CST3 Rat

Cystatin C Rat Recombinant
Shipped with Ice Packs
In Stock

Description

Total 134 AA, Mw: 14.93 kDa (calculated). N-terminal His-tag (14AA).

Product Specs

Introduction
Cystatins are a large family of proteins that inhibit cysteine proteases. These inhibitors are present in various tissues and bodily fluids of both plants and animals. They are known for their ability to form strong complexes with cysteine proteases, such as cathepsin B, H, L, and S. Among them, Cystatin C stands out due to its significance in biochemistry, medicine, and evolutionary studies. This secreted molecule, with a molecular weight of 13,260 Da, consists of 120 amino acids, lacks carbohydrate structures, and has two disulfide bridges near its carboxyl terminus. Elevated levels of Cystatin C are observed in individuals with malignant diseases and kidney dysfunction, suggesting its potential as a more accurate marker than creatinine for assessing renal function. Conversely, low levels of Cystatin C have been linked to the degradation of elastic laminae, potentially leading to conditions like atherosclerosis and abdominal aortic aneurysm.
Description
This recombinant Cystatin C protein consists of 134 amino acids, with a calculated molecular weight of 14.93 kDa. It includes a 14-amino acid His-tag located at the N-terminus.
Physical Appearance
White powder, sterile-filtered and lyophilized.
Formulation
Lyophilized from a 0.5 mg/ml solution in 0.03M Acetate buffer, pH 4. The solution was sterile-filtered using a 0.4-micron filter before lyophilization.
Solubility
To reconstitute the lyophilized protein, add 0.2 ml of 0.1M Acetate buffer (pH 4) to the vial and allow the pellet to dissolve completely. For use in buffers with a higher pH, we recommend diluting the reconstituted protein with the desired buffer to a final concentration of 0.1 mg/ml. Please note that the solubility of this protein is limited at higher concentrations.
Stability
Store the lyophilized protein at -20°C. After reconstitution, aliquot the protein and store at 4°C to minimize freeze-thaw cycles. Reconstituted protein remains stable at 4°C for up to two weeks without any noticeable degradation.
Purity
Purity is greater than 96% as determined by SDS-PAGE analysis.
Synonyms
Cystatin-C, Cystatin-3, CYSC, MGC105556.
Source
Escherichia Coli.
Amino Acid Sequence
MRGSHHHHHH GMASGTSRPP PRLLGAPQEA DASEEGVQRA LDFAVSEYNK GSNDAYHSRA IQVVRARKQLVAGINYYLDV EMGRTTCTKS QTNLTNCPFH DQPHLMRKAL CSFQIYSVPW KGTHTLTKSS.

Q&A

What is CST3 and what are its fundamental characteristics in rat models?

Cystatin C (CST3) is a secreted extracellular cysteine protease inhibitor belonging to the cystatin superfamily. In rats, it is a protein of approximately 13 kDa that exists in both non-glycosylated and glycosylated forms. Rat Cystatin C shares 72% amino acid sequence identity with human Cystatin C and 88% sequence identity with mouse Cystatin C. It is produced in all tissues and present in all biological fluids. Its primary function is inhibiting cysteine proteases of the papain family, such as Cathepsins B, H, K, L, and S . Cystatin C is freely filtered by the glomeruli and then taken up by proximal tubule epithelial cells via megalin-mediated endocytosis, where it is metabolized rather than returning to the bloodstream . This characteristic makes CST3 valuable as a biomarker in renal function assessment.

How is CST3 regulated in the rat central nervous system following injury?

Following axonal injury in rats, such as facial nerve axotomy, Cystatin C expression is significantly upregulated in the central nervous system. In situ hybridization studies have revealed that Cystatin C mRNA in the facial nucleus markedly increases by day 7 post-axotomy and gradually decreases to normal levels by day 50. This upregulation occurs primarily in microglial cells within the damaged facial nucleus, as confirmed by both light and electron microscopic immunohistochemistry . The temporal expression pattern suggests CST3 plays a significant role in neuronal degeneration, regeneration, and repair processes following injury. Furthermore, immunoreactivity has been found in the extracellular space, indicating that microglia likely secrete this protein into the surrounding environment where it may influence regenerative processes .

