CHI3 Antibody

Shipped with Ice Packs
In Stock

Description

Molecular and Functional Overview of CHI3L1

CHI3L1 (YKL-40 in humans, BRP39 in mice) is a 39 kDa glycoprotein belonging to the glycosyl hydrolase 18 family. Key features include:

  • Structural properties: Encoded by a 381-amino-acid sequence with a signal peptide and two disulfide bonds .

  • Expression: Secreted by macrophages, neutrophils, endothelial cells, and cancer cells .

  • Biological roles:

    • Modulates inflammation, cell survival, and extracellular matrix remodeling .

    • Promotes tumor angiogenesis, metastasis, and immune evasion .

Mechanism of CHI3L1 Antibody Action

Anti-CHI3L1 antibodies inhibit pathogenic pathways by:

  • Blocking CHI3L1-receptor interactions: Targets the chitosome receptor complex (IL-13Rα2/TMEM219), disrupting downstream MAPK, Akt, and Wnt/β-catenin signaling .

  • Suppressing M2 macrophage polarization: Reduces STAT6-dependent M2 polarization, limiting tumor-supportive microenvironments .

  • Neutralizing NETosis: Inhibits neutrophil extracellular trap (NET) formation, restoring T cell infiltration into tumors .

Table 1: Key Mechanisms and Outcomes of Anti-CHI3L1 Antibodies

MechanismFunctional ImpactDisease ModelSource
STAT6 inhibitionReduced M2 polarization, tumor growthLung cancer
NETosis suppressionEnhanced T cell cytotoxicityTriple-negative breast cancer
Chitosome disruptionAttenuated metastasis, inflammationGlioblastoma, NSCLC

Antitumor Efficacy

  • Lung cancer: Anti-CHI3L1 antibodies reduced tumor growth and metastasis in murine models by 60–70% via STAT6 inhibition .

  • Breast cancer: Blockade of CHI3L1 decreased NET-mediated T cell exclusion, suppressing tumor progression in preclinical trials .

  • Glioblastoma: Antibodies restored therapeutic sensitivity to tyrosine kinase inhibitors, inducing tumor cell death .

Beyond Oncology

  • Acetaminophen-induced liver injury (AILI): Antibody C59 (humanized anti-CHI3L1) reduced serum ALT levels by 10–200-fold in mice, demonstrating hepatoprotective effects .

Advantages

  • Multifunctional targeting: Simultaneously inhibits tumor growth, metastasis, and immunosuppression .

  • Cross-reactivity: Certain antibodies (e.g., C59) bind both murine and human CHI3L1, enhancing translational potential .

Challenges

  • Complex signaling: CHI3L1 interacts with plasminogen and heparan sulfate, complicating target specificity .

  • Biomarker variability: Serum CHI3L1 levels correlate with prognosis in cancer and asthma but lack disease specificity .

Current Research and Development

  • Ocean Biomedical’s anti-CHI3L1 antibody: Demonstrated efficacy in breast cancer, NSCLC, and glioblastoma across multiple preclinical studies .

  • Humanized antibodies: C59 and similar candidates are advancing toward clinical trials for AILI and cancer .

Future Directions

  • Combination therapies: Pairing anti-CHI3L1 antibodies with checkpoint inhibitors or chemotherapy .

  • Pan-cancer applications: Targeting CHI3L1 in EGFR-driven cancers (e.g., colon, glioblastoma) .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Made-to-order (14-16 weeks)
Synonyms
CHI3 antibody; CHI-L1 antibody; CHIL antibody; At5g05270 antibody; K18I23.7Probable chalcone--flavonone isomerase 3 antibody; Chalcone isomerase 3 antibody; EC 5.5.1.6 antibody; Chalcone isomerase-like 1 antibody
Target Names
CHI3
Uniprot No.

Target Background

Function
CHI3 is involved in anthocyanin biosynthesis.
Gene References Into Functions
  1. CHI3 functions in conjunction with TT5 to promote flavonoid production. PMID: 26347569
Database Links

KEGG: ath:AT5G05270

STRING: 3702.AT5G05270.1

UniGene: At.27434

Protein Families
Chalcone isomerase family

Q&A

What is CHI3L1 and why is it significant in immunological research?

