CML12 Antibody

Shipped with Ice Packs
In Stock

Description

CXCL12 Antibodies: Key Features and Applications

CXCL12 (also known as stromal cell-derived factor 1, SDF-1) is a chemokine involved in immune cell trafficking, cancer metastasis, and autoimmune disorders. Antibodies targeting CXCL12 are designed to modulate its interaction with receptors CXCR4 and ACKR3, which are critical in pathological processes like cancer progression and alopecia areata (AA) .

Mechanisms of Action

  • Immune Modulation: CXCL12-neutralizing antibodies reduce autoimmune attacks by inhibiting T-cell chemotaxis. In AA models, humanized CXCL12 antibodies decreased skin-infiltrating CD8+ T cells and dendritic cells, delaying hair loss .

  • Cancer Therapy: CXCL12/CXCR4 axis blockade inhibits tumor-stromal interactions, reducing metastasis and enhancing immunotherapy efficacy .

Table 1: Functional Effects of CXCL12 Antibodies

MechanismObserved OutcomeSource
CD8+ T-cell reductionDelayed AA onset in mice
Dendritic cell suppressionAttenuated autoimmune inflammation
Synergy with anti-PD-L1Enhanced glioblastoma tumor regression

CCL12 Antibodies in Preclinical Research

CCL12 (Monocyte Chemoattractant Protein-5) is a murine chemokine analog of human CCL2. Antibodies targeting CCL12 are used to study inflammatory diseases and cancer.

Key Applications

  • Chemotaxis Inhibition: Rat anti-mouse CCL12 antibodies (e.g., MAB428) neutralize CCL12-induced migration of CCR2A-expressing cells, with an ND50 of 0.5–2.5 µg/mL .

  • Inflammatory Models: Used to dissect pathways in fibrosis, atherosclerosis, and tumor-associated macrophage recruitment.

CXCL12 Antibodies

ParameterDetailsSource
Target EpitopeC-terminal region (AA 64–92)
ReactivityHuman, Mouse, Rat
ApplicationsWB, IHC, IF, ELISA
Commercial ExamplesOAAB09710 (Aviva Systems Biology), ABIN6139271 (Antibodies-Online)

CCL12 Antibodies

ParameterDetailsSource
Clone63205
Host SpeciesRat
NeutralizationBlocks BaF3 cell migration (80% efficacy at 10 µg/mL)

CXCL12 in Autoimmunity

  • In AA mice, CXCL12 antibody treatment downregulated genes linked to T-cell activation (Ifng, Cd8a, Ccl5) and reduced interferon-γ pathways .

  • Single-cell RNA sequencing revealed suppressed lymphocyte chemotaxis (GO:0048247) and type II interferon responses (GO:0034341) .

CXCL12 in Cancer

  • Glioblastoma models showed that CXCR7 agonists (e.g., VUF11207) synergize with anti-PD-L1 antibodies, improving CD8+ T-cell infiltration and survival .

  • In breast cancer, CA12 siRNA reversed paclitaxel resistance by modulating mitochondrial apoptosis pathways (Bax, Bcl-2) .

Potential Misinterpretation: CML12 DIRECT Trial

The term "CML12" appears in a phase II trial (CML12 DIRECT) investigating dasatinib dosing in chronic myeloid leukemia (CML) . This study does not involve antibody therapies but focuses on tyrosine kinase inhibitor optimization.

Challenges and Future Directions

  • Off-Target Effects: CXCL12 antibodies may transiently alter calcium signaling and cell-matrix adhesion pathways .

  • Delivery Optimization: Antibody-drug conjugates (ADCs) require mg/kg dosing parity between preclinical models and humans to ensure tissue penetration .

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
CML12 antibody; CAL4 antibody; TCH3 antibody; At2g41100 antibody; T3K9.13Calmodulin-like protein 12 antibody; Touch-induced calmodulin-related protein 3 antibody
Target Names
CML12
Uniprot No.

Target Background

Function
CML12 Antibody is a potential calcium sensor that exhibits calcium binding in vitro.
Database Links

KEGG: ath:AT2G41100

STRING: 3702.AT2G41100.1

UniGene: At.12547

Protein Families
Calmodulin family

Q&A

What is CXCL12 and why is it relevant for antibody development?

