CXE12 Antibody

Shipped with Ice Packs
In Stock

Description

Preclinical Efficacy in Alopecia Areata

In murine AA models, CXCL12 antibody demonstrated:

  • Delayed Disease Onset: Treated mice showed 40% slower progression of hair loss compared to controls .

  • Transcriptional Normalization: 78% of upregulated genes in AA models (e.g., Ifng, Cd8a, Ccr5) were downregulated post-treatment .

  • Immune Cell Redistribution:

    Cell TypeAA Model (%)AA + Antibody (%)
    CD8+ T Cells4.22.5
    DC/Macrophages1.20.9
    Source: Single-cell RNA-seq analysis of AA mouse models .

Broader Therapeutic Applications

  • Cancer: Disrupts CXCL12-mediated metastasis and angiogenesis .

  • Inflammatory Diseases: Reduces neutrophil chemotaxis in conditions like rheumatoid arthritis .

Pharmacodynamic Profile

  • Ligand Inhibition: Blocks CXCL12 binding at picomolar affinity (IC₅₀: 0.5–9.5 nM for related chemokine receptors) .

  • Safety: Minimal off-target effects, with <10% of DEGs linked to non-AA pathways .

Clinical Development Landscape

CXCL12 antibody is part of a growing class of chemokine-targeted therapies. Related clinical trials include:

TargetAntibody NamePhaseConditionStatus
CXCR4MDX-1338I/IIMultiple MyelomaActive
CCR4MogamulizumabIIICutaneous T-Cell LymphomaRecruiting

Adapted from chemokine receptor antibody trials .

Challenges and Future Directions

  • Biomarker Identification: Correlating CXCR4/ACKR3 expression levels with treatment response .

  • Combination Therapies: Potential synergy with PD-1/CTLA-4 inhibitors in cancer .

  • Long-Term Safety: Ongoing studies to assess immune tolerance and off-target risks .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M Phosphate Buffered Saline (PBS), pH 7.4
Form
Liquid
Lead Time
Made-to-order (14-16 weeks)
Synonyms
CXE12 antibody; At3g48690 antibody; T8P19.200Probable carboxylesterase 12 antibody; AtCXE12 antibody; EC 3.1.1.1 antibody
Target Names
CXE12
Uniprot No.

Target Background

Function
CXE12 Antibody is a carboxylesterase that exhibits activity on esters with varying acyl chain lengths.
Database Links

KEGG: ath:AT3G48690

STRING: 3702.AT3G48690.1

UniGene: At.24698

Protein Families
'GDXG' lipolytic enzyme family
Tissue Specificity
Expressed in leaves, stems, flowers and siliques.

Q&A

What is CXCL12 and how do CXCL12 antibodies function?

CXCL12 (also known as stromal cell-derived factor 1 or SDF-1) is a CXC chemokine traditionally classified as homeostatic, contributing to physiological processes such as embryogenesis, hematopoiesis, and angiogenesis. CXCL12 primarily signals through the CXCR4 receptor and can also interact with ACKR3 (formerly CXCR7). In pathological conditions, increased expression of CXCL12 or specific CXCL12 splicing variants has been observed across various diseases . CXCL12 antibodies function by neutralizing CXCL12, preventing its interaction with receptors, thereby blocking downstream signaling cascades that mediate inflammatory responses, cell migration, and immune cell activation .

How does one evaluate the specificity of a CXCL12 antibody?

To evaluate CXCL12 antibody specificity, researchers should employ multiple complementary approaches:

  • Direct binding assays: Assess binding affinity (KD values) through surface plasmon resonance (SPR), enzyme-linked immunosorbent assay (ELISA), or yeast surface display methods.

  • Cross-reactivity testing: Examine binding to related chemokines, particularly other CXC family members.

  • Functional neutralization assays: Measure the antibody's ability to block CXCL12-mediated cellular responses, such as calcium mobilization in neutrophils.

  • Epitope mapping: Determine the specific region of CXCL12 recognized by the antibody using techniques like hydrogen-deuterium exchange mass spectrometry.

For example, researchers evaluating crossreactive antibodies have employed two complementary configurations: soluble CXC chemokines with yeast-displayed antibodies and soluble antibodies with yeast-displayed CXC chemokines to comprehensively characterize binding properties .

What are the primary research applications for CXCL12 antibodies?

CXCL12 antibodies are valuable research tools for:

  • Disease modeling: Investigating the role of CXCL12/CXCR4 signaling in preclinical models of inflammatory and autoimmune conditions.

  • Immunomodulation studies: Examining how CXCL12 neutralization affects immune cell recruitment, activation, and function.