Chitinase-3-like 1 (CHI3L1) is a glycoprotein that binds to chitin but lacks enzymatic activity, distinguishing it from true chitinases. It functions as a critical mediator in inflammatory processes, particularly in conditions like atopic dermatitis where it contributes to skin inflammation progression . CHI3L1 has gained significant attention because it operates at the intersection of multiple inflammatory pathways and influences processes ranging from tissue remodeling to inflammatory cell recruitment. In viral research, CHI3L1 has been identified as a host factor that enhances viral entry, particularly for SARS-CoV-2, by augmenting ACE2 receptor expression on epithelial cells . This unique positioning across different disease mechanisms makes CHI3L1 an attractive target for therapeutic antibody development.

How do anti-CHI3L1 antibodies exert their therapeutic effects?

Anti-CHI3L1 antibodies function through several interconnected mechanisms to produce their therapeutic effects. Primarily, they bind to CHI3L1 protein, inhibiting its activity and downstream signaling pathways. In atopic dermatitis models, anti-CHI3L1 antibody treatment suppresses epidermal thickening, reduces clinical scores, decreases IgE levels, and inhibits inflammatory cell infiltration . A key molecular mechanism involves inhibition of STAT3 phosphorylation and activity, which subsequently reduces the expression of inflammatory cytokines including IL-1β, IL-4, CXCL8, and TSLP . In viral infection models, particularly for SARS-CoV-2, anti-CHI3L1 antibodies (like the monoclonal antibody "FRG") demonstrate efficacy by reducing ACE2 receptor expression and inhibiting viral entry across multiple variants . These antibodies affect both exogenous and endogenous CHI3L1, providing comprehensive pathway inhibition.

What experimental models are used to evaluate CHI3L1 antibody efficacy?

Researchers employ several complementary models to evaluate anti-CHI3L1 antibody efficacy:

  • Phthalic anhydride-induced atopic dermatitis animal model: This in vivo system allows assessment of systemic and local effects on clinical parameters including epidermal thickening, inflammatory cell infiltration, and IgE levels .

  • Reconstructed Human Skin (RHS) model: This in vitro system provides a controlled environment using human skin equivalents to study molecular and cellular responses to CHI3L1 antibody treatment .

  • Pseudovirus infection models: For viral research, particularly SARS-CoV-2, cell lines like Calu-3 are infected with pseudoviruses expressing different spike protein variants to assess antibody efficacy against viral entry .

  • Molecular knockdown approaches: siRNA-mediated knockdown of CHI3L1 or STAT3 is used to verify the specificity of antibody effects and confirm mechanistic pathways .

These models collectively provide robust evidence for CHI3L1 antibody efficacy across different disease contexts and allow for detailed mechanistic studies.

What techniques are most effective for measuring CHI3L1 antibody activity?

Several complementary methodologies provide comprehensive assessment of CHI3L1 antibody activity:

  • Enzyme-linked immunosorbent assay (ELISA): Used to quantify CHI3L1 protein levels and associated inflammatory cytokines in tissue homogenates or biological fluids .

  • RT-qPCR: Employed to assess changes in mRNA expression of CHI3L1 and related inflammatory mediators (IL-1β, IL-4, CXCL8, TSLP) .

  • Western blotting: Critical for evaluating protein expression and activation status of downstream signaling molecules, particularly STAT3 phosphorylation .

  • Histological analysis: Includes H&E staining for tissue architecture assessment, immunohistochemistry for protein localization, and specific stains for inflammatory cell infiltration .

  • Flow cytometry (FACS): Particularly valuable in viral infection models to quantify viral uptake by target cells following antibody treatment .

  • Protein-association network analysis: Used to identify and validate interactions between CHI3L1 and other inflammatory mediators like CXCL8 .

The integration of these techniques allows researchers to comprehensively characterize both the direct binding of antibodies to CHI3L1 and their downstream functional effects on inflammatory and infectious processes.

How should researchers design experiments comparing CHI3L1 antibodies with established treatments?

When designing comparative experiments between CHI3L1 antibodies and established treatments (such as IL-4 antibodies), researchers should implement the following design elements:

  • Parallel treatment groups: Include untreated controls, CHI3L1 antibody treatment groups, established treatment groups (e.g., IL-4 antibody), and potentially combination treatment groups .