CXCL12 is a CXC chemokine traditionally classified as a homeostatic chemokine that contributes to physiological processes such as embryogenesis, hematopoiesis, and angiogenesis . Despite these homeostatic functions, increased expression of CXCL12 (or specific CXCL12 splicing variants) has been demonstrated in various pathologies . CXCL12 is highly expressed in dermal fibroblasts and mediates inflammatory processes in the skin, making it a valuable target for therapeutic antibody development . The CXCL12/CXCR4/ACKR3 axis constitutes a potential therapeutic target for a wide variety of inflammatory diseases, not only by interfering with cell migration but also by modulating immune responses .

How does the humanization process affect CXCL12 antibody function?

Antibody humanization is critical for reducing immunogenicity while maintaining therapeutic efficacy. For CXCL12 antibodies, the humanization process involves genetic engineering to replace non-human regions with human antibody sequences while preserving the antigen-binding domains. Research demonstrates that humanized CXCL12 antibodies retain their ability to effectively neutralize CXCL12 in vivo . When tested in mouse models of alopecia areata, subcutaneous injection of humanized CXCL12 antibody significantly delayed disease onset, confirming that the humanization process preserved the antibody's therapeutic function . Researchers should verify preservation of binding affinity and neutralizing capability through both in vitro and in vivo experiments following humanization.

What experimental controls are essential when testing CXCL12 antibody efficacy?

When designing experiments to test CXCL12 antibody efficacy, researchers should include:

  • Negative controls (untreated subjects) to establish baseline conditions without intervention

  • Disease model controls (subjects with the pathology but no antibody treatment)

  • Antibody treatment groups with appropriate dosing schedules

  • Isotype control antibodies to control for non-specific effects of antibody administration

This approach is exemplified in studies where researchers used three experimental groups: negative controls (Neg), AA model subjects (AA), and antibody-treated AA model subjects (AA + Ab) . These controls allow researchers to distinguish between disease-specific effects and antibody-mediated responses.

How should researchers design single-cell RNA sequencing studies to evaluate CXCL12 antibody effects?

Single-cell RNA sequencing (scRNA-seq) provides critical insights into cellular responses to CXCL12 antibody treatment. An effective experimental design requires:

  • Collection of tissue samples from control and experimental groups at appropriate timepoints

  • Gentle tissue dissociation to maintain cell viability and RNA integrity

  • Cell sorting and library preparation optimized for immune cell capture

  • Sequencing depth sufficient for detecting low-abundance transcripts (minimum 50,000 reads per cell)

  • Computational pipeline including quality control, normalization, clustering, and differential expression analysis

In CXCL12 antibody research, this approach successfully identified changes in immune cell populations and gene expression profiles following treatment . Researchers identified cell types based on marker gene expression (e.g., T cells expressing Cd3e, dendritic cells/macrophages expressing Cd68, Cd74, and Cd209a) . This detailed cellular characterization enabled identification of specific immune cell responses to antibody treatment.

What bioinformatics approaches are most effective for analyzing transcriptomic data from CXCL12 antibody experiments?

When analyzing transcriptomic data from CXCL12 antibody experiments, researchers should implement:

  • Differential Expression Analysis: Compare gene expression between experimental groups using robust statistical methods (e.g., DESeq2 or edgeR) with appropriate cutoffs (typically >2-fold change, adjusted p-value <0.05) .

  • Pseudobulk RNA-seq Analysis: Aggregate transcript counts from single cells for each experimental group to increase statistical power for detecting changes across conditions .

  • Protein-Protein Interaction Networks: Use tools like STRING to organize differentially expressed genes based on known interactions, facilitating identification of functional clusters .

  • Gene Ontology (GO) Enrichment Analysis: Determine which biological processes are significantly affected by antibody treatment .

  • Gene Set Enrichment Analysis (GSEA): Evaluate changes in predefined gene sets representing specific pathways or biological processes .

This methodological approach identified 153 differentially expressed genes that were upregulated in an AA model and downregulated after antibody treatment, revealing key mediators of both disease pathogenesis and therapeutic response .

How can researchers effectively compare CXCL12 antibody effects across different immune cell populations?

To comprehensively assess CXCL12 antibody effects across immune cell populations:

  • Cell Type Annotation and Quantification: Use canonical marker genes to identify distinct immune cell populations through clustering analysis of scRNA-seq data .

  • Proportional Analysis: Calculate the proportion of each cell type among total cells for each experimental group and assess statistical significance using appropriate tests (e.g., binomial test) .

  • Subclustering Analysis: Perform secondary clustering within major cell types to identify functional subpopulations .

  • Gene Expression Profiling: Analyze expression of key functional genes within each cell type to assess activation state .

