CST8 Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Background

  1. CST8 as Cystatin 8:

    • The CST8 gene encodes cystatin 8, a member of the cystatin family of cysteine protease inhibitors. This protein is primarily expressed in salivary glands and epithelial tissues .

    • No studies in the reviewed literature explicitly describe antibodies targeting cystatin 8.

  2. Potential Typographical Confusion:

    • The term may refer to caspase-8 (CASP8) antibodies, which target the apoptosis-related protease caspase-8. One commercial product, Caspase-8 Antibody #4927, is available for research use (e.g., Western blotting) .

    • Alternatively, "CST8" might be conflated with CD8 antibodies, which target the CD8 glycoprotein on cytotoxic T cells. These are well-documented in immunology research .

Caspase-8 Antibody #4927

PropertyDetails
TargetCaspase-8 (CASP8)
ReactivityMouse
ApplicationsWestern Blotting (1:1000 dilution)
SourceRabbit monoclonal antibody
Molecular Weight45 kDa (pro-form), 57 kDa (cleaved form)
Commercial UseResearch-only; not approved for diagnostic or therapeutic purposes

Key Notes:

  • Caspase-8 plays a critical role in apoptosis and necroptosis pathways.

  • Antibody #4927 detects endogenous caspase-8 in mouse models but has no reported clinical applications .

Anti-CD8 Antibodies

Anti-CD8 antibodies are extensively studied for their roles in T cell activation and therapeutic applications. Key findings include:

Antibody NameFunctionalityResearch Findings
OKT8Activates CD8+ T cells via FcγRIIa engagementInduces cytokine release and cytotoxicity in human CD8+ T cells .
CT-CD8a/CD8bEnhances TCR/pMHCI binding in miceImproves tetramer staining and T cell visualization .
GAALIE VariantEngineered IgG with FcγRIIa selectivityBoosts dendritic cell maturation and CD8+ T cell responses in viral infections .

Mechanistic Insights:

  • Anti-CD8 antibodies like OKT8 trigger effector functions (e.g., cytokine release) without requiring TCR engagement .

  • Fc-engineered anti-CD8 antibodies (e.g., GAALIE) enhance adaptive immunity by activating dendritic cells, leading to protective CD8+ T cell responses against viral pathogens .

Clinical and Therapeutic Relevance

  • CD8 Antibodies in Oncology:

    • Depleting intratumoral regulatory T cells (itTregs) via anti-CCR8 antibodies (e.g., CHS-114) enhances anti-PD-1 therapy efficacy in preclinical models .

    • Bispecific antibodies like cibisatamab (CEA-CD3) redirect CD8+ T cells to tumors, increasing intra-tumoral T cell infiltration .

  • SARS-CoV-2 Mutations:

    • Chronic infection in immunocompromised patients drives mutations in CD8+ T cell epitopes, suggesting immune evasion pressures .

Data Gaps and Limitations

  • No peer-reviewed studies or commercial products explicitly targeting CST8 (cystatin 8) were identified in the provided sources.

  • Research on caspase-8 and CD8 antibodies remains siloed, with limited cross-disciplinary integration.

Product Specs

Buffer
PBS with 0.02% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze/thaw cycles.
Lead Time
Typically, we can ship your order within 1-3 business days of receiving it. Delivery time may vary depending on the purchase method or location. Please contact your local distributor for specific delivery information.
Synonyms
CRES antibody; CST 8 antibody; Cst8 antibody; CST8_HUMAN antibody; cystatin 8 (cystatin related epididymal specific) antibody; Cystatin 8 antibody; Cystatin related epididymal specific antibody; Cystatin related epididymal specific protein antibody; Cystatin related epididymal spermatogenic protein antibody; Cystatin related protein epididymis specific antibody; Cystatin-8 antibody; Cystatin-related epididymal spermatogenic protein antibody
Target Names
CST8
Uniprot No.

Target Background

Function
CST8 Antibody plays a specialized role in sperm development and maturation.
Gene References Into Functions
  1. Phylogenetic studies indicate that CTES genes are grouped into three distinct categories: testatin, Cres, and CstL1. PMID: 20565543
  2. Research suggests that cystatin-related epididymal spermatogenic protein is localized to the sperm equatorial segment and becomes accessible to the external environment during fertilization. PMID: 12193387
Database Links

HGNC: 2480

OMIM: 608683

KEGG: hsa:10047

STRING: 9606.ENSP00000246012

UniGene: Hs.121602

Protein Families
Cystatin family
Subcellular Location
Secreted.
Tissue Specificity
Proximal caput region of the epididymis. Lower expression in the testis. Within the testis it is localized to the elongating spermatids, whereas within the epididymis it is exclusively synthesized by the proximal caput epithelium.

