localized to the endolysosomal compartment where it acts as an endogenous inhibitor of cysteine proteases such as cathepsin L and cathepsin C . Unlike other cystatins that are secreted or found in the cytosol, cystatin F plays a regulatory role within the endolysosomal system, potentially influencing protein degradation and processing pathways critical for cellular homeostasis. In microglia, CST7 appears to regulate lysosomal function, which has implications for key microglial activities including phagocytosis and degradation of pathological proteins.

Where is CST7 primarily expressed in the human brain?

CST7 shows highly cell-specific expression in the brain. Based on single-cell RNA sequencing data, CST7 is expressed almost exclusively in disease-associated microglial clusters with minimal expression in homeostatic microglia and negligible expression in other CNS cell types including neurons, astrocytes, and oligodendrocytes . This restricted expression pattern makes CST7 a valuable marker for identifying disease-associated microglial states, particularly in the context of neurodegenerative conditions like Alzheimer's disease.

What methodological approaches are used to study CST7 expression?

Multiple complementary approaches are employed to investigate CST7 expression:

TechniqueApplicationAdvantages
Fluorescence-Activated Cell Sorting (FACS)Isolation of microglia for expression analysisAllows purification of specific cell populations
Immunomagnetic SeparationIsolation of microglia from brain tissueLess potential for activation artifacts
RiboTag Translational ProfilingAnalysis of actively translated mRNAsFocuses on proteins being actively synthesized
Single-cell RNA SequencingCell-type specific expression profilingProvides high-resolution data on expression heterogeneity
In Situ HybridizationVisualization of mRNA in intact tissuePreserves spatial information

How does CST7 expression differ in male versus female models of Alzheimer's disease?

Research demonstrates that CST7 expression exhibits sexual dimorphism in Alzheimer's disease models. In both APP/PS1 and App NL-G-F mouse models, CST7 appears marginally more highly expressed in female disease brains compared to males . This pattern is consistent with broader findings suggesting that the disease-associated microglial (DAM/ARM/MGnD) state develops more rapidly in female mice. Additionally, other endolysosomal genes that are characteristic of disease-associated microglia, such as Spp1 and Gpnmb, also show higher expression in female microglia compared to males, suggesting a broader sex-dependent pattern in microglial responses to disease.

What are the functional consequences of CST7 deletion in Alzheimer's disease models?

The functional consequences of CST7 deletion demonstrate striking sex-dependent differences:

ParameterFemale App NL-G-F CST7-/-Male App NL-G-F CST7-/-
Transcriptomic EffectsUpregulation of endolysosomal genesDownregulation of inflammatory genes
Differentially Expressed Genes26 genes upregulated, 5 downregulated8 genes upregulated, 51 downregulated
Key Affected PathwaysEndolysosomal functionInflammatory response, NF-κB signaling
Lysosomal BurdenIncreasedReduced
Microglial Aβ BurdenIncreasedNo significant change
Phagocytic CapacityEnhancedNot significantly affected

These divergent effects indicate that CST7 plays fundamentally different roles in male versus female microglia in the context of Alzheimer's disease pathology .

How does CST7 influence microglial phagocytosis of amyloid beta?

In female microglia derived from App NL-G-F mice, CST7 deletion leads to enhanced phagocytosis of amyloid beta in vitro . This suggests that CST7 normally functions as a negative regulator of microglial phagocytosis, potentially by inhibiting lysosomal proteases involved in the degradation of engulfed material. Importantly, the increased microglial amyloid beta burden observed in female CST7-deficient mice appears to be due to increased uptake rather than impaired degradation, as demonstrated by in vitro phagocytosis assays. This finding has significant implications for understanding microglial clearance of pathological proteins in neurodegenerative diseases.

What is the relationship between CST7 and inflammation in neurodegenerative disease?

The relationship between CST7 and inflammation shows pronounced sexual dimorphism. In male App NL-G-F mice, CST7 deletion leads to downregulation of classical proinflammatory mediators such as Il1b, Il1a, Tnf, and Cxcl2, suggesting that CST7 normally promotes inflammatory responses in males . Gene ontology analyses of CST7-regulated genes in males revealed enrichment for inflammatory response and NF-κB signaling pathways. In contrast, CST7 deletion in females primarily affects endolysosomal rather than inflammatory pathways. This dichotomy suggests that CST7 plays different roles in orchestrating microglial responses in males versus females, with implications for sex differences in neuroinflammation.

