CX3CR1 Antibody

Shipped with Ice Packs
In Stock

Description

CX33CR1 Antibody Overview

CX3CR1 antibodies bind to the transmembrane chemokine receptor CX3CR1, which interacts exclusively with fractalkine (CX3CL1). This receptor is expressed on immune cells (monocytes, NK cells, microglia) and certain cancer cells . Antibodies targeting CX3CR1 are designed to:

  • Block ligand-receptor interactions (e.g., inhibiting CX3CL1-mediated chemotaxis)

  • Modulate immune signaling (e.g., reducing NLRP3 inflammasome activity)

  • Facilitate diagnostic detection (e.g., flow cytometry, immunohistochemistry)

Cancer Immunotherapy

  • Tumor Migration Inhibition: CRISPR deletion or monoclonal antibody blockade of CX3CR1 reduces metastasis in pancreatic, breast, and colorectal cancers by >50% .

  • Combination Therapy: Anti-CX3CR1 antibodies enhance anti-PD-1 efficacy, increasing survival in CT26 colon carcinoma models by remodeling myeloid compartments (↓MDSC, ↑mature macrophages) .

  • Immune Suppression Reversal: CX3CR1 blockade decreases secretion of CXCL1, IL-6, and CCL5, reversing tumor-mediated immunosuppression .

Neuroinflammation Studies

  • Microglial Activity: Anti-CX3CR1 antibodies (e.g., ACR-058) label microglia in rat hippocampus, enabling studies on synaptic pruning and neurodegenerative diseases .

  • HIV Neuropathology: CX3CR1 serves as an HIV-1 co-receptor; antibodies help map viral entry mechanisms in microglia .

Flow Cytometry Profiling

  • Immune Cell Subsets: Clone 2A9-1 identifies CX3CR1+ CD8+ T cells and CD16+ NK cells in human blood, correlating with cytotoxic potential (perforin/granzyme B expression) .

Mechanistic Insights from Preclinical Studies

  • Signaling Pathways: CX3CR1 antibodies disrupt GPCR-mediated activation of PI3K/AKT, MAPK, and NLRP3 pathways, reducing tumor survival and inflammasome activity .

  • Myeloid Recruitment: In melanoma models, CX3CR1+ tumor cells secrete chemoattractants (e.g., CXCL1, G-CSF), recruiting immunosuppressive MDSCs; antibody treatment reduces MDSC infiltration by 40% .

Challenges and Future Directions

  • Specificity Issues: Small-molecule CX3CR1 antagonists may cross-react with other GPCRs, whereas monoclonal antibodies offer superior selectivity .

  • Therapeutic Potential: Phase I trials are exploring CX3CR1 antibodies in combination with checkpoint inhibitors for solid tumors .

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 orders within 1-3 business days of receiving them. Delivery time may vary depending on the purchasing method or location. Please consult your local distributors for specific delivery time estimates.
Synonyms
CX3CR1; CMKBRL1; GPR13; CX3C chemokine receptor 1; C-X3-C CKR-1; CX3CR1; Beta chemokine receptor-like 1; CMK-BRL-1; CMK-BRL1; Fractalkine receptor; G-protein coupled receptor 13; V28
Target Names
Uniprot No.