What is the biological significance of CST3 expression in rat tissues?

Cystatin C expression in rat tissues serves several critical biological functions. As a potent inhibitor of cysteine proteinases, CST3 helps regulate proteolytic activity that could otherwise cause tissue damage if uncontrolled. Changes in Cystatin C expression have been documented in numerous pathological conditions, including cardiovascular diseases such as atherosclerosis, amyloid angiopathy, and myocardial infarction . In the central nervous system, its upregulation by microglia in response to injury suggests roles in modulating inflammatory responses and tissue remodeling during recovery . Additionally, CST3 may have immune-regulatory functions, with recent evidence indicating sex-dependent effects in experimental autoimmune models, where it demonstrates disease-promoting activity specifically in females .

What are the validated methods for measuring CST3 in rat biological samples?

The most widely utilized method for measuring rat Cystatin C is the enzyme-linked immunosorbent assay (ELISA). Commercially available ELISA kits can quantify Cystatin C in rat serum, plasma, and cell culture medium . These assays employ a sandwich ELISA principle where a target-specific antibody is pre-coated in microplate wells, followed by sample addition and binding to this immobilized capture antibody. Detection involves a second antibody and substrate solution that produces a measurable signal proportional to CST3 concentration . For accurate quantification, researchers should generate a standard curve using computer software capable of four-parameter logistic curve-fitting or plot the mean absorbance against concentration . The sensitivity of typical assays reaches a minimum detectable dose of approximately 3.93 pg/mL, with a range from 2.47-12.9 pg/mL across different kit evaluations .

What technical considerations should researchers address when measuring rat CST3?

When measuring rat CST3, researchers must consider several technical aspects to ensure reliable results. Current assays demonstrate good reliability with intra-assay precision CV% typically around 2.8-3.4% and inter-assay precision CV% between 5.4-9.4% . Sample dilution is often necessary, as high-concentration samples should be serially diluted to produce values within the assay's dynamic range. Linearity assessments show that samples can generally be diluted 1:2 to 1:16 with recovery percentages between 80-110% across various sample types . For cell culture supernatants, tissue lysates, serum, plasma, and urine samples, specific dilution protocols may be required to optimize detection. Additionally, researchers should be aware that different sample matrices may affect results differently, necessitating appropriate controls and validation steps for each sample type .

What are the optimal protocols for sample preparation when measuring CST3 in different rat specimens?

Sample preparation protocols vary depending on the specimen type being analyzed. For serum samples, blood should be collected following standard protocols, allowed to clot, then centrifuged to separate serum. For plasma, appropriate anticoagulants should be used during collection. Both serum and plasma samples typically require dilution before assay, with 1:2 to 1:16 dilutions showing good linearity in validation studies . Cell culture supernatants should be centrifuged to remove debris before analysis, while tissue samples require homogenization in appropriate buffers followed by centrifugation to collect the supernatant. For urine samples, centrifugation to remove particulate matter is recommended before dilution and analysis. Across all sample types, it's crucial to avoid repeated freeze-thaw cycles that may degrade the protein. Recovery testing in cell culture media has shown excellent results, with average recovery rates of 107% (range 98-113%) .

What is the role of CST3 in microglial activation following neuronal injury in rats?

Following neuronal injury in rats, CST3 plays a significant role in microglial activation and subsequent neuroinflammatory processes. In situ hybridization studies have shown that CST3 mRNA is markedly upregulated in microglia within the damaged facial nucleus after axotomy . This upregulation follows a specific temporal pattern, increasing by day 7 post-injury and returning to baseline by day 50, suggesting a time-dependent role in the injury response. Electron microscopic immunohistochemistry has confirmed that microglia in the damaged facial nucleus strongly express CST3 protein, and importantly, the protein is also detected in the extracellular space . This extracellular presence indicates that microglia secrete CST3 into the surrounding environment, where it likely influences the injury microenvironment through inhibition of cysteine proteases. This regulated expression pattern suggests CST3 functions in modulating neuroinflammation, potentially limiting excessive protease activity that could exacerbate tissue damage while participating in processes of neuronal degeneration, regeneration, and repair .