How do sex-specific effects of CST7 relate to sexual dimorphism in Alzheimer's disease?

Alzheimer's disease exhibits significant sexual dimorphism, with women up to twice as likely to be diagnosed than men . The sex-dependent effects of CST7 on microglial function may contribute to this dimorphism through several mechanisms:

  • Female microglia show accelerated adoption of the disease-associated (DAM/ARM/MGnD) phenotype in which CST7 is a key component

  • CST7 appears to have more pronounced effects on endolysosomal function in females

  • The inhibitory effect of CST7 on phagocytosis may particularly impact female microglia

These findings suggest that sex-specific differences in microglial function, partly regulated by genes like CST7, may contribute to the increased vulnerability of females to Alzheimer's disease. Understanding these mechanisms may provide insights into developing sex-specific therapeutic approaches.

What experimental approaches can be used to investigate CST7 function in human models?

Translating findings from mouse models to human disease contexts requires specialized approaches:

ApproachDescriptionConsiderations
Human iPSC-derived MicrogliaMicroglia differentiated from human induced pluripotent stem cellsMay require disease-relevant stimuli to induce CST7 expression
TREM2 OverexpressionDriving CST7 expression through TREM2 overexpressionRecent studies show this approach reveals CST7-dependent inhibition of phagocytosis
Ex vivo Human Tissue AnalysisExamination of CST7 expression in human post-mortem brain samplesProvides direct evidence of expression in human disease
CRISPR-Cas9 ModificationGenetic manipulation of CST7 in human cell modelsAllows precise examination of gene function
Humanized Mouse ModelsMice engrafted with human immune cellsBridges gap between mouse models and human disease

Since CST7 function appears to be revealed primarily in disease contexts, complex models that recapitulate disease environments are necessary for translational studies .

What are the methodological challenges in studying CST7 function in microglia?

Several methodological challenges exist in studying CST7 function:

  • Context-Dependent Expression: CST7 is expressed at very low levels under baseline conditions and becomes dramatically upregulated only in disease contexts, necessitating appropriate disease models.

  • Sex-Dependent Effects: The markedly different consequences of CST7 deletion in males versus females require sex-stratified experimental designs.

  • Cell-Type Specificity: The highly specific expression of CST7 in disease-associated microglia requires techniques for isolating and studying these specific cell populations.

  • Translation to Human Disease: Translating findings from mouse models to human disease contexts requires specialized approaches such as human iPSC-derived microglia and post-mortem tissue analysis.

  • Functional Redundancy: Potential compensatory mechanisms in genetic knockout models may mask certain phenotypes.

These challenges highlight the importance of using multiple complementary approaches and careful experimental design when investigating CST7 function .

How might CST7-related findings inform therapeutic approaches for Alzheimer's disease?

The emerging understanding of CST7 function suggests several potential therapeutic implications:

  • Sex-Specific Interventions: Given the dramatically different effects of CST7 in males versus females, sex-specific therapeutic approaches may be necessary. Inhibiting CST7 might be beneficial in females but potentially counterproductive in males.

  • Endolysosomal Targeting: Since CST7 regulates endolysosomal proteases, approaches that modulate specific aspects of endolysosomal function affected by CST7 might offer more precise intervention.

  • Combination Approaches: Targeting CST7 in combination with other microglial genes involved in Alzheimer's disease, such as TREM2, might yield synergistic effects.

  • Disease Stage-Specific Interventions: The dynamic changes in microglial states across disease progression suggest that the timing of CST7-targeted interventions may be critical for therapeutic success.

  • Biomarker Development: The robust upregulation of CST7 in disease-associated microglia might be leveraged for developing biomarkers of microglial activation in neurodegenerative diseases.

These approaches highlight the potential for translating basic research on CST7 into clinically relevant interventions .

What cellular mechanisms underlie the sex-specific effects of CST7?