Target Background

Function
CX3CR1 is a receptor for the chemokine fractalkine (CX3CL1), found on many early leukocyte cells. CX3CR1-CX3CL1 signaling plays diverse roles in different tissues, including immune response, inflammation, cell adhesion, and chemotaxis. CX3CR1-CX3CL1 signaling is crucial for cell migration, recruiting natural killer (NK) cells to inflamed tissues. It regulates inflammatory processes contributing to atherosclerosis by mediating macrophage and monocyte recruitment to inflamed atherosclerotic plaques and promoting cell survival. It is involved in airway inflammation by enhancing interleukin 2-producing T helper (Th2) cell survival in inflamed lungs. It participates in the migration of circulating monocytes to non-inflamed tissues, where they differentiate into macrophages and dendritic cells. It acts as a negative regulator of angiogenesis, likely by promoting macrophage chemotaxis. CX3CR1 plays a key role in brain microglia, regulating inflammatory responses in the central nervous system (CNS) and synapse maturation. It is required to restrain microglial inflammation in the CNS and prevent subsequent parenchymal damage in response to pathological stimuli. It is involved in brain development by participating in synaptic pruning, a natural process where brain microglia eliminate extra synapses during postnatal development. Synaptic pruning by microglia is necessary for the maturation of circuit connectivity during brain development. CX3CR1 acts as a significant regulator of the gut microbiota by controlling immunity to intestinal bacteria and fungi. It is expressed in lamina propria dendritic cells in the small intestine, which form transepithelial dendrites capable of absorbing bacteria to provide defense against pathogenic bacteria. CX3CR1 is essential for initiating innate and adaptive immune responses against the dissemination of commensal fungi (mycobiota) in the gut, expressed in mononuclear phagocytes (MNPs) and promoting the induction of antifungal IgG antibody responses to confer protection against disseminated C.albicans or C.auris infections. CX3CR1 also serves as a receptor for the C-C motif chemokine CCL26, inducing cell chemotaxis. It acts as a coreceptor with CD4 for the HIV-1 virus envelope protein, potentially having more potent HIV-1 coreceptor activity than isoform 1.
Gene References Into Functions
  1. A study demonstrated that CX3CL1/CX3CR1 was overexpressed in prostate cancer tissues with spinal metastasis compared to primary tumors. Overexpression of CX3CR1 increased cell proliferation, migration, and invasion. The study also observed that the EGFR/Src/FAK pathway was activated by CX3CL1/CX3CR1. PMID: 30066854
  2. CX3CR1 major allele carriers V249 and T280 are significantly associated with increased total arterial blood volume in the whole brain, particularly around the bilateral precuneus, left posterior cingulate cortex, and left posterior parietal cortex. PMID: 29485193
  3. A study investigated the association of CX3CR1 839C/T and CX3CR1 745G/A polymorphisms with Age-Related Macular Degeneration (AMD) risk. These polymorphisms were associated with an increased AMD risk (CX3CR1 839C/T, additive model: aOR=2.682, 95% CI=1.119-5.709, P=0.022, recessive model: aOR=2.729, 95% CI=1.141-6.048, P=0.010; CX3CR1 745G/A, additive model: aOR=2.614, 95% CI=1.231-6.012, P=0.020, recessive model: aOR=2.340, 95% CI=1.227-5.993, P=0.011). PMID: 29565837
  4. CX3CR1 regulated chondrocyte proliferation. PMID: 29217163
  5. A study detected a statistically significant association between the variant Ala55Thr in CX3CR1 with schizophrenia and autism spectrum disorder phenotypes. PMID: 28763059
  6. This study showed that CX3CR1 expression in both Microglia and Astrocytes in the hippocampus is affected by stroke, Alzheimer's disease, and Lewy body dementia. PMID: 28398520
  7. The US28 gene product has maintained the function of the ancestral gene and has the ability to bind and signal in response to human CX3CL1, the natural ligand for CX3CR1. PMID: 28315475
  8. Findings demonstrate that motility, invasion, and contact-independent growth of PDAC cells all increase following CX3CL1 exposure, and that antagonism of CX3CR1 by the inhibitor JMS-17-2 reduces each of these phenotypes and correlates with a downregulation of AKT phosphorylation. PMID: 29274778
  9. In Crohn's disease patients, a missense mutation in the gene encoding CX3CR1 was identified and found to be associated with impaired antifungal responses. PMID: 29326275
  10. Soluble FKN that was efficiently shed from the surface of LPS-activated ECs in response to binding of CD16(+) monocytes to ECs, diminished monocyte adhesion in down-regulating CX3CR1 expression on the surface of CD16(+) monocytes resulting in decreased TNF-secretion. PMID: 27031442
  11. CX3CR1 genetic variants were not associated with the risk of atherosclerotic coronary heart disease and glucometabolic traits in a European ancestry cohort. In a South Asian cohort, identified CX3CR1 SNP associated with myocardial infarction and type II diabetes mellitus. PMID: 27013693
  12. FKN and CX3CR1 expression was significantly increased in pancreatic ductal adenocarcinoma (PDAC) tissues, especially in the metastatic samples, and was highly-correlated with the severity of PDAC. Ectopic expression of FKN promoted the proliferation and migration of PDAC, while knockdown of CX3CR1 reversed the function of FKN. PMID: 28986258
  13. CX3CL1 is upregulated in both human and murine tumors following VEGF signaling blockade, resulting in the recruitment of CX3CR1+Ly6Clo monocytes into the tumor. PMID: 28691930
  14. The fractalkine functions on the activation of the AKT/NF-kappaB/p65 signaling cascade and regulation of the antiapoptosis process in pancreatic cancer cells. PMID: 28845524
  15. High expression of CX3CR1 correlates with significantly shorter survival, specifically in post-menopausal patients with advanced and terminal stages of the disease. Taken together, this supports a key regulatory role for the fractalkine axis in advanced and relapsed peritoneal metastasis in epithelial ovarian carcinoma. PMID: 27941884