  • Dose-response relationships: Test multiple concentrations to identify optimal dosing and potential synergistic effects between treatments .

  • Comprehensive endpoint analysis: Measure multiple parameters including:

    • Clinical scores (for in vivo models)

    • Histological changes (epidermal thickness, inflammatory cell infiltration)

    • Molecular markers (cytokine profiles, signaling pathway activation)

    • Functional outcomes relevant to the specific disease model

  • Temporal dynamics assessment: Include time-course experiments to evaluate both immediate and sustained effects of treatments .

  • Positive and negative controls: Use appropriate control antibodies (e.g., isotype-matched IgG) to distinguish specific from non-specific effects .

This structured approach allows for rigorous comparison of efficacy, mechanistic differences, and potential complementary effects between CHI3L1 antibodies and established therapeutic approaches.

What methods verify the specificity of anti-CHI3L1 antibodies in experimental settings?

To verify antibody specificity, researchers should employ multiple complementary approaches:

  • Knockdown validation: Using CHI3L1 siRNA alongside antibody treatment to confirm that both approaches produce similar effects on downstream targets and inflammatory markers .

  • Recombinant protein competition: Testing whether excess recombinant CHI3L1 can compete with endogenous protein for antibody binding and reverse antibody effects .

  • Isotype control comparisons: Including appropriate IgG controls to distinguish specific from non-specific effects of antibody treatment .

  • Functional recovery experiments: Determining if adding recombinant CHI3L1 can overcome the inhibitory effects of the antibody on inflammatory processes or viral entry .

  • Cross-reactivity assessment: Confirming the antibody specifically recognizes CHI3L1 without binding to related proteins through techniques like Western blotting and immunoprecipitation .

These approaches collectively provide strong evidence for antibody specificity when positive results align across multiple methods, strengthening the validity of experimental findings.

What evidence supports CHI3L1 antibody efficacy in atopic dermatitis?

Substantial evidence supports the investigation of CHI3L1 antibody in atopic dermatitis, with key findings including:

  • Clinical manifestations: Anti-CHI3L1 antibody treatment suppresses phthalic anhydride-induced epidermal thickening and reduces clinical scores in animal models of atopic dermatitis .

  • Inflammatory markers: Treatment significantly decreases IgE levels and inflammatory cell infiltration in skin tissue, two hallmarks of atopic dermatitis .

  • Cytokine modulation: The antibody reduces concentrations of atopic dermatitis-related inflammatory cytokines including IL-1β, IL-4, CXCL8, and TSLP in both animal models and reconstructed human skin systems .

  • Comparative efficacy: The inhibitory effects of CHI3L1 antibody are comparable or superior to those of IL-4 antibody, which is an established therapeutic target for atopic dermatitis .

  • Mechanistic insights: CHI3L1 antibody inhibits STAT3 activity and disrupts the CHI3L1-CXCL8 inflammatory axis, providing clear molecular mechanisms for its therapeutic effects .

These findings collectively suggest that CHI3L1 antibody represents a promising therapeutic approach for atopic dermatitis, potentially offering advantages over or complementing existing treatments.

How does CHI3L1 antibody treatment affect histological parameters in skin inflammation?

CHI3L1 antibody administration produces several significant histological improvements in inflamed skin tissue:

  • Epidermal normalization: One of the most consistent findings is reduction of epidermal thickening, which is typically increased in atopic dermatitis .

  • Decreased inflammatory cell infiltration: Treatment significantly reduces the infiltration of inflammatory cells into the dermis and epidermis, improving tissue architecture .

  • Reduced vascular changes: Decreased vasodilation and endothelial activation are observed after treatment, contributing to reduced inflammation .

  • Molecular marker changes: Immunohistochemical analyses show decreased expression of inflammatory mediators within the tissue following CHI3L1 antibody treatment .

  • STAT3 signaling inhibition: Histological assessment demonstrates reduced phosphorylated STAT3 in treated tissues, confirming the molecular mechanism of action .

These histological improvements correlate with clinical score improvements and provide visual confirmation of the anti-inflammatory effects of CHI3L1 antibody at the tissue level, supporting its therapeutic potential in inflammatory skin conditions.