How can researchers effectively visualize and quantify CST3 expression in rat brain tissue?

To effectively visualize and quantify CST3 expression in rat brain tissue, researchers should employ complementary techniques that provide information about both spatial distribution and expression levels. In situ hybridization using RNA probes specific for rat Cystatin C has been successfully used to visualize mRNA expression patterns and identify cells actively transcribing CST3 . This can be complemented with immunohistochemistry using antibodies specific for Cystatin C to detect protein localization. For quantitative assessment, researchers can use Western blotting of tissue homogenates or ELISA of tissue extracts. When designing these experiments, it's important to consider the secreted nature of CST3, as the protein may accumulate in locations distant from its production site. Dual-labeling approaches combining CST3 detection with cell-type-specific markers (e.g., Iba1 for microglia, GFAP for astrocytes, NeuN for neurons) can help identify the specific cell populations expressing or interacting with CST3 in different pathological contexts .

What experimental models are most appropriate for studying CST3 in rat neurodegeneration?

Several experimental models are appropriate for studying CST3 in rat neurodegeneration, each offering distinct advantages. The facial nerve axotomy model has been well-validated for studying microglial responses and CST3 upregulation following peripheral nerve injury . This model allows for precise timing of the injury and clear delineation of the affected region, facilitating time-course studies of CST3 expression. For more central neurodegenerative processes, models of traumatic brain injury, stroke (middle cerebral artery occlusion), or chemically-induced neurodegeneration can be employed. When studying CST3 in these contexts, researchers should consider both acute injury responses and chronic degenerative processes, as CST3 functions may differ between these phases. Additionally, transgenic approaches using Cst3 knockout or overexpressing rats can be particularly valuable for establishing causality in CST3's role in neurodegeneration. When selecting models, researchers should consider the specific pathological mechanisms they wish to study, as CST3's functions may vary across different forms of neurodegeneration .

How does CST3 function differ between male and female rats in experimental disease models?

Research has revealed striking sex-dependent differences in CST3 function in experimental autoimmune encephalomyelitis (EAE) models. Female Cst3 null (Cst3−/−) mice display significantly attenuated clinical signs of disease compared to wild-type littermates, indicating a disease-promoting function for CST3 in females . This difference is associated with reduced interleukin-6 production and lower expression of key proteins involved in antigen presentation in female Cst3−/− animals. In contrast, male wild-type and Cst3−/− mice show no significant differences in EAE clinical signs or antigen-presenting cell function after the initial disease onset, despite a slight delay in disease initiation . Furthermore, female Cst3-overexpressing (Cst3Tg) animals demonstrated enhanced clinical disability at peak disease, while male Cst3Tg animals showed no differences compared to controls . These findings indicate that CST3's role in immune-mediated disease is sex-dependent and likely influenced by gonadal hormones, highlighting the importance of considering sex as a biological variable in CST3 research.

What contradictory findings exist regarding CST3's role in inflammation, and how should researchers interpret them?

Contradictory findings regarding CST3's role in inflammation present a complex picture that requires careful experimental design to unravel. Some studies suggest an immune-limiting role for CST3, where its absence leads to enhanced T and B cell responses and where activation of macrophages with inflammatory stimuli decreases CST3 secretion . Conversely, other findings suggest a pro-inflammatory role, particularly in female animals with EAE, where CST3 deficiency reduces disease severity and CST3 overexpression enhances clinical disability . Additionally, decreased CST3 expression has been linked to reduced MHC II expression and inflammation in IL-6-activated dendritic cells, supporting a pro-inflammatory function in specific contexts . These contradictions suggest that CST3's role in inflammation is context-dependent and may vary based on cell type, disease model, timing, and sex. Researchers should interpret these findings by considering that CST3 likely has multifaceted functions that depend on the specific microenvironment, cell population, and pathological context. Comprehensive studies incorporating both sexes, multiple time points, and various cell types are necessary to fully elucidate CST3's complex roles in inflammation.

What analytical approaches can differentiate cell-specific versus systemic effects of CST3 in rat disease models?