  16. rs3732378 and rs3732379 susceptibility loci for developmental dysplasia of the hip. PMID: 27176135
  17. The V249I genotype of the fractalkine receptor showed a protective role in patients with type 2 diabetes. The T280M genotype is associated with increased carotid intima-media thickness in Mexican individuals with or without type 2 diabetes. PMID: 28128806
  18. CX3CR1 genetic variation shows a possible association with hypertension, diabetes mellitus, and atherosclerosis comorbidities in patients treated with hemodialysis. PMID: 27118566
  19. This study shows that the expression of CX3CR1 on tonsillar CD8-positive cells is higher in IgA nephropathy patients. PMID: 28196748
  20. Low CCRL1 expression is associated with hepatocellular carcinoma. PMID: 26813566
  21. Recent works show that, in allergic diseases, there is an increased expression of fractalkine/CX3CL1 and its unique receptor CX3CR1 and that this chemokine does not act as a chemoattractant. In allergic asthma, CX3CR1 expression regulates Th2 and Th1 cell survival in the inflammatory lung, while, in atopic dermatitis, it regulates Th2 and Th1 cell retention into the inflammatory site. [review] PMID: 27011244
  22. cxc3cr1 is a biomarker for Alzheimer's disease. PMID: 26567742
  23. CX3CR1 is expressed in the normal, cancer adjacent normal, inflammatory, and malignant fallopian epithelium. PMID: 26633537
  24. Genetic polymorphism is associated with delayed allograft function in Polish kidney transplant recipients. PMID: 25898802
  25. The CX3CR1 T allele of rs3732379 might have a positive association with the susceptibility of age-related macular degeneration. PMID: 26464724
  26. CX3CR1 is expressed differentially in human monocytes during differentiation. PMID: 25502213
  27. This meta-analysis suggested that CX3CR1 T280M and V249I polymorphisms may not be associated with an increased risk of AMD based on current published data. PMID: 26651305
  28. Data show that fractalkine receptor CX3CR1 expression is decreased in both murine and human glioblastoma (GBM) tissue. PMID: 25987130
  29. Respiratory syncytial virus G protein and host CX3CR1 interaction is important in infection and infection-induced responses of the airway epithelium. PMID: 26297201
  30. Association of hydrogen sulfide with alterations of monocyte chemokine receptors, CCR2 and CX3CR1 in patients with coronary artery disease. PMID: 26123579
  31. Specific pro-inflammatory monocyte subpopulations positive for CD16 and the co-expressed chemokine receptor, CX3CR1, are discriminative for chronic kidney disease stage 5 on hemodialysis patients. PMID: 25830914
  32. CX3CR1 is expressed in differentiated human ciliated airway cells and co-localizes with respiratory syncytial virus on cilia in a G protein-dependent manner. PMID: 26107373
  33. CX3CL1 and CX3CR1 may contribute to the formation of coronary atherosclerotic plaque in coronary artery disease. PMID: 25845619
  34. Fractalkine receptor polymorphisms may not contribute to the molecular pathogenesis of ulcerative colitis. PMID: 26042517
  35. The results from the present study support the concept of CX3CL1-mediated activation of the progression of multiple myeloma via CX3CR1. PMID: 25962684
  36. This is the first demonstration of the role of DNA methylation in regulating the expression of the CX3CR1 gene by CD8+ T cells and the potential biological relevance of increased expression of CX3CR1 by IL-7Ra(low) effector memory CD8+ T cells in humans. PMID: 26276874
  37. The study found a significantly lower expression of CX3CR1 on CD8+ T cells in the neovascular age-related macular degeneration group compared to the control group (p = 0.04). PMID: 25503251
  38. The high expression of CX3CR1 was associated with prolonged isolated thrombocytopenia after allogeneic hematopoietic stem cell transplantation. PMID: 26141368
  39. The progression rate of ALS symptoms and the survival time is affected in patients with one or two copies of the CX3CR1 249I allele. CX3CR1 is the most potent ALS survival genetic factor reported to date. PMID: 24806473
  40. An inverse pattern was observed in gene expression levels of fractalkine receptor (CX3CR1), which might be a compensatory mechanism. PMID: 24930044
  41. Findings suggest that overexpression of CX3CR1 promotes gastric cancer metastasis, proliferation, and survival and might play a physiological role in normal gastric tissue renewal and/or tissue remodeling after injury. PMID: 25482732
  42. Fractalkine and CX3CR1 may play a role in the pathogenesis of pSS, including extraglandular manifestations. PMID: 25320221
  43. Cx3cr1-deficient mononuclear phagocytes express increased P2X7 receptors, which stimulates IL-1Beta secretion. PMID: 25948251
  44. A higher percentage of circulating CD4(+) T-cells expressed CX3CR1 in relapsing-remitting multiple sclerosis compared to healthy controls. PMID: 25596452
  45. Data show that the chemokine domain of fractalkine (FKN-CD) can activate alphavbeta3 integrin in the absence of fractalkine receptor CX3CR1, but that this activation requires the direct binding of FKN-CD to alphavbeta3. PMID: 24789099
  46. These findings suggest that CCR2, CX3CR1, RANTES, and SDF1 gene variants seem to play an important role in the dynamics of HIV infection and could be used as drug or vaccine targets. PMID: 25313609
  47. Insulin resistance increases plaque vulnerability by augmenting the CX3CL1/CX3CR1 axis, which is mechanistically linked to reduced vascular smooth muscle cell survival. PMID: 24788416
  48. Our results confirm previous findings on the predominance of R5 tropism among HIV-1 treatment-naive subjects and reveal an association between a higher frequency of R5 tropism and the CX3CR1 A allele and a lower additive genetic score. PMID: 24750723
  49. CX3CR1 (T280M and V249I) and PLEKHA1 (A320T) polymorphisms were not found to be associated with age-related macular degeneration in an Indian population. PMID: 25050486
  50. Suggest that CCRL1 impairs chemotactic events associated with CCR7 in the progression and metastasis of hepatocellular carcinoma. PMID: 25255875