How do CHI3L1 antibodies compare with IL-4 antibodies in treating skin inflammation?

Direct comparisons between CHI3L1 and IL-4 antibodies have revealed important similarities and differences:

  • Efficacy comparison:

    • The inhibitory effects of CHI3L1 antibody were found to be similar or more effective compared to IL-4 antibody in reducing inflammatory markers and clinical scores .

    • Both antibodies effectively reduced epidermal thickening and inflammatory cell infiltration in atopic dermatitis models .

  • Mechanistic differences:

    • IL-4 antibody primarily targets the Th2 inflammatory pathway by neutralizing IL-4, a central cytokine in atopic dermatitis.

    • CHI3L1 antibody appears to have broader effects through STAT3 inhibition, affecting multiple cytokines including IL-1β, IL-4, CXCL8, and TSLP .

  • Target specificity:

    • IL-4 antibody specifically neutralizes the IL-4 cytokine, affecting downstream Th2 responses.

    • CHI3L1 antibody targets both exogenous and endogenous CHI3L1, potentially offering more comprehensive pathway inhibition .

These comparisons suggest that CHI3L1 antibody may offer certain advantages over IL-4 antibody treatment, particularly in its broader spectrum of anti-inflammatory effects through STAT3 pathway inhibition, though further clinical studies are needed for definitive comparison.

How does CHI3L1 influence SARS-CoV-2 infection and what is the role of CHI3L1 antibodies?

CHI3L1 has been identified as a critical host factor influencing SARS-CoV-2 infection through several mechanisms:

  • ACE2 regulation: CHI3L1 significantly augments the expression of ACE2 (the primary receptor for SARS-CoV-2) on epithelial cells. In experimental models, treatment with recombinant CHI3L1 increased ACE2 accumulation on Calu-3 cells .

  • Enhanced viral uptake: Studies using pseudoviruses with various SARS-CoV-2 spike protein mutations demonstrated that CHI3L1 significantly enhances viral uptake by epithelial cells across multiple variants including alpha, beta, gamma, and delta .

  • Universal mechanism: The stimulatory effect of CHI3L1 on viral entry appears to be conserved across different SARS-CoV-2 variants, positioning it as a "universal" host factor facilitating infection .

CHI3L1 antibodies counteract these effects through:

  • ACE2 expression inhibition: The monoclonal anti-CHI3L1 antibody (FRG) significantly reduces ACE2 expression on epithelial cells, limiting the receptor availability for viral binding .

  • Viral entry blockade: FRG effectively blocked pseudovirus uptake by epithelial cells across multiple SARS-CoV-2 variants, both under baseline conditions and when cells were stimulated with recombinant CHI3L1 .

  • Variant-independent action: The antibody maintained efficacy against pseudoviruses expressing spike proteins from multiple variants, suggesting potential as a broadly effective therapeutic approach .

These findings position CHI3L1 antibodies as promising host-directed therapeutic candidates that may remain effective despite viral mutations affecting traditional virus-directed approaches.

What evidence suggests CHI3L1 antibodies might be effective against multiple SARS-CoV-2 variants?

Several lines of experimental evidence support the potential of CHI3L1 antibodies as effective therapeutics against multiple SARS-CoV-2 variants:

  • Cross-variant efficacy:

    • The monoclonal anti-CHI3L1 antibody (FRG) effectively blocked pseudovirus uptake by Calu-3 cells across multiple SARS-CoV-2 variants including the ancestral strain with G614 mutation, and alpha, beta, gamma, and delta variants .

    • This inhibition was consistent regardless of the specific spike protein mutations present in different variants.

  • Host-directed mechanism:

    • By targeting a host factor (CHI3L1) rather than viral components, the antibody's mechanism of action circumvents the immune evasion strategies of different variants .

    • This differs fundamentally from spike-targeting antibodies that may lose efficacy as the spike protein mutates.

  • ACE2 modulation:

    • Immunocytochemical evaluations demonstrated that FRG antibody treatment significantly reduced ACE2 expression on epithelial cells across variants .

    • Since ACE2 is the primary entry receptor for all SARS-CoV-2 variants, this mechanism provides broad protection.