  • Mechanistic investigations: Elucidating molecular pathways downstream of CXCL12/CXCR4 signaling.

  • Therapeutic development: Serving as lead compounds for developing novel targeted therapies.

Recent research has demonstrated that administration of CXCL12-neutralizing antibodies delays disease onset or prevents disease progression in diverse conditions including cancer, viral infections, inflammatory bowel diseases, rheumatoid arthritis, osteoarthritis, and alopecia areata .

How should researchers design experiments to evaluate CXCL12 antibody efficacy in vitro?

A comprehensive in vitro evaluation protocol for CXCL12 antibody efficacy should include:

  • Binding characterization:

    • Determine binding affinity (KD) using SPR or other quantitative techniques

    • Assess binding kinetics (kon and koff rates)

    • Evaluate pH and temperature dependence of binding

  • Functional assays:

    • Chemotaxis inhibition assays using primary neutrophils or relevant immune cells

    • Calcium mobilization assays to assess CXCR4 signaling blockade

    • Cell proliferation/survival assays in CXCL12-dependent cell lines

  • Cellular mechanism studies:

    • Analysis of downstream signaling pathway inhibition (JAK/STAT, MAPK)

    • Receptor internalization assays

    • Competition assays with labeled CXCL12

These approaches should include appropriate controls, including isotype control antibodies and cells lacking CXCR4/CXCR7 expression .

What are the optimal methods for humanizing CXCL12 antibodies for therapeutic development?

Humanization of CXCL12 antibodies typically follows these methodological steps:

  • CDR grafting: Transfer of complementarity-determining regions (CDRs) from a non-human (typically mouse) antibody to a human antibody framework.

  • Framework back-mutations: Identification and reversion of key framework residues that support CDR conformation to maintain binding affinity.

  • Affinity maturation:

    • Error-prone PCR to introduce genetic diversity

    • Phage or yeast display selections using decreasing concentrations of target

    • Sequential screening against multiple chemokines to maintain crossreactivity

  • Biophysical optimization:

    • Thermal stability assessment (Tm values)

    • Aggregation propensity analysis

    • Long-term stability studies

Recent research successfully developed humanized CXCL12 antibodies that retained therapeutic efficacy in alopecia areata models while minimizing immunogenicity potential .

How can single-cell RNA sequencing be utilized to evaluate CXCL12 antibody mechanisms of action?

Single-cell RNA sequencing (scRNA-seq) provides a powerful approach to delineate the cellular and molecular mechanisms of CXCL12 antibody action:

Methodology workflow:

  • Treat relevant disease models with CXCL12 antibody and appropriate controls

  • Isolate tissues of interest (e.g., skin in alopecia models)

  • Prepare single-cell suspensions and perform scRNA-seq

  • Analyze data using:

    • Cell clustering and annotation

    • Differential gene expression analysis

    • Pseudotime trajectory analysis

    • Cell-cell interaction modeling

    • Receptor-ligand pair identification

Key insights obtainable:

  • Cell type-specific responses to CXCL12 antibody treatment

  • Temporal dynamics of gene expression changes

  • Identification of key mediators and pathways affected

  • Characterization of immune cell recruitment and activation states

Recent research employed scRNA-seq to demonstrate that CXCL12 antibody treatment in an alopecia areata model decreased the proportion of T cells (from 4.2% to 2.5%) and dendritic cells/macrophages (from 1.2% to 0.9%), identifying 153 differentially expressed genes associated with treatment response .

What approaches can be used to develop CXCL12 antibodies with enhanced crossreactivity while maintaining high affinity?

Developing CXCL12 antibodies with both high affinity and broad crossreactivity requires sophisticated engineering approaches:

  • Co-evolutionary selection strategy:

    • Create genetic diversity through error-prone PCR

    • Implement equilibrium-based selection using decreasing concentrations of targets

    • Employ combinatorial exposure to multiple chemokines

    • Screen in sequence from lowest to highest affinity chemokines

  • Structural biology-guided engineering:

    • Crystallize antibody-antigen complexes to identify key contact residues

    • Design mutations that enhance conserved epitope recognition

    • Employ computational modeling to predict crossreactive binding modes

  • Combinatorial library screening:

    • Generate libraries of 10^11 members or larger

    • Use yeast or phage display technologies

    • Implement negative selection to remove unwanted specificities

This approach has successfully yielded antibodies with improved crossreactivity and affinity. For example, the engineered CK138 clone recognized double the number of chemokines (from three to six) and achieved roughly a 30- to 340-fold improvement in affinity (KD values ranging from 5.8 to 193 nM) relative to its parental clone .