Show More

Hide All

Database Links

HGNC: 2558

OMIM: 601470

KEGG: hsa:1524

STRING: 9606.ENSP00000351059

UniGene: Hs.78913

Involvement In Disease
Macular degeneration, age-related, 12 (ARMD12)
Protein Families
G-protein coupled receptor 1 family
Subcellular Location
Cell membrane; Multi-pass membrane protein.
Tissue Specificity
Expressed in lymphoid and neural tissues. Expressed in lymphocyte subsets, such as natural killer (NK) cells, gamma-delta T-cells and terminally differentiated CD8(+) T-cells. Expressed in smooth muscle cells in atherosclerotic plaques.

Q&A

What is CX3CR1 and why is it important in research?

CX3CR1, also known as fractalkine receptor or G-protein coupled receptor 13 (GPR13), is a transmembrane protein belonging to the G protein-coupled receptor 1 (GPCR1) family. It is the sole member of the CX3C chemokine receptor subfamily and is encoded by the CX3CR1 gene in humans. Its significance stems from its critical role in inflammatory processes, cell migration, and tissue-specific immune responses. CX3CR1 is constitutively expressed in various hematopoietic-derived cells including T lymphocytes, natural killer cells, dendritic cells, monocytes, macrophages, microglia, and neutrophils, making it a valuable marker in immunology research . Recent studies have implicated the CX3CL1-CX3CR1 axis in fibrotic disorders, such as systemic sclerosis, highlighting its potential as a therapeutic target .

Which cell types express CX3CR1 and how can antibodies help identify them?

CX3CR1 is expressed in multiple cells of hematopoietic lineage, including:

  • T lymphocytes

  • Natural killer (NK) cells

  • Dendritic cells

  • B lymphocytes

  • Mast cells

  • Monocytes

  • Macrophages

  • Neutrophils

  • Microglia

  • Osteoclasts

  • Thrombocytes

Anti-CX3CR1 antibodies enable identification of these cell populations through techniques like immunofluorescence, flow cytometry, and immunohistochemistry. For example, flow cytometry using anti-CX3CR1 antibodies can distinguish CX3CR1-expressing myeloid cells, as demonstrated in studies with the M1 mouse myeloid leukemia cell line . When combined with other lineage markers, CX3CR1 antibodies can help identify specific subpopulations in complex tissues like the central nervous system or inflammatory microenvironments .