To differentiate cell-specific versus systemic effects of CST3 in rat disease models, researchers should employ multimodal analytical approaches. Cell-type specific analyses can be performed using techniques like flow cytometry with intracellular staining for CST3 combined with surface markers for different immune cell populations. Single-cell RNA sequencing can provide comprehensive insights into which cells express CST3 and which respond to it across different tissues. For in vivo studies, bone marrow chimeras or adoptive transfer experiments can help determine whether disease phenotypes are driven by CST3 from circulating cells versus tissue-resident cells. Conditional knockout models, where CST3 is deleted only in specific cell types, offer another powerful approach to dissect cell-autonomous effects. To assess systemic effects, researchers should measure CST3 levels in multiple compartments (serum, CSF, tissue) and correlate these with disease parameters. Comparing local versus systemic administration of recombinant CST3 or CST3-neutralizing antibodies can also help distinguish between local and systemic effects. Finally, ex vivo functional assays with isolated cells from different tissues can determine whether CST3's effects are direct or indirect .

What are the sensitivity and specificity parameters of current rat CST3 detection methods?

Current rat CST3 detection methods demonstrate well-defined sensitivity and specificity parameters that researchers should consider when selecting appropriate assays. For ELISA-based detection, sensitivity evaluations across 41 assays showed minimum detectable doses (MDD) ranging from 2.47-12.9 pg/mL, with a mean MDD of 3.93 pg/mL . These values were determined by adding two standard deviations to the mean optical density of zero standard replicates. Regarding specificity, commercial assays are designed to exclusively recognize both natural and recombinant rat Cystatin C . Validation studies using natural mouse or rat Cystatin C have demonstrated dose-response curves parallel to standard curves obtained with recombinant proteins, indicating good specificity for the native protein . The linearity of these assays is also well-characterized, with dilution series of high-concentration samples showing average percent recovery generally within 80-110% of expected values across multiple sample types .

How should researchers interpret CST3 concentration data in different rat sample types?

Interpreting CST3 concentration data requires consideration of sample-specific factors that influence baseline levels and variability. For serum and plasma samples, researchers should establish reference ranges specific to their rat strain, age, and sex, as these factors can influence baseline CST3 levels. When interpreting urine CST3 concentrations, values should be normalized to creatinine to account for variations in urine concentration. For cell culture supernatants, consideration should be given to cell number, culture conditions, and time of collection. The table below summarizes key considerations for different sample types:

Sample TypeTypical DilutionLinearity RangeSpecific Considerations
Rat Serum1:2 - 1:1681-99%Sex differences may affect baseline levels
EDTA Plasma1:2 - 1:1695-100%Less susceptible to ex vivo release from cells
Heparin Plasma1:2 - 1:1689-93%May show different values from EDTA plasma
Urine1:2 - 1:1690-98%Should be normalized to creatinine
Cell Culture1:2 - 1:1691-94%Account for cell number and culture time

When comparing CST3 levels across different experimental groups, researchers should ensure consistent sample collection, processing, and assay conditions to minimize technical variability .

What quality control measures ensure reliable CST3 measurements in rat research?

To ensure reliable CST3 measurements in rat research, several quality control measures should be implemented throughout the experimental workflow. During sample collection and processing, standardized protocols should be followed to minimize pre-analytical variability. Samples should be processed promptly and stored at appropriate temperatures (-20°C or lower) with avoidance of repeated freeze-thaw cycles. When performing assays, internal quality controls at known concentrations (low, medium, and high) should be included on each plate to monitor assay performance. Standard curves should be generated using appropriate curve-fitting algorithms, preferably four-parameter logistic regression . Assay validation should include assessments of intra-assay precision (CV typically 2.8-3.4%) and inter-assay precision (CV typically 5.4-9.4%) to ensure reproducibility . Recovery experiments, where known amounts of CST3 are added to samples, help verify accuracy (typical recovery 98-113%) . Dilution linearity testing confirms that samples can be appropriately diluted while maintaining proportional results. Finally, parallel analysis of selected samples using alternative methods (e.g., Western blot) can provide additional verification of ELISA results.