How does the mechanism of CXCL12 antibody treatment differ from direct CXCR4 antagonism in autoimmune conditions?

The mechanistic differences between CXCL12 neutralization and direct CXCR4 antagonism are significant:

ParameterCXCL12 AntibodyCXCR4 Antagonist
TargetSoluble ligand (CXCL12)Membrane receptor (CXCR4)
Effect on alternative ligandsNo direct effect on other CXCR4 ligandsBlocks all ligand interactions
Impact on ACKR3 signalingReduces CXCL12-ACKR3 signalingNo direct effect on ACKR3
Tissue penetrationGenerally good for soluble targetsVaries based on compound properties
Immune cell targetingAffects cells responding to CXCL12Affects all CXCR4-expressing cells
Off-target concernsGenerally fewerMay affect essential CXCR4 functions

Gene ontology analysis of CXCL12 antibody treatment in alopecia areata revealed specific downregulation of immune cell chemotaxis and cellular response to type II interferon pathways, with CXCL12 antibody selectively reducing activation of CD8+ T cells via the JAK/STAT pathway . This targeted modulation contrasts with the broader effects of direct CXCR4 antagonism.

What are the key considerations for analyzing and interpreting transcriptional changes following CXCL12 antibody treatment?

Analyzing transcriptional responses to CXCL12 antibody treatment requires careful consideration of several factors:

  • Distinguishing direct vs. indirect effects:

    • Identify genes directly regulated by CXCL12/CXCR4 signaling

    • Map secondary response genes activated downstream of primary targets

    • Consider temporal dynamics of gene expression changes

  • Cell type-specific analysis:

    • Perform cell type-specific differential expression analysis

    • Consider relative proportions of cell populations and their changes

    • Identify cell types most responsive to treatment

  • Pathway integration:

    • Conduct pathway and network analyses to identify key biological processes

    • Distinguish disease-related vs. treatment-specific pathways

    • Evaluate interaction between CXCL12/CXCR4 and other signaling networks

  • Validation approaches:

    • Confirm key findings with orthogonal methods (qPCR, protein analysis)

    • Perform functional studies of identified targets

    • Evaluate consistency across experimental models

Recent research using pseudobulk RNA sequencing of an alopecia model identified 153 differentially expressed genes that were upregulated in disease and downregulated after antibody treatment. STRING network analysis grouped these into three major clusters associated with immune cell chemotaxis, cytokine response pathways, and complement system functions .

How does CXCL12 antibody treatment modulate immune responses in alopecia areata?

CXCL12 antibody treatment exerts multifaceted effects on immune responses in alopecia areata:

  • Immune cell population changes:

    • Decreases T cell infiltration (reduced from 4.2% to 2.5% of total cells)

    • Reduces dendritic cell/macrophage populations (reduced from 1.2% to 0.9%)

    • Modulates the activation state of residual immune cells

  • Molecular pathway modulation:

    • Downregulates genes involved in immune cell chemotaxis

    • Reduces cellular responses to type II interferon

    • Affects key immune cell-related genes including Ifng, Cd8a, Ccr5, Ccl4, Ccl5, and Il21r

  • CD8+ T cell-specific effects:

    • Significantly decreases CD8+ T cell activation

    • Inhibits JAK/STAT pathway signaling in these cells

    • Reduces coexpression of Cxcr4 and Ifng (from 9.5% in disease to 1.3% after treatment)

  • Disease progression impact:

    • Delays onset of alopecia areata in mouse models

    • Normalizes approximately 78% of disease-associated transcriptional alterations

    • Shows minimal off-target effects unrelated to disease treatment

What is the evidence supporting epitope-guided antibody selection for targeting chemokine receptors versus ligands?

Epitope-guided antibody selection offers distinct advantages when targeting chemokine receptors:

  • Selectivity advantages:

    • Allows targeting of specific receptor domains involved in ligand binding

    • Can distinguish between closely related receptor family members

    • Enables modulation of specific signaling pathways while preserving others

  • Mechanistic benefits:

    • Directly blocks ligand-receptor interactions at the cell surface

    • Can target regions of receptors involved in specific functions

    • Potentially modulates receptor conformation and basal signaling

  • Research evidence:

    • Antibodies targeting the N-terminal region of CXCR2 (part of the IL-8 epitope) demonstrated high selectivity and tight binding

    • Such antibodies strongly inhibited IL-8-induced and CXCR2-mediated neutrophil chemotaxis

    • Receptor-targeting antibodies alleviated experimental autoimmune encephalomyelitis symptoms in mice

This receptor-targeting approach contrasts with ligand neutralization strategies by addressing the signal-receiving rather than signal-producing component, which may be advantageous when multiple ligands activate the same receptor or when targeting tissue-specific receptor variants .