While the search results demonstrate clear sex-dependent effects of CST7 deletion, the underlying mechanisms remain incompletely understood. Future research should investigate:

  • Cell-intrinsic versus hormonal influences on CST7 function through ovariectomy/hormone replacement studies

  • Potential interactions between CST7 and sex chromosome genes

  • Sex-specific differences in CST7 protein processing or localization

  • Temporal dynamics of CST7 expression across disease progression in males versus females

Understanding these mechanisms will provide deeper insights into the fundamental biology of sex differences in microglial function and neurodegenerative disease .

How does CST7 interact with other risk factors for Alzheimer's disease?

Future studies should investigate potential interactions between CST7 and other established risk factors for Alzheimer's disease:

  • Genetic risk factors such as APOE and TREM2 variants

  • Age-related changes in microglial function

  • Comorbidities such as diabetes and cardiovascular disease

  • Environmental factors including systemic inflammation

These interactions may further clarify the role of CST7 in Alzheimer's disease pathogenesis and potentially identify synergistic effects that could be targeted therapeutically .

What is the translational potential of CST7-focused research?

Translating findings on CST7 from mouse models to human applications requires addressing several key questions:

  • Is the sex-dependent function of CST7 conserved in human microglia?

  • How does CST7 expression correlate with disease progression in human Alzheimer's disease?

  • Are there functional polymorphisms in the human CST7 gene that modulate disease risk?

  • Can CST7 expression or activity be modulated pharmacologically?

  • Would CST7-targeted interventions need to be personalized based on sex, disease stage, or genetic background?

Addressing these questions will be essential for realizing the therapeutic potential of CST7-focused research .

What techniques can be developed to better study CST7 dynamics in living systems?

Development of new experimental approaches would enhance our understanding of CST7 function:

  • CST7 reporter systems to visualize expression dynamics in real-time

  • Activity-based probes to monitor cathepsin inhibition by CST7 in living cells

  • Inducible and cell-type specific CST7 knockout or overexpression systems

  • High-resolution imaging techniques to visualize CST7 trafficking within the endolysosomal system

  • Computational models to predict the effects of CST7 modulation on microglial function

These technical advances would provide more nuanced insights into CST7 biology and facilitate the development of targeted interventions .

Product Science Overview

Introduction

Cystatin 7, also known as Cystatin F, is a member of the cystatin superfamily, which consists of natural cysteine protease inhibitors. These inhibitors are present in a wide variety of organisms and play crucial roles in regulating protease activity. Cystatin 7 is particularly notable for its involvement in immune regulation and its potential implications in cancer research.

Structure and Expression

Cystatin 7 is a type 2 cystatin, which means it belongs to a class of cysteine proteinase inhibitors found in various human fluids and secretions. The protein is glycosylated and has a molecular weight of approximately 15.2 kDa, although it is observed to be around 25 kDa due to glycosylation . The recombinant form of Cystatin 7 is typically expressed in HEK293 cells, a human embryonic kidney cell line commonly used for protein production .

Biological Function

Cystatin 7 is primarily expressed in hematopoietic cells, with the highest levels observed in monocytes, dendritic cells, and certain types of T-cells . Its expression is up-regulated during the maturation of dendritic cells, suggesting a specific role in immune regulation . Cystatin 7 functions as an inhibitor of cysteine proteases, enzymes that break down proteins by cleaving peptide bonds. By inhibiting these proteases, Cystatin 7 helps regulate various physiological processes, including immune responses and inflammation.

Role in Cancer Research

Cystatin 7 has been observed in various human cancer cell lines, indicating its potential role in cancer progression and metastasis . The protein is produced as a dimer, an inactive form that becomes an active cathepsin inhibitor upon chemical reduction . This activation mechanism suggests that Cystatin 7 could be involved in controlling protease activity in the tumor microenvironment, thereby influencing cancer cell behavior.

Applications in Research

Recombinant Cystatin 7 is widely used in research to study its role in immune regulation and cancer. The protein is typically provided as a lyophilized powder, which can be reconstituted for experimental use . It is essential to handle and store the protein properly to maintain its stability and activity. Researchers often use Cystatin 7 to investigate its interactions with cysteine proteases and to explore its potential as a therapeutic target for immune-related disorders and cancer.

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.