What applications are CX3CR1 antibodies validated for?

CX3CR1 antibodies have been validated for multiple research applications:

ApplicationValidated SpeciesTypical DilutionsReference
ImmunofluorescenceMouse, Rat1:100
ImmunohistochemistryMouse, Rat, Human1:100-1:500
Western BlotMouse, Human1 μg/mL
Flow CytometryMouse, HumanApplication-dependent
ImmunocytochemistryHumanApplication-dependent

Validation studies have confirmed specificity in multiple species, with antibodies detecting bands at approximately 40-50 kDa in western blot analysis, consistent with the predicted molecular weight of CX3CR1 .

How should I optimize CX3CR1 antibody staining protocols for immunofluorescence?

Optimizing CX3CR1 antibody staining requires careful consideration of several factors:

  • Fixation: For membrane proteins like CX3CR1, a mild fixation protocol is recommended. Typically, 2-4% paraformaldehyde for 10-15 minutes preserves epitope accessibility while maintaining tissue architecture.

  • Antigen retrieval: Heat-induced epitope retrieval in citrate buffer (pH 6.0) often enhances CX3CR1 detection, particularly in formalin-fixed tissues.

  • Blocking: Thorough blocking with 5-10% normal serum (matching the species of secondary antibody) with 0.1-0.3% Triton X-100 for 1-2 hours minimizes non-specific binding.

  • Antibody concentration: Start with the manufacturer's recommended dilution (typically 1:100 for immunofluorescence) and optimize as needed .

  • Incubation conditions: Overnight incubation at 4°C typically yields optimal staining with minimal background.

  • Secondary antibody selection: Choose secondary antibodies with minimal cross-reactivity to the species being examined, and include appropriate controls to verify specificity.

For optimal results in rodent tissues, validated anti-CX3CR1 antibodies have demonstrated reliable performance at 1:100 dilution with standard blocking protocols .

What are the critical controls for validating CX3CR1 antibody specificity?

Rigorous validation of CX3CR1 antibody specificity requires multiple controls:

  • Positive controls: Include known CX3CR1-expressing cells or tissues, such as:

    • Microglia in brain tissue

    • NK-92 human natural killer lymphoma cell line

    • M1 mouse myeloid leukemia cell line

  • Negative controls:

    • Primary antibody omission

    • Isotype controls (e.g., Catalog # AB-108-C has been successfully used as an isotype control for CX3CR1 antibodies)

    • CX3CR1 knockout tissues when available

    • Cell lines known not to express CX3CR1

  • Peptide competition: Pre-absorption of the antibody with the immunizing peptide should abolish specific staining.

  • Multiple detection methods: Confirm expression using two or more techniques (e.g., western blot and immunostaining).

  • Cross-validation with different antibody clones: Comparing results from different antibodies targeting distinct epitopes enhances confidence in specificity.

Research data supports this approach, as demonstrated in validation studies showing consistent detection patterns across multiple techniques .

How can I troubleshoot weak or non-specific CX3CR1 staining?

When encountering issues with CX3CR1 antibody staining, consider these solutions:

For weak signals:

  • Increase antibody concentration incrementally

  • Extend primary antibody incubation time (overnight at 4°C)

  • Optimize antigen retrieval methods (try different buffers or retrieval times)

  • Use signal amplification systems (e.g., biotin-streptavidin, tyramide signal amplification)

  • Ensure samples are properly fixed and processed

  • Check for protein degradation in older samples

For high background or non-specific staining:

  • Increase blocking time and concentration

  • Dilute antibody further

  • Reduce detergent concentration in wash buffers

  • Add 0.1-0.3% Triton X-100 to antibody diluent

  • Include 1-5% BSA or normal serum in antibody diluent

  • Increase washing steps and duration

  • Ensure secondary antibody specificity

For inconsistent results:

  • Standardize tissue processing protocols

  • Consider that CX3CR1 expression can be modulated by inflammation, affecting detection levels

  • Verify antibody storage conditions and avoid freeze-thaw cycles

  • Check microscope settings for consistency across experiments

Published studies have successfully detected CX3CR1 in multiple tissue contexts using these optimization approaches .

How do CX3CR1 antibodies perform across different species and what are the cross-reactivity considerations?