Q&A

What is CST8 and why is it a significant research target?

CST8 (Cystatin 8), also known as CRES (Cystatin-related epididymal spermatogenic protein), is a member of the cystatin superfamily that functions primarily as an inhibitor of cysteine proteases. These proteases play crucial roles in various physiological processes including protein degradation and immune response regulation. The significance of CST8 as a research target stems from evidence linking dysregulation of cysteine proteases to multiple pathological conditions including cancer, neurodegenerative disorders, and inflammatory diseases. Understanding CST8's inhibitory mechanisms provides valuable insights into these disease processes and potential therapeutic interventions. Research utilizing CST8-specific antibodies enables detailed investigation of its expression patterns, interactions, and functional roles in different tissues and disease states .

How do CST8 polyclonal and monoclonal antibodies differ in research applications?

The fundamental difference between polyclonal and monoclonal CST8 antibodies lies in their epitope recognition patterns and consequent experimental utility. Polyclonal CST8 antibodies, typically raised in rabbits against recombinant fusion proteins containing CST8 amino acid sequences, recognize multiple epitopes on the CST8 antigen. This multi-epitope recognition provides robust signal amplification particularly beneficial in applications where protein abundance is low. For example, the rabbit polyclonal antibody to CST8 detailed in the search results demonstrates versatility across Western blot, IHC, and ELISA applications .

Monoclonal antibodies, conversely, recognize a single epitope on the CST8 protein, offering superior specificity but potentially reduced sensitivity compared to polyclonal variants. This specificity is particularly valuable in research scenarios requiring discrimination between closely related cystatin family members or specific CST8 conformational states. When selecting between these antibody types, researchers should consider experimental priorities: polyclonal antibodies excel in detection sensitivity while monoclonal antibodies provide greater epitope specificity.

What explains the discrepancy between calculated and observed molecular weights for CST8?

The discrepancy between CST8's calculated molecular weight (16kDa) and its observed molecular weight in experimental conditions (27kDa) represents a common phenomenon in protein research that can be attributed to several post-translational factors . This molecular weight difference occurs primarily due to:

  • Post-translational modifications (PTMs): CST8 undergoes various PTMs including glycosylation, phosphorylation, and other covalent modifications that increase its apparent molecular weight on SDS-PAGE.

  • Protein structure and charge distribution: Non-uniform SDS binding due to unusual charge distributions or structural elements can alter migration patterns.

  • Signal peptide retention: Incomplete processing of signal sequences may contribute to higher observed molecular weights.

When conducting Western blot analyses of CST8, researchers should anticipate this molecular weight discrepancy and utilize appropriate positive controls (such as mouse testis or liver samples as indicated in the antibody specifications) to confirm band identity . Additionally, validation through complementary techniques such as immunoprecipitation followed by mass spectrometry can provide definitive confirmation of protein identity when molecular weight variations raise concerns about specificity.

How should researchers optimize Western blot protocols specifically for CST8 detection?

Optimizing Western blot protocols for CST8 detection requires methodical attention to several critical parameters. Based on the available data for CST8 antibodies, the following optimization approach is recommended:

  • Sample preparation: For CST8 detection, tissue lysates from testis or liver (positive control tissues) should be prepared using RIPA buffer supplemented with protease inhibitors. Complete denaturation is essential, requiring heating at 95°C for 5 minutes in reducing sample buffer.

  • Gel percentage selection: Given CST8's observed molecular weight of 27kDa despite its calculated 16kDa size, 12-15% polyacrylamide gels provide optimal resolution in this molecular weight range .

  • Transfer conditions: Semi-dry transfer at 15V for 30 minutes or wet transfer at 100V for 60 minutes using PVDF membranes (0.22μm pore size) typically yields optimal results for CST8.

  • Antibody dilution: The recommended dilution range for CST8 polyclonal antibodies in Western blot applications is 1:500 - 1:2000 . Researchers should conduct titration experiments to determine optimal concentration for their specific samples.