  • Dual activity:

    • FRG was effective against both endogenous and exogenous CHI3L1, suggesting comprehensive pathway inhibition .

    • This dual activity enhances the antibody's potential effectiveness across different infection scenarios.

These findings collectively indicate that CHI3L1 antibodies represent a promising host-directed therapeutic approach with potential efficacy against both current and emerging SARS-CoV-2 variants.

How does CHI3L1 antibody modulate the STAT3 signaling pathway?

CHI3L1 antibody modulates the STAT3 signaling pathway through several interconnected mechanisms:

  • Inhibition of STAT3 phosphorylation:

    • Anti-CHI3L1 antibody treatment significantly reduces STAT3 phosphorylation (activation) in both atopic dermatitis animal models and in vitro systems .

    • Western blotting analysis confirms decreased phosphorylated STAT3 (p-STAT3) relative to total STAT3 protein following antibody treatment.

  • Functional validation through knockdown studies:

    • siRNA-mediated knockdown of either CHI3L1 or STAT3 produced similar effects on inflammatory cytokine expression, confirming their functional relationship .

    • Specifically, siRNA of CHI3L1 blocked the expression of CHI3L1 and p-STAT3, while siRNA of STAT3 reduced the mRNA levels of multiple inflammatory cytokines .

  • Downstream gene expression effects:

    • Inhibition of STAT3 activation leads to reduced expression of multiple inflammatory genes including IL-1β, IL-4, CXCL8, and TSLP .

    • This broad inhibitory effect explains the comprehensive anti-inflammatory activity of CHI3L1 antibody in atopic dermatitis and potentially other inflammatory conditions.

  • Correlation with therapeutic outcomes:

    • The degree of STAT3 inhibition correlates with reduced clinical scores and histological improvement in atopic dermatitis models .

These findings establish STAT3 signaling as a central mechanism through which CHI3L1 antibody exerts its therapeutic effects, providing a molecular basis for its efficacy in inflammatory conditions.

What is the relationship between CHI3L1 and CXCL8 in inflammation?

Research has uncovered a significant functional relationship between CHI3L1 and CXCL8 in inflammatory processes:

  • Network association:

    • Protein-association network analysis identified CXCL8 as a key mediator connected to CHI3L1 function .

    • This association appears to be critical for inflammatory signaling in conditions like atopic dermatitis.

  • Expression correlation:

    • CHI3L1 levels positively correlate with CXCL8 expression in inflammatory conditions .

    • When CHI3L1 is inhibited by antibody treatment or siRNA knockdown, CXCL8 expression significantly decreases .

  • Mechanistic connection through STAT3:

    • The relationship between CHI3L1 and CXCL8 appears to be mediated through STAT3 signaling .

    • CHI3L1 and p-STAT3 expression levels correlate with CXCL8 levels in experimental models, particularly in the reconstructed human skin model .

    • siRNA knockdown of STAT3 reduces the mRNA levels of both CHI3L1 and CXCL8, confirming this signaling axis .

  • Functional significance:

    • CXCL8 (also known as IL-8) is a potent chemoattractant for neutrophils and plays a key role in acute inflammation.

    • The CHI3L1-STAT3-CXCL8 axis represents a central pathway in inflammatory amplification in multiple disease contexts .

This relationship provides a mechanistic explanation for how CHI3L1 antibody reduces neutrophilic inflammation and suggests that CXCL8 inhibition is an important component of its therapeutic effect in inflammatory conditions.

How might AI-based technologies enhance the design of next-generation CHI3L1 antibodies?

AI-based technologies offer several promising approaches to enhance CHI3L1 antibody design:

  • De novo antibody sequence generation:

    • Language models like IgLM can generate novel CDRH3 sequences specifically targeting CHI3L1 .

    • These models can explore sequence space more efficiently than traditional methods, potentially identifying novel binding solutions .

    • Generated sequences can exhibit substantial diversity in composition and length while maintaining target specificity .

  • Structure-based optimization:

    • Tools like ImmuneBuilder can model the structure of generated antibody sequences bound to CHI3L1 .

    • Structural similarity to known effective antibodies can be used to down-select promising candidates .