CX3CR1 antibodies exhibit variable cross-reactivity depending on the epitope targeted and species homology:

SpeciesValidated AntibodiesNotes on Cross-Reactivity
MouseMultiple options availableGenerally high specificity, well-characterized
RatSeveral validated optionsGood cross-reactivity with mouse-targeting antibodies
HumanLimited but increasing optionsRequires human-specific validation
Non-human primatesCase-by-caseMay cross-react with human-specific antibodies
Other mammalsLimited validationSequence alignment recommended before use

When selecting antibodies for cross-species applications, consider:

  • Epitope conservation: For the rat anti-CX3CR1 polyclonal antibody, the synthetic peptide immunogen is identical between mouse and rat sequences, showing 66.7% homology to human sequences . This explains its validated reactivity in both rodent species.

  • Domain targeting: Antibodies targeting conserved domains (such as AF5825, which targets multiple conserved epitopes) often show better cross-reactivity between human and mouse samples .

  • Validation method relevance: Antibodies validated by western blot may not necessarily work for immunohistochemistry in the same species due to differences in epitope accessibility.

To ensure reliability across species, preliminary validation experiments are strongly recommended before proceeding with full-scale studies.

How can CX3CR1 antibodies be used to study neuroinflammation and microglial function?

CX3CR1 antibodies are valuable tools for investigating neuroinflammation due to the high expression of CX3CR1 on microglia. Research applications include:

  • Microglial identification: Anti-CX3CR1 antibodies can specifically label microglia in brain tissue, distinguishing them from other CNS cells.

  • Activation state analysis: Changes in CX3CR1 expression correlate with microglial activation states. Quantitative analysis of staining intensity can reveal activation patterns in disease models.

  • Co-localization studies: Combining CX3CR1 antibodies with markers for proinflammatory cytokines or activation markers provides insights into microglial phenotypes.

  • In vivo tracking: Using CX3CR1 antibodies to identify microglia in tissue sections from experimental models allows tracking of migration and morphological changes.

  • Therapeutic target validation: As reported in mouse models, the CX3CL1-CX3CR1 axis represents a potential therapeutic target for neuroinflammatory conditions .

Methodologically, when studying microglia, consider:

  • Using thin sections (10-30 μm) to capture microglial processes

  • Employing z-stack confocal microscopy for 3D reconstruction

  • Combining with other markers (Iba1, TMEM119) for comprehensive characterization

  • Quantifying morphological parameters (process length, branching) in addition to expression levels

These approaches have been validated in studies examining microglial responses in neurodegenerative disease models .

What role does the CX3CL1-CX3CR1 axis play in fibrotic disorders and how can antibodies help investigate this?

The CX3CL1-CX3CR1 axis has emerged as a critical pathway in fibrotic disorders, particularly in systemic sclerosis (SSc) and related conditions. Research findings reveal:

  • Elevated serum biomarkers: Increased soluble CX3CL1 levels are observed in patients with severe SSc, interstitial lung disease, and digital ulcers .

  • Therapeutic potential: Anti-CX3CL1 monoclonal antibody treatment significantly inhibits TGF-β1-induced expression of type I collagen and fibronectin in human dermal fibroblasts, suggesting a direct anti-fibrotic effect .

  • Multi-organ benefits: Anti-CX3CL1 mAb therapy reduced both skin and lung fibrosis in sclerodermatous chronic graft-versus-host disease (Scl-cGVHD) mouse models without apparent adverse events .

  • Cellular mechanism: The therapeutic effects correlate with reduced tissue-infiltrating inflammatory cells, particularly CX3CR1-expressing macrophages and T cells, and decreased α-SMA-positive myofibroblasts in affected tissues .

CX3CR1 antibodies can be employed to investigate these mechanisms through:

  • Immunohistochemical analysis to quantify CX3CR1-expressing cells in fibrotic tissues

  • Flow cytometry to characterize infiltrating immune cell populations

  • Co-localization studies with fibrosis markers like α-SMA and collagen

  • In vitro studies examining the effects of CX3CL1/CX3CR1 blockade on fibroblast activation

These approaches have yielded valuable insights in preclinical studies, demonstrating that disruption of CX3CL1-CX3CR1 signaling may represent a rational therapeutic strategy for fibrotic disorders .

How do I design multi-color flow cytometry panels that include CX3CR1 for immune cell phenotyping?