  • Detection system: Enhanced chemiluminescence (ECL) provides sufficient sensitivity for most CST8 detection purposes. For challenging samples with low CST8 expression, amplified detection systems may be necessary.

When troubleshooting Western blots for CST8, researchers should account for the molecular weight discrepancy (observed 27kDa vs. calculated 16kDa) and verify specificity using positive control tissues like mouse testis or liver as indicated in antibody specifications .

What are the critical parameters for successful immunohistochemical detection of CST8?

Successful immunohistochemical (IHC) detection of CST8 in tissue samples depends on careful optimization of several parameters to maximize signal specificity while minimizing background. The following methodological approach is recommended based on the performance characteristics of available CST8 antibodies:

  • Fixation and antigen retrieval: Formalin-fixed paraffin-embedded tissues typically require heat-induced epitope retrieval using citrate buffer (pH 6.0) or EDTA buffer (pH 9.0). For CST8, citrate buffer-based retrieval (10mM, pH 6.0) at 95-100°C for 20 minutes often yields optimal results.

  • Blocking parameters: A dual blocking approach is recommended - first with hydrogen peroxide (3% for 10 minutes) to quench endogenous peroxidases, followed by protein blocking (5% normal serum from the same species as the secondary antibody) for 1 hour at room temperature.

  • Primary antibody incubation: CST8 antibodies should be applied at dilutions ranging from 1:100 to 1:500 depending on the specific antibody. Overnight incubation at 4°C typically provides the best balance between specific signal development and background minimization .

  • Detection system selection: For chromogenic detection, polymer-based systems offer superior sensitivity compared to conventional ABC methods for CST8 visualization. DAB (3,3'-diaminobenzidine) substrate provides good contrast for evaluating CST8 expression patterns.

  • Controls: Implementation of both positive controls (tissues known to express CST8 such as testis) and negative controls (primary antibody omission and isotype controls) is essential for validating staining specificity.

When evaluating CST8 expression patterns by IHC, researchers should pay particular attention to its secreted cellular localization and interpret staining patterns accordingly .

How can researchers validate antibody specificity for CST8 versus other cystatin family members?

Validating CST8 antibody specificity against other cystatin family members requires a multi-layered approach that addresses both experimental and computational validation. The following comprehensive strategy ensures confident discrimination between CST8 and related cystatins:

  • Sequence-based validation: Begin with computational analysis comparing the immunogen sequence (amino acids 53-142 of human CST8 as indicated in the specification ) against other cystatin family members using tools like BLAST. Identify unique epitopes that distinguish CST8 from related proteins.

  • Knockout/knockdown validation: Utilize CST8 knockout or knockdown models (siRNA, CRISPR-Cas9) to confirm antibody specificity. Absence or reduction of signal in these models provides strong evidence for specificity.

  • Recombinant protein panel testing: Test antibody reactivity against a panel of purified recombinant cystatin family proteins. Quantitative analysis of binding affinities can reveal cross-reactivity profiles.

  • Peptide competition assays: Pre-incubate CST8 antibody with increasing concentrations of synthetic peptides corresponding to the immunogen sequence. Specific binding should be progressively inhibited by the cognate peptide but not by control peptides from other cystatins.

  • Mass spectrometry validation: Perform immunoprecipitation using the CST8 antibody followed by mass spectrometry analysis of the precipitated proteins to confirm identity and detect any cross-reacting proteins.

What methodological approaches can address contradictory results when multiple anti-CST8 antibodies yield different experimental outcomes?

Conflicting results between different anti-CST8 antibodies represent a significant challenge in research that requires systematic investigation. When faced with discrepant findings, researchers should implement the following methodological approach:

  • Epitope mapping analysis: Determine the specific epitopes recognized by each antibody. Differences in epitope recognition may explain discrepant results if certain epitopes are masked in particular experimental conditions or protein conformations. Epitope mapping can be performed through peptide arrays or hydrogen-deuterium exchange mass spectrometry.

  • Antibody validation matrix: Create a comprehensive validation matrix analyzing each antibody across multiple techniques (Western blot, IHC, IP, ELISA) and samples. This systematic approach can reveal technique-specific limitations of particular antibodies.

  • Orthogonal detection methods: Implement non-antibody-based detection methods such as mass spectrometry or RNA expression analysis (RT-qPCR, RNA-seq) to provide antibody-independent verification of protein presence.