How can genetic manipulation of CST3 in rats advance understanding of its biological functions?

Genetic manipulation of CST3 in rats provides powerful approaches for elucidating its biological functions in both physiological and pathological contexts. Creating Cst3 knockout rats allows for loss-of-function studies to determine the consequences of CST3 deficiency on various physiological processes and disease susceptibility. Conversely, Cst3-overexpressing transgenic rats enable gain-of-function studies to assess the effects of elevated CST3 levels. The striking findings from studies with Cst3 null mice showing significantly attenuated clinical signs of EAE in females but not males highlight the value of genetic approaches in revealing sex-dependent functions that might otherwise remain undiscovered . By comparing the phenotypes of these genetically modified rats with wild-type controls across different disease models, researchers can establish causality rather than merely correlation. Conditional knockout models, where CST3 expression can be selectively eliminated in specific cell types or at specific times, offer even more refined approaches to dissect the cell-specific and temporal aspects of CST3 function. These genetic approaches can be particularly valuable in resolving contradictory findings regarding CST3's roles in inflammation and disease processes .

What experimental designs can elucidate sex-dependent effects of CST3 in rat disease models?

To properly elucidate sex-dependent effects of CST3 in rat disease models, researchers should implement comprehensive experimental designs that account for hormonal influences and sex-specific biological variables. Studies should include both male and female rats in sufficient numbers to allow for sex-stratified statistical analyses. Age-matched animals should be used, with consideration for estrous cycle stage in females, which can be determined through vaginal cytology. Gonadectomy experiments, where male and female rats undergo castration or ovariectomy respectively, can help determine whether sex differences are mediated directly by gonadal hormones . Hormone replacement studies can further clarify the specific hormones involved. For genetic studies using Cst3 knockout or transgenic rats, littermate controls of both sexes should be used to control for genetic background effects. When possible, parallel in vitro studies using primary cells derived from male and female rats can help identify cell-autonomous sex differences in CST3 function. Comprehensive phenotyping should include both clinical disease scores and molecular analyses of immune parameters, particularly focusing on antigen presentation machinery components like MHC II, CD80, CD86, and IL-6 production, which have shown sex-dependent relationships with CST3 .

What are the most promising therapeutic applications based on CST3 biology in rat disease models?

Research on CST3 biology in rat disease models has revealed several promising therapeutic applications, particularly in neurological and immunological disorders. The finding that CST3 deficiency attenuates EAE severity in female animals suggests that CST3 inhibition could potentially benefit female patients with multiple sclerosis or similar autoimmune conditions . Since this effect is associated with reduced interleukin-6 production and decreased expression of antigen presentation machinery, therapeutic strategies targeting these pathways might be particularly effective. The upregulation of CST3 by microglia following axonal injury indicates potential applications in promoting neural repair and regeneration after trauma . This could involve either enhancing or inhibiting CST3 function depending on the specific pathological context and timing. As CST3 is a potent inhibitor of cysteine proteases implicated in various pathological processes, modulating its activity could help control excessive proteolytic activity in conditions like neurodegeneration, cancer, and inflammatory disorders. Development of recombinant CST3 variants, peptide mimetics, or small molecule modulators that can enhance or inhibit specific CST3-protease interactions represents a promising avenue for therapeutic intervention based on rat model findings .

What emerging technologies will advance CST3 research in rat models?

Emerging technologies are poised to significantly advance CST3 research in rat models by providing unprecedented insights into its functions and regulatory mechanisms. CRISPR/Cas9 gene editing now enables the creation of more sophisticated rat models, including point mutations that mimic human CST3 variants associated with disease or domain-specific modifications that alter particular functional aspects of the protein. Single-cell RNA sequencing can reveal cell-specific expression patterns of CST3 and its receptors across different tissues and disease states, providing a comprehensive atlas of CST3 biology. Advanced imaging techniques such as two-photon intravital microscopy allow for real-time visualization of CST3 dynamics in living tissues, while super-resolution microscopy provides nanoscale insights into CST3 localization and interactions. Mass spectrometry imaging can map CST3 distribution across tissue sections with high spatial resolution while simultaneously detecting post-translational modifications. Computational approaches integrating multi-omics data can help identify novel CST3 interaction networks and predict functional relationships. Finally, organ-on-a-chip technologies incorporating multiple cell types can model complex CST3-mediated cell-cell interactions in controlled microenvironments that better recapitulate in vivo conditions .