How can researchers conduct comparative analyses of CXCL12 antibody efficacy across different disease models?

To effectively compare CXCL12 antibody efficacy across disease models, researchers should implement a structured analytical framework:

  • Standardized dosing and administration:

    • Establish pharmacokinetic/pharmacodynamic relationships across models

    • Normalize dosages based on target engagement metrics

    • Use consistent administration routes and schedules

  • Cross-disease biomarker panel:

    • Identify common mechanisms (e.g., T cell infiltration, cytokine profiles)

    • Develop a core set of cellular and molecular measurements

    • Include disease-specific metrics alongside universal parameters

  • Quantitative comparison methodology:

    • Calculate effect sizes relative to disease severity

    • Determine EC50 values for key biological processes

    • Employ multivariate analyses to identify determinants of response

  • Predictive biomarker identification:

    • Correlate baseline parameters with treatment response

    • Perform early on-treatment measurements to predict outcomes

    • Identify molecular signatures associated with efficacy

Research has demonstrated CXCL12 antibody efficacy across multiple conditions including alopecia areata, cancer, viral infections, inflammatory bowel diseases, rheumatoid arthritis, and osteoarthritis, suggesting common mechanisms relating to immune cell migration and activation .

How can researchers address variability in CXCL12 antibody neutralization assays?

Variability in CXCL12 neutralization assays can be addressed through systematic methodology optimization:

  • Source considerations:

    • Use consistent cell sources for functional assays (e.g., primary neutrophils)

    • Standardize isolation procedures and confirm cell viability

    • Consider donor variability in primary cell assays

  • Assay standardization:

    • Establish precise concentration ranges for CXCL12 stimulus

    • Validate CXCL12 activity before each experiment

    • Include internal standards and calibrators in each assay

  • Technical refinements:

    • Optimize incubation times and temperatures

    • Control for CXCL12 oligomerization effects

    • Consider matrix effects in complex biological samples

  • Advanced analytical approaches:

    • Employ full dose-response curves rather than single-point measurements

    • Calculate IC50 values with appropriate statistical models

    • Use area-under-curve analyses for time-course experiments

Researchers have found that exploring dual configurations (soluble chemokines with displayed antibodies and soluble antibodies with displayed chemokines) helps address variability issues related to chemokine oligomerization .

What strategies can optimize the translation of findings from mouse models to human applications of CXCL12 antibodies?

Optimizing translational relevance requires addressing species differences systematically:

  • Antibody engineering approaches:

    • Develop antibodies with cross-species reactivity where possible

    • Create species-specific antibodies with comparable epitope targeting

    • Consider humanized mouse models expressing human CXCL12/CXCR4

  • Comparative biology assessments:

    • Characterize species differences in CXCL12 expression patterns

    • Map variances in downstream signaling pathways

    • Identify conserved vs. divergent disease mechanisms

  • Translational model selection:

    • Use humanized mouse models where appropriate

    • Consider ex vivo human tissue assays as complementary approaches

    • Develop organoid or tissue-on-chip models incorporating human cells

  • Biomarker harmonization:

    • Identify translatable biomarkers of target engagement

    • Develop assays applicable to both preclinical and clinical samples

    • Establish quantitative relationships between animal and human metrics

Successful translation has been demonstrated with humanized CXCL12 antibodies that maintain therapeutic efficacy while minimizing immunogenicity, suggesting the core mechanisms are conserved across species .

How should researchers interpret contradictory results between in vitro and in vivo CXCL12 antibody studies?

Resolving contradictions between in vitro and in vivo findings requires systematic analysis:

  • Pharmacological considerations:

    • Assess antibody bioavailability and tissue penetration in vivo

    • Consider half-life and clearance mechanisms

    • Examine potential neutralization by anti-drug antibodies

  • Biological complexity factors:

    • Evaluate compensatory mechanisms active in vivo but not in vitro

    • Consider cell-cell interactions present only in intact tissues

    • Assess contributions of tissue microenvironment to response differences

  • Experimental design reconciliation:

    • Match concentrations/doses between systems when possible

    • Develop ex vivo models that bridge the complexity gap

    • Use systems biology approaches to identify missing components

  • Integration framework:

    • Develop quantitative models incorporating both datasets

    • Identify parameters explaining divergent results

    • Design targeted experiments to test specific hypotheses about discrepancies

When addressing contradictions, researchers should consider the complex interplay between different immune cell populations observed in vivo. For example, in alopecia models, CXCL12 antibody treatment affected multiple cell types simultaneously, with interactions that would not be captured in simplified in vitro systems .

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.