  • Structural considerations: Assess whether discrepancies might be explained by recognition of different CST8 conformational states, splice variants, or post-translationally modified forms by different antibodies.

  • Combined antibody approach: For critical experiments, consider using multiple antibodies in parallel or in sequence (e.g., different antibodies for capture and detection in sandwich ELISA) to provide more comprehensive detection.

How does CST8 antibody performance compare in detecting native versus denatured protein conformations?

The differential performance of CST8 antibodies in detecting native versus denatured protein conformations represents a critical consideration for experimental design. Based on available data, CST8 antibodies exhibit distinctive performance characteristics across these conformational states:

  • Conformational epitope recognition: Polyclonal CST8 antibodies, particularly those generated against recombinant fusion proteins containing amino acids 53-142 of human CST8 , typically recognize both linear (denatured) and conformational (native) epitopes, though with varying efficiency.

  • Application-specific performance profiles:

    • Western blot: Excellent performance detecting denatured CST8 (observed at 27kDa despite calculated 16kDa)

    • ELISA: Generally effective for native protein detection

    • Immunohistochemistry: Variable performance depending on fixation/retrieval methods that may partially denature the protein

    • Immunoprecipitation: Requires antibodies capable of recognizing native conformations

  • Buffer and condition considerations: Native condition detection requires non-denaturing buffers that preserve protein folding. Detergents like NP-40 or Triton X-100 at low concentrations (0.1-0.5%) are preferred over stronger denaturing agents like SDS for maintaining CST8 native conformation.

  • Cross-linking considerations: When studying protein-protein interactions involving CST8, mild cross-linking (0.5-1% formaldehyde) may help preserve interaction complexes for subsequent antibody-based detection.

Researchers should select CST8 antibodies based on the specific conformational state required for their experimental objectives. For studies requiring detection of native CST8 (such as investigating protein-protein interactions or enzymatic inhibitory functions), antibodies validated specifically for native condition applications should be prioritized .

What are the most common causes of false positive and false negative results in CST8 antibody applications?

Reliable interpretation of CST8 antibody-based experiments requires awareness of potential sources of false results. The following analysis details common causes and mitigation strategies for both false positive and false negative outcomes:

False Positive Results:

  • Cross-reactivity with related cystatins: The structural similarity between CST8 and other cystatin family members can lead to non-specific binding. Mitigation requires thorough validation using peptide competition assays and testing in CST8-deficient systems.

  • Non-specific secondary antibody binding: Particularly in tissues with high endogenous immunoglobulin content. Control by including secondary-only controls and using appropriate blocking (5% serum from secondary antibody host species).

  • Endogenous peroxidase or phosphatase activity: Can cause false signals in IHC/ICC applications. Address through appropriate quenching steps (3% H₂O₂ treatment for peroxidases).

  • Sample-specific artifacts: Certain tissues may contain components that non-specifically bind antibodies. Validation across multiple sample types and with multiple detection methods is essential.

False Negative Results:

  • Epitope masking: Post-translational modifications or protein-protein interactions may obscure antibody binding sites. Test multiple antibodies targeting different CST8 epitopes.

  • Insufficient antigen retrieval: Critical for FFPE samples in IHC. Optimize using different retrieval buffers (citrate pH 6.0 vs. EDTA pH 9.0) and conditions.

  • Protein degradation: CST8 may be degraded during sample preparation. Include protease inhibitors and process samples rapidly at cold temperatures.

  • Insufficient sensitivity: Low abundance of CST8 may require signal amplification. Consider using polymer-based detection systems or tyramide signal amplification.

  • Incorrect molecular weight assessment: Remember that CST8 runs at 27kDa despite its calculated 16kDa size . Looking at incorrect molecular weight regions may lead to false negative interpretation.

Implementation of appropriate positive controls (mouse testis or liver for CST8) and negative controls (isotype controls, antigen competition) is essential for distinguishing true from false results in all CST8 antibody applications.

How should researchers interpret multiple bands or unexpected molecular weight patterns in CST8 Western blots?

The interpretation of multiple bands or unexpected molecular weight patterns in CST8 Western blots requires systematic analysis considering both technical and biological factors. The following structured approach guides researchers through this interpretive process:

  • Expected pattern baseline: CST8 typically presents as a 27kDa band despite its calculated molecular weight of 16kDa . This discrepancy is well-documented and should serve as the reference point for interpretation.