  • Receptor-Ligand Interaction Analysis: Evaluate expression of CXCL12 receptors (CXCR4, ACKR3) in relation to response genes .

Using this approach, researchers identified significant changes in T cell and dendritic cell/macrophage populations following CXCL12 antibody treatment, with proportions decreasing from 4.2% to 2.5% for T cells and from 1.2% to 0.9% for dendritic cells/macrophages after treatment .

How does CXCL12 antibody modulate T cell function in autoimmune conditions?

CXCL12 antibody demonstrates sophisticated modulation of T cell function in autoimmune conditions through multiple mechanisms:

  • Reduction of T Cell Recruitment: By neutralizing CXCL12, the antibody disrupts chemokine gradients that normally attract T cells to inflammatory sites, as evidenced by decreased T cell proportions in skin tissue following antibody treatment .

  • Alteration of T Cell Activation: The antibody treatment suppresses expression of genes involved in T cell activation pathways, although this effect shows a trend toward suppression rather than significant downregulation .

  • Inhibition of CD8+ T Cell Effector Functions: CD8+ T cells, which play a central role in autoimmune attack in conditions like alopecia areata, show reduced activation following antibody treatment, potentially through inactivation of the Jak/Stat pathway .

  • Regulation of Key Immune-Related Genes: Treatment with CXCL12 antibody downregulates genes critical for T cell function including Ifng, Cd8a, Ccr5, Ccl4, Ccl5, and Il21r, which are colocalized with Cxcr4 in T cells .

These mechanisms collectively contribute to the immunomodulatory effects of CXCL12 antibody in autoimmune conditions, particularly those mediated by T cell responses.

What molecular pathways are directly and indirectly affected by CXCL12 antibody treatment?

CXCL12 antibody treatment affects multiple molecular pathways, with differential impacts on direct and indirect targets:

Directly Affected Pathways:

  • Immune Cell Chemotaxis: Pathways related to lymphocyte and monocyte chemotaxis are significantly downregulated following antibody treatment .

  • Chemokine-Mediated Signaling: The antibody disrupts signaling cascades initiated by chemokine-receptor interactions .

  • Cellular Response to Type II Interferon: Gene set enrichment analysis identified significant suppression of interferon response pathways following antibody treatment .

Indirectly Affected Pathways:

  • Leukocyte Differentiation: Pathways regulating immune cell differentiation show altered activity, potentially as a secondary effect of chemokine signaling disruption .

  • Cytokine Response Networks: Both type I and type II interferon response pathways show modulated activity, indicating broad effects on cytokine networks .

  • Complement System: Genes linked to complement function show altered expression, particularly within dendritic cells and macrophages .

Interestingly, the antibody induced relatively few significant changes in biological processes unrelated to immune function, suggesting minimal off-target effects .

How can researchers distinguish between on-target and off-target effects of CXCL12 antibody treatment?

Distinguishing between on-target and off-target effects requires rigorous analytical approaches:

  • Pathway-Specific Analysis: Compare differentially expressed genes (DEGs) that represent the primary therapeutic target versus those potentially indicating off-target effects .

  • Quantitative Comparison: Assess the number of biological processes affected by common DEGs versus antibody-specific DEGs; research shows that common DEGs were associated with approximately 30 biological processes, while antibody-specific DEGs affected only 5-7 processes despite similar numbers of DEGs .

  • Temporal Analysis: Evaluate the kinetics of gene expression changes, as on-target effects often follow specific temporal patterns.

  • Dose-Response Relationships: Test multiple antibody concentrations to identify threshold effects that may separate on-target from off-target responses.

  • Receptor Specificity Controls: Use cells lacking CXCL12 receptors to identify effects that occur independent of target engagement.

Research demonstrates that CXCL12 antibody treatment shows high specificity with limited off-target effects, though it may increase expression of genes involved in TLR receptor pathways (potentially an immunomodulatory effect) and reduce expression of genes related to calcium transport, muscle contraction, and cell-matrix adhesion .

What are the optimal protocols for testing CXCL12 antibody efficacy in animal models?

Effective protocols for testing CXCL12 antibody efficacy in animal models should include:

  • Disease Model Selection: Choose models that recapitulate key features of the target disease. For alopecia areata research, lymph node cell injection models effectively induce disease with relevant immune responses .

  • Administration Route and Schedule: Subcutaneous injection has proven effective for CXCL12 antibody delivery, allowing for local effects in skin conditions . Researchers should establish appropriate dosing schedules based on antibody pharmacokinetics.