    • Virtual screening can prioritize antibodies with optimal binding characteristics before experimental validation.

  • Efficiency improvements:

    • AI-designed antibodies have achieved hit rates of approximately 15% in some studies, significantly reducing the resources required for antibody discovery .

    • This approach bypasses traditional requirements for source samples with previous exposure to the target antigen .

    • Virtual testing can prioritize candidates before resource-intensive experimental validation.

  • Novel binding solutions:

    • AI methods may identify binding approaches that differ from naturally occurring antibodies .

    • Generated antibody sequences typically show distinction from known natural antibodies against the same target .

These technologies could lead to CHI3L1 antibodies with enhanced specificity, improved therapeutic properties, and novel mechanisms of action beyond what traditional discovery methods might achieve.

What potential combination therapies involving CHI3L1 antibodies should researchers investigate?

Several promising combination therapy approaches involving CHI3L1 antibodies warrant investigation:

  • Combinations with cytokine-targeting antibodies:

    • CHI3L1 + IL-4/IL-13 antibodies: Given the comparative efficacy data between CHI3L1 and IL-4 antibodies, their combination might provide enhanced benefits in Th2-driven conditions like atopic dermatitis .

    • CHI3L1 + other inflammatory cytokine inhibitors: Targeting multiple inflammatory pathways simultaneously could provide more comprehensive disease control.

  • Antiviral combination approaches:

    • CHI3L1 antibodies + direct-acting antivirals for SARS-CoV-2: The host-directed mechanism of CHI3L1 antibodies could complement virus-directed therapies .

    • CHI3L1 antibodies + spike-targeting antibodies: Potential synergy through targeting both viral entry mechanisms and host factors .

  • Pathway-specific combinations:

    • CHI3L1 antibodies + JAK inhibitors: Since both affect STAT signaling through different mechanisms, this combination could provide enhanced pathway inhibition .

    • CHI3L1 antibodies + targeted CXCL8 inhibitors: Directly targeting both elements of the identified CHI3L1-CXCL8 axis could provide synergistic benefits .

  • Delivery system innovations:

    • Tissue-targeted delivery approaches: Specifically delivering CHI3L1 antibodies to affected tissues could enhance efficacy while reducing systemic exposure.

    • Combination with small molecule inhibitors of related pathways.

Systematic investigation of these combination approaches should include assessment of additive versus synergistic effects, optimal dosing ratios, and potential for reduced side effects through dose-sparing of individual components.

What methodological approaches are needed to advance CHI3L1 antibody research?

To advance CHI3L1 antibody research, several methodological approaches should be prioritized:

  • Receptor identification and characterization:

    • Definitive identification of specific receptor(s) through which CHI3L1 signals.

    • Characterization of receptor distribution across different cell types and tissues.

    • Development of tools to monitor receptor-ligand interactions in real-time.

  • Expanded disease models:

    • Development of genetic models with CHI3L1 alterations to complement the current induced models .

    • Humanized animal models to better predict translational outcomes.

    • Patient-derived organoid systems for personalized response testing.

  • Advanced imaging techniques:

    • Intravital microscopy to visualize CHI3L1-antibody interactions in living tissues.

    • Multi-parameter imaging to simultaneously track multiple inflammatory markers.

    • Correlative light and electron microscopy to link molecular events with ultrastructural changes.

  • Systems biology approaches:

    • Network analysis to position CHI3L1 within broader inflammatory cascades .

    • Multi-omics integration (transcriptomics, proteomics, metabolomics) to comprehensively assess antibody effects.

    • Computational modeling of CHI3L1 signaling networks to predict intervention points.

  • Biomarker development:

    • Identification of predictive biomarkers for treatment response.

    • Development of companion diagnostics for patient selection.

    • Pharmacodynamic markers for dose optimization.

  • Antibody engineering advances:

    • Bispecific antibodies targeting CHI3L1 and complementary inflammatory mediators.

    • Format optimization (Fab, F(ab')2, IgG subclasses) for specific disease applications.

    • Site-specific modifications to enhance tissue penetration or half-life.

These methodological advances would address current knowledge gaps and accelerate the translation of CHI3L1 antibody research into clinical applications across multiple disease areas.

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.