  • Higher molecular weight bands interpretation:

    • ~34-35kDa: May represent glycosylated CST8 forms

    • ~50-55kDa: Could indicate CST8 dimers resistant to denaturation

    • 75kDa: Might represent CST8 in complex with binding partners

  • Lower molecular weight bands interpretation:

    • 14-16kDa: May represent proteolytic degradation products or non-modified CST8

    • <14kDa: Typically indicates degradation artifacts

  • Validation approaches for multiplet band patterns:

    • Peptide competition assays: Specific bands should disappear with increasing concentrations of blocking peptide

    • Deglycosylation experiments: Treatment with PNGase F or similar enzymes will collapse glycosylation-based band multiplicity

    • Sample preparation variations: Alter lysis buffers, detergent concentrations, and reducing agent strength to assess effects on band patterns

  • Tissue-specific variation considerations: CST8 expression and post-translational modifications can differ substantially between tissues. Compare experimental samples with documented positive controls (mouse testis or liver) .

When encountering unexpected band patterns, researchers should systematically document conditions (sample source, preparation method, gel percentage, transfer time) and perform validation experiments to distinguish biologically relevant signals from technical artifacts. Particularly for CST8, with its known molecular weight discrepancy, rigorous validation is essential for accurate interpretation.

What quality control procedures should be implemented when working with CST8 antibodies across different experimental batches?

Implementing robust quality control procedures when working with CST8 antibodies across different experimental batches is essential for research reproducibility. The following comprehensive quality control framework ensures consistent antibody performance:

Pre-Experimental Quality Control:

  • Initial validation panel: For each new lot of CST8 antibody, perform:

    • Titration experiments to determine optimal working concentration

    • Western blot using positive control tissues (mouse testis/liver) to confirm expected 27kDa band

    • Peptide competition assay to verify specificity

    • ELISA-based binding curves against recombinant CST8

  • Reference standard preparation: Create aliquots of well-characterized positive control samples (recombinant protein and tissue lysates) sufficient for comparison across multiple experiments.

  • Stability testing: Assess antibody performance after multiple freeze-thaw cycles to establish handling guidelines.

Intra-Experimental Controls:

  • Internal controls for each experiment:

    • Positive control (known CST8-expressing sample)

    • Negative control (sample known to lack CST8)

    • Loading/processing controls appropriate to the technique

  • Antibody cocktail preparation: When possible, prepare sufficient antibody dilution for all samples within an experimental series.

  • Technical replicates: Include at least duplicate measurements for critical samples.

Batch Comparison Analysis:

  • Quantitative performance metrics:

    • Signal-to-noise ratio calculation for each batch

    • Coefficient of variation (CV) determination across technical replicates

    • Limit of detection comparison between batches

  • Calibration curve inclusion: For quantitative applications, include a calibration curve in each batch to enable mathematical normalization.

  • Statistical analysis plan: Implement appropriate statistical methods to account for batch effects in data analysis.

This systematic quality control approach provides a framework for detecting and mitigating batch-related variability in CST8 antibody performance, enhancing data reliability and reproducibility across extended research programs .

How can CST8 antibodies be effectively utilized in multiplex immunoassays with other autoantibodies?

Multiplex immunoassays incorporating CST8 antibodies alongside other autoantibodies require careful optimization to maintain specificity while enabling simultaneous detection. The following methodological approach maximizes multiplex assay performance:

  • Cross-reactivity matrix assessment: Before multiplexing, systematically evaluate cross-reactivity between each antibody pair (primary-primary and primary-secondary) to identify and mitigate potential interference. This is particularly important when combining CST8 antibodies with other autoantibodies, as demonstrated in studies of autoimmune encephalitis where multiple antibodies coexist .

  • Spectral overlap mitigation strategies:

    • For fluorescence-based multiplexing: Select fluorophores with minimal spectral overlap and implement appropriate compensation matrices

    • For chromogenic multiplexing: Utilize spatially distinct substrates (membrane vs. nuclear) or sequential detection protocols

  • Optimization of antibody concentrations: In multiplex formats, optimal antibody concentrations often differ from those in single-plex assays. Titrate each antibody independently and in combination to identify concentrations that maximize specific signal while minimizing background.