Designing effective multi-color flow cytometry panels incorporating CX3CR1 requires strategic planning:

  • Panel design considerations:

    • CX3CR1 is expressed at variable levels across immune cells, so pairing with lineage-defining markers is essential

    • Common combinations include:

      • CX3CR1/CD14/HLA-DR for monocyte subsets

      • CX3CR1/CD3/CD56 for NK and T cell populations

      • CX3CR1/CD11b/Ly6C for mouse myeloid populations

  • Fluorophore selection:

    • Choose brighter fluorophores (PE, APC) for CX3CR1 when expression levels might be variable

    • Consider tandem dyes with minimal compensation requirements

    • Validated conjugates include PE-conjugated anti-goat IgG secondary antibodies used successfully with primary CX3CR1 antibodies

  • Staining protocol optimization:

    • Surface staining: Perform on ice to minimize receptor internalization

    • Buffer selection: Include sodium azide to prevent endocytosis during staining

    • Titrate antibody concentration to achieve optimal signal-to-noise ratio

    • When using indirect staining approaches, include appropriate blocking steps

  • Controls and analysis strategies:

    • Include isotype controls (such as Catalog # AB-108-C) to establish positive population boundaries

    • Use fluorescence-minus-one (FMO) controls for setting gates

    • Consider density plots rather than histograms for visualizing CX3CR1+ populations

  • Validation approaches:

    • Confirm staining patterns with alternative techniques

    • Compare with known population frequencies from literature

    • Use cell sorting followed by functional assays to confirm identity

These approaches have been successfully employed in studies characterizing CX3CR1+ cell populations in various disease models and tissues .

How can CX3CR1 antibodies contribute to therapeutic development for inflammatory and fibrotic diseases?

CX3CR1 antibodies play crucial roles in therapeutic development through several research applications:

  • Target validation: Antibodies against CX3CR1 and its ligand CX3CL1 have demonstrated efficacy in preclinical models of fibrotic disorders, validating this pathway as a therapeutic target. For example, anti-CX3CL1 monoclonal antibody treatment significantly reduced skin and lung fibrosis in sclerodermatous chronic graft-versus-host disease (Scl-cGVHD) mouse models .

  • Biomarker development: CX3CR1 antibodies enable quantification of receptor expression in patient samples, potentially identifying individuals more likely to respond to therapies targeting this pathway. Increased serum soluble CX3CL1 levels correlate with severe systemic sclerosis, suggesting utility as a stratification biomarker .

  • Mechanism of action studies: Using CX3CR1 antibodies in histopathological analyses revealed that therapeutic effects of CX3CL1 blockade correlate with reduced infiltration of CX3CR1-expressing macrophages and T cells in affected tissues, providing mechanistic insights .

  • Safety assessment: The absence of adverse events in preclinical studies with anti-CX3CL1 mAb treatment supports further exploration of this therapeutic approach .

  • Companion diagnostic potential: CX3CR1 antibodies could potentially serve as tools for companion diagnostics to identify patients with elevated pathway activity.

Research data supports these applications, as anti-CX3CL1 mAb therapy has shown promise across multiple fibrosis models, suggesting broader utility in human fibrotic disorders .

What are the technical considerations for using CX3CR1 antibodies in human tissue samples for translational research?

Translational research using CX3CR1 antibodies in human samples requires careful technical considerations:

  • Antibody selection:

    • Confirm human reactivity with validation data

    • Consider antibodies targeting conserved epitopes between species

    • Select antibodies with demonstrated performance in similar applications

    • Human/mouse cross-reactive antibodies (like AF5825) have been validated for both species

  • Sample preparation:

    • Fresh frozen tissues generally yield better results than FFPE for membrane proteins like CX3CR1

    • For FFPE samples, optimize antigen retrieval protocols (citrate buffer pH 6.0 or EDTA buffer pH 9.0)

    • Consider section thickness (5-10 μm optimal for most applications)

    • Test multiple fixation protocols when possible

  • Validation in human samples:

    • Always include known positive controls (e.g., NK cells, certain monocyte populations)

    • Perform peptide competition assays to confirm specificity

    • Compare staining patterns with published literature

    • Consider dual staining with antibodies against other established markers

  • Interpretation challenges:

    • Account for variability in CX3CR1 expression levels between individuals

    • Consider effects of medications on receptor expression

    • Disease state may alter receptor internalization or shedding

    • Tissue processing delays can impact membrane protein detection

  • Ethical and practical considerations:

    • Ensure appropriate ethical approvals and consent

    • Document clinical information relevant to interpretation

    • Consider time from collection to fixation as a variable

    • Standardize protocols across cohorts to enable comparison

Successful applications in human samples have been reported, including detection of CX3CR1 in oral squamous cell carcinoma cells and organoid cultures .