How can researchers integrate CST3 findings from rat models with human clinical data?

Integrating CST3 findings from rat models with human clinical data requires thoughtful approaches that account for species differences while leveraging translational opportunities. Researchers should first establish the degree of conservation between rat and human CST3 in terms of protein structure (72% amino acid identity), expression patterns, and regulatory mechanisms . Comparative studies examining CST3 function in both rat cells/tissues and human samples (e.g., peripheral blood mononuclear cells, induced pluripotent stem cell-derived models) can identify conserved versus species-specific aspects of CST3 biology. Biobank resources containing human samples with associated clinical data provide opportunities to validate rat model findings in human populations. Researchers can look for associations between CST3 levels or genetic variants and disease parameters that parallel those observed in rat models. The sex-dependent effects of CST3 observed in rat EAE models warrant particular attention in human studies, suggesting that clinical data should be stratified by sex when analyzing CST3 associations . Additionally, pharmacological studies targeting CST3 or its downstream pathways in rats should assess whether similar interventions might be feasible in humans, considering species differences in drug metabolism and distribution.

What methodological improvements would enhance precision in rat CST3 research?

Several methodological improvements could significantly enhance precision in rat CST3 research. Development of more sensitive and specific antibodies that can distinguish between different forms of CST3 (e.g., glycosylated versus non-glycosylated, full-length versus truncated) would allow for more nuanced analysis of CST3 biology . Implementation of absolute quantification methods using isotope-labeled internal standards would improve the accuracy of CST3 measurement across different laboratories and studies. Standardization of sample collection, processing, and storage protocols would reduce pre-analytical variability. For genetic models, improved methods for conditional and inducible gene manipulation would provide better temporal and spatial control of CST3 expression. Development of activity-based probes that can measure functional interactions between CST3 and its target proteases in vivo would move beyond simple expression analysis to functional assessment. Integration of machine learning approaches for image analysis could enhance the quantification of CST3 distribution in tissue sections. Finally, establishment of open-access repositories for rat CST3 data would facilitate meta-analyses and systematic reviews, accelerating knowledge synthesis across multiple studies and reducing publication bias .

Product Science Overview

Classification and Structure

Cystatin C is a secreted protein with a molecular weight of approximately 14 kDa . The recombinant form of rat Cystatin C is typically produced in a mouse myeloma cell line (NS0) and includes a C-terminal 6-His tag for purification purposes . The protein sequence of rat Cystatin C consists of 140 amino acids, starting from Met1 to Ala140 .

Biological Properties and Functions

Cystatin C is ubiquitously expressed in human tissues and body fluids . Its primary function is to inhibit cysteine proteases, which are enzymes that degrade proteins by cleaving peptide bonds. By inhibiting these proteases, Cystatin C helps regulate protein turnover and prevent excessive proteolysis, which can lead to tissue damage .

Mode of Action

The inhibitory activity of Cystatin C is measured by its ability to inhibit the cleavage of a fluorogenic peptide substrate by papain, a model cysteine protease . The IC50 value, which represents the concentration of Cystatin C required to inhibit 50% of the protease activity, is less than 15 nM . This high affinity for cysteine proteases underscores its effectiveness as a protease inhibitor.

Regulatory Mechanisms

Cystatin C levels in the serum are closely correlated with the glomerular filtration rate (GFR), making it a valuable biomarker for kidney function . Elevated levels of Cystatin C are associated with an increased risk of coronary artery disease and cardiovascular events . Additionally, its expression and activity can be influenced by various physiological and pathological conditions, including inflammation and cancer .

Applications

Recombinant rat Cystatin C is widely used in research to study its role in various diseases and to develop therapeutic strategies targeting cysteine proteases. It is also utilized in enzyme-linked immunosorbent assays (ELISAs) to quantify Cystatin C levels in biological samples .

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.