  • Blocking strategy enhancement: In multiplex settings, more robust blocking is typically required. Consider implementing:

    • Multi-component blocking solutions (combination of different proteins)

    • Sequential blocking steps targeting different binding mechanisms

    • Extended blocking durations (2+ hours at room temperature)

  • Validation using defined sample panels:

    • Samples containing only CST8 antibodies

    • Samples containing only other target autoantibodies

    • Samples containing defined mixtures of antibodies

    • Negative control samples

Research on autoimmune encephalitis demonstrates that approximately 8% of antibody-positive patients have two or more coexisting antibodies , highlighting the clinical relevance of multiplex detection capabilities. Effective multiplexing enables more comprehensive characterization of complex immunological profiles while conserving limited sample materials.

What role do T-cell responses play alongside CST8 antibodies in immune protection mechanisms?

The interplay between CST8-specific antibodies and T-cell responses represents a critical yet under-investigated dimension of immune protection mechanisms. Recent research on immune responses provides valuable insights applicable to CST8 biology:

  • Coordinated humoral and cellular immunity: Studies on vaccine breakthrough infections demonstrate that protection correlates with both antibody responses and CD4+/CD8+ T-cell activity . In the context of CST8, this suggests optimal immune responses likely require both antibody-mediated neutralization of secreted CST8 and T-cell recognition of CST8-expressing cells.

  • CD4+ T-cell contributions: CD4+ T cells provide essential support for high-quality antibody responses against CST8 through:

    • Germinal center formation enabling affinity maturation

    • Cytokine production directing appropriate antibody isotype switching

    • Memory B-cell development for sustained antibody responses

  • CD8+ T-cell effector functions: CD8+ T cells complement antibody-mediated protection through:

    • Direct cytolysis of cells expressing aberrant levels of CST8

    • IFN-γ production enhancing antigen presentation

    • Establishment of tissue-resident memory providing rapid local responses

  • Cross-reactive T-cell responses: Evidence indicates that cross-reactive T cells may play protective roles , suggesting T-cell responses targeting conserved epitopes across cystatin family members might provide broader protection than highly specific antibody responses.

  • Methodological approaches for integrated analysis:

    • ELISpot assays to quantify CST8-specific T-cell responses

    • Intracellular cytokine staining to characterize T-cell functionality

    • Tetramer analysis to enumerate epitope-specific T cells

Research examining breakthrough infections found evidence supporting roles for both CD4+ and CD8+ T cells alongside antibodies in protection , highlighting the importance of investigating integrated immune responses rather than focusing exclusively on antibody-mediated mechanisms when studying CST8 biology.

How can biophysics-informed computational modeling improve CST8 antibody design for therapeutic applications?

Biophysics-informed computational modeling offers sophisticated approaches to CST8 antibody design that transcend traditional experimental limitations. This advanced methodology enables precise engineering of antibodies with customized binding profiles particularly valuable for therapeutic applications:

  • Energy function optimization approach: The methodology employs energy functions (E) associated with specific binding modes (w) to generate antibodies with desired properties. For CST8-targeted therapeutics, this allows:

    • Designing highly specific antibodies that bind CST8 but not related cystatins by minimizing energy functions for CST8 binding while maximizing those for unwanted interactions

    • Creating cross-specific antibodies that interact with predetermined subsets of cystatin family members by jointly minimizing energy functions for desired targets

  • Epitope-focused design strategy: Rather than selecting from existing antibody libraries, this approach computationally designs complementarity-determining regions (CDRs) optimized for:

    • Targeting functionally critical epitopes on CST8 that may be under-represented in natural antibody repertoires

    • Maximizing binding affinity while controlling cross-reactivity with related proteins

    • Enhancing stability and manufacturability parameters critical for therapeutic development

  • Integration with experimental validation:

    • Computational predictions guide focused experimental testing of promising candidates

    • Iterative refinement based on experimental feedback improves model accuracy

    • Combined approach addresses limitations of both computational prediction and traditional selection methods

  • Therapeutic advantage analysis:

    • Enables targeting of specific CST8 conformational states associated with disease

    • Facilitates development of antibodies that modulate rather than simply bind CST8

    • Potential for reduced immunogenicity through strategic framework selection

This biophysics-informed approach has demonstrated success in creating antibodies with customized specificity profiles and offers particular value for therapeutic applications where precise targeting is essential . For CST8-related therapeutic development, this methodology provides a powerful framework for designing antibodies that specifically modulate CST8 function while minimizing off-target effects on related cystatin family members.

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 2025 TheBiotek. All Rights Reserved.