How is CX3CR1 expression being studied in the context of cancer immunology and metastasis?

CX3CR1 has emerged as a significant factor in cancer biology, with antibodies enabling several key research applications:

  • Tumor microenvironment characterization: CX3CR1 antibodies help identify specific immune infiltrates in tumors, revealing the composition and potential function of myeloid and lymphoid populations. Recent research indicates CX3CR1 expression influences cell migration and invasion in oral squamous cell carcinoma through ICAM-1 expression .

  • Metastatic process investigation: CX3CR1-expressing cells may facilitate tumor cell migration and invasion. Antibody-based detection systems allow tracking of CX3CR1+ cells in primary and metastatic sites.

  • Therapeutic target assessment: As the CX3CL1-CX3CR1 axis influences tumor-immune interactions, antibodies enable validation of this pathway as a potential therapeutic target in cancer models.

  • Prognostic biomarker evaluation: Quantification of CX3CR1+ cells in tumor samples may correlate with clinical outcomes, potentially serving as a prognostic indicator.

Methodological approaches include:

  • Multiplex immunofluorescence to analyze CX3CR1+ cells in relation to other immune markers and tumor cells

  • Flow cytometry to quantify CX3CR1 expression on circulating immune cells in cancer patients

  • In vitro migration and invasion assays using CX3CR1 antibodies to block function

  • Single-cell analysis combining CX3CR1 antibodies with other markers to identify specific cellular subsets

These applications have provided insights into cancer progression mechanisms involving CX3CR1-expressing cells .

What are the latest methodological advances in CX3CR1 detection for single-cell and spatial biology applications?

Recent technological advances have expanded CX3CR1 detection capabilities:

  • Single-cell protein analysis:

    • Mass cytometry (CyTOF) incorporation of CX3CR1 antibodies enables high-dimensional analysis of dozens of markers simultaneously

    • Spectral flow cytometry allows more fluorophore combinations with CX3CR1 antibodies

    • Single-cell Western blot techniques can verify CX3CR1 expression at the individual cell level

  • Spatial biology applications:

    • Multiplex immunofluorescence techniques combining CX3CR1 with multiple markers in single tissue sections

    • Imaging mass cytometry for high-parameter spatial analysis of CX3CR1+ cells

    • In situ hybridization combined with CX3CR1 immunostaining to correlate protein and mRNA expression

    • Proximity ligation assays to study CX3CR1 interactions with signaling partners

  • Live cell imaging advances:

    • Fluorescently-tagged CX3CR1 antibody fragments for dynamic imaging

    • Calcium flux imaging combined with CX3CR1 staining to correlate expression with functional responses

    • Optogenetic approaches incorporating CX3CR1 targeting

  • Multi-omics integration:

    • Antibody-based cell sorting followed by single-cell RNA sequencing

    • CITE-seq approaches incorporating CX3CR1 antibodies for simultaneous protein and transcriptome analysis

    • Spatial transcriptomics combined with CX3CR1 immunostaining for comprehensive tissue analysis

These methodological advances enable unprecedented insights into CX3CR1 biology in complex tissues and disease states, supporting deeper understanding of its role across biological systems .

What are the recommended resources for CX3CR1 antibody validation and protocol optimization?

For comprehensive CX3CR1 antibody information, researchers should consult:

  • Validated antibody repositories:

    • Antibodypedia for independent validation data

    • The Antibody Registry for unique identifier tracking

    • RRID portal for antibody standardization

  • Technical resources:

    • Manufacturer technical support (BiCell Scientific, R&D Systems, Abcam)

    • Application-specific protocols with optimization guidelines

    • Troubleshooting guides for common detection methods

  • Literature resources:

    • Systematic reviews of CX3CR1 detection methods

    • Primary research articles demonstrating successful applications

    • Method papers describing optimized protocols

  • Online communities and databases:

    • Research Gate forums for technical discussions

    • BioCompare for antibody comparison data

    • LinkedIn professional groups focused on antibody technologies

  • Standardization initiatives:

    • International Working Group for Antibody Validation guidelines

    • Reproducibility initiatives like the Antibody Validation Initiative

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.