  • Endpoint Measurements:

    • Clinical assessment of disease progression (e.g., hair loss onset and severity)

    • Histological evaluation of affected tissues

    • Flow cytometric analysis of immune cell infiltration

    • Molecular analysis of target pathways using techniques like scRNA-seq

  • Controls and Blinding: Include appropriate controls (as discussed in question 1.3) and implement blinding procedures to minimize bias in outcome assessments.

  • Statistical Considerations: Determine sample sizes through power analysis and use appropriate statistical tests for data evaluation, such as the binomial test for cell proportion comparisons .

This systematic approach allows for comprehensive evaluation of CXCL12 antibody efficacy in preclinical models.

What methodologies are available for assessing CXCL12 antibody effects in human samples or clinical studies?

For human studies, several methodologies can assess CXCL12 antibody effects:

  • Ex Vivo Tissue Analysis:

    • Skin biopsies or other relevant tissues can be collected before and after antibody administration

    • Immunohistochemistry to evaluate changes in immune cell infiltration

    • Single-cell analysis techniques to characterize cellular responses at high resolution

  • Circulating Biomarkers:

    • Measure serum levels of inflammatory cytokines

    • Evaluate circulating immune cell populations via flow cytometry

    • Assess CXCL12 levels to confirm target engagement

  • Functional Assays:

    • Ex vivo stimulation of patient samples to assess immune responsiveness

    • Chemotaxis assays to evaluate immune cell migration capacity

  • Molecular Response Assessment:

    • Similar to techniques used in CML patients, standardized molecular response criteria can help quantify treatment effects

    • Categorize responses using established frameworks (e.g., MR4.0) to track changes over time

  • Longitudinal Design:

    • Sample at multiple timepoints (pre-treatment, early post-treatment, late post-treatment)

    • Compare with control subjects or standard-of-care treatments

These approaches can be tailored to specific clinical contexts, providing comprehensive assessment of CXCL12 antibody effects in human subjects.

How might CXCL12 antibody research intersect with other immunomodulatory approaches?

CXCL12 antibody research presents several opportunities for integration with complementary immunomodulatory approaches:

  • Combination with Small Molecule Inhibitors: Combining CXCL12 antibodies with small molecule antagonists of CXCR4 (like AMD3100) may provide synergistic effects by targeting both the ligand and receptor .

  • Integration with Targeted Immunotherapies: Exploring combinations with agents that target other aspects of immune dysfunction, similar to how anti-CCL2 antibody has been combined with chemotherapeutic agents like etoposide .

  • Vaccine Response Modulation: Investigating how CXCL12 pathway modulation affects vaccine responses, particularly in patients with conditions like CML who may have altered immune function .

  • JAK-STAT Pathway Integration: Since CXCL12 antibody treatment appears to inactivate the JAK-STAT pathway in CD8+ T cells , exploring combination with JAK inhibitors might yield enhanced therapeutic effects in autoimmune conditions.

  • TLR Pathway Modulation: Given the observed effects on TLR receptor pathways , investigating combinations with TLR agonists or antagonists could yield novel therapeutic approaches.

Future research should systematically evaluate these combinatorial approaches to identify optimal strategies for specific disease contexts.

What are the key considerations for translating CXCL12 antibody research from animal models to clinical applications?

Translating CXCL12 antibody research to clinical applications requires careful consideration of:

  • Species-Specific Differences: Human and mouse CXCL12 signaling pathways may differ in key aspects, necessitating validation in human systems before clinical testing.

  • Safety Profiling: Thoroughly evaluate potential off-target effects identified in preclinical studies, particularly those affecting calcium transport, muscle contraction, and cell-matrix adhesion .

  • Dosing Optimization: Establish optimal dosing regimens based on pharmacokinetic/pharmacodynamic modeling that accounts for differences between preclinical models and humans.

  • Biomarker Development: Identify reliable biomarkers of target engagement and therapeutic response to guide clinical development.

  • Patient Selection: Define appropriate patient populations based on disease mechanisms and expression patterns of CXCL12 and its receptors.

  • Long-term Effects: Design studies to assess long-term effects, as immunomodulatory treatments may have delayed consequences on immune surveillance and response.

  • Route of Administration: Determine optimal delivery methods based on target tissue and systemic vs. local effects desired.

Addressing these considerations systematically will facilitate successful translation of CXCL12 antibody therapies to clinical applications.

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.