The protein is produced via E. coli expression, followed by chromatographic purification. Quality assurance includes:
SDS-PAGE Analysis: Confirms molecular weight and homogeneity .
Functional Validation: Chemotaxis assays using BaF3 cells transfected with CX3CR1 show an ED50 of 0.3–1.5 ng/mL .
Stability: Lyophilized formulations retain activity for ≥12 months at -20°C .
Leukocyte Recruitment: Soluble CX3CL1 induces chemotaxis in monocytes and T-cells via CX3CR1 binding .
Integrin Activation: Enhances integrin-ligand binding through CX3CR1-dependent and independent pathways .
Microglial Regulation: Reduces microglial motility and toxicity in co-culture models exposed to HIV-1 Tat and morphine .
Calcium Flux Inhibition: Protects neurons from NMDA-induced apoptosis via ERK activation .
Neuroinflammatory Disorders: Attenuates synergistic neurotoxicity in HIV-opioid interaction models .
Autoimmune Diseases: Investigated in rheumatoid arthritis and atherosclerosis due to its role in leukocyte adhesion .
Receptor Signaling: Used to study CX3CR1 downregulation under inflammatory conditions .
Structural Analysis: Soluble chemokine domain facilitates crystallography and binding assays .
NeuroAIDS: Exogenous fractalkine rescues neurons from Tat-morphine toxicity by normalizing microglial motility (Figure 3E, ).
Chemokine Stability: N-terminal pyroglutamate modification enhances receptor interaction stability .
Dual Signaling: Functions as both chemoattractant (soluble form) and adhesion molecule (membrane-bound form) .
Recombinant Human CX3CL1, also known as Fractalkine, is a multifunctional chemokine involved in immune responses. It plays a critical role in the recruitment and activation of leukocytes[1]. Our recombinant human CX3CL1 is a partial protein, encompassing amino acids 25-100, with a molecular weight of 8.6 kDa. Expressed in Escherichia coli, this tag-free recombinant protein is available in liquid or lyophilized powder form, making it suitable for diverse research applications in the field of immunology.
Our Recombinant Human CX3CL1 exhibits a purity exceeding 97%, as determined by SDS-PAGE and HPLC, ensuring high quality for your research endeavors. The endotoxin level is less than 1.0 EU/ug, as established by the LAL method. The biological activity of our CX3CL1 has been validated by a chemotaxis bioassay utilizing human T-lymphocytes, demonstrating activity within a concentration range of 5.0-10 ng/ml.
Extensive studies have demonstrated that CX3CL1 is implicated in a wide array of immune-mediated disorders, including atherosclerosis, multiple sclerosis, rheumatoid arthritis, and inflammatory bowel disease[2]. Furthermore, CX3CL1 has been linked to the regulation of tumor progression and metastasis in specific types of cancer[3].
References:
1. Bazan JF, et al. A new class of membrane-bound chemokine with a CX3C motif. Nature. 1997;385(6617): 640-4.
2. Jones BA, et al. Fractalkine/CX3CL1: A potential new target for inflammatory diseases. Mol Interv. 2010;10(5): 263-70.
3. Park MH, et al. Serum fractalkine levels are elevated in patients with advanced non-small cell lung cancer. Korean J Intern Med. 2010;25(2): 146-52.
CX3CL1 (Fractalkine) is the only known member of the CX3C chemokine family, distinguished by a unique structural arrangement. Unlike other chemokines, CX3CL1 exists in both membrane-anchored and soluble forms, each mediating distinct biological activities . The full-length protein consists of several domains: a 24-amino acid signal peptide, a 76-amino acid chemokine domain (CKD), a 241-amino acid mucin stalk, a 31-amino acid transmembrane domain, and a 35-amino acid intracellular domain .
The chemokine domain contains the CX3C motif that gives the protein its classification. When membrane-bound, CX3CL1 functions as an adhesion molecule, while the soluble form (released through proteolytic cleavage) acts as a conventional chemokine that stimulates cellular migration . This dual functionality makes CX3CL1 particularly interesting for research in inflammation, immunity, and neuroscience.
Membrane-bound and soluble CX3CL1 exhibit different functional properties despite sharing the same chemokine domain:
Form | Primary Function | Signal Transduction | Cellular Response |
---|---|---|---|
Membrane-bound | Cell adhesion | Sustained signaling | Anti-inflammatory effects in some contexts |
Soluble | Chemotaxis | Transient signaling | Pro-inflammatory effects in some contexts |
Soluble CX3CL1 (containing only the chemokine domain) primarily functions as a chemoattractant for immune cells expressing CX3CR1. Experimental data shows that soluble recombinant human CX3CL1 activates CX3CR1 in a concentration-dependent manner with an EC50 of approximately 0.91nM, reaching maximal efficacy at 10nM .
In contrast, membrane-bound CX3CL1 serves as an adhesion molecule that facilitates direct cell-cell contact. Studies using Membrane Tethered Ligand (MTL) constructs suggest that membrane-bound CX3CL1 may initiate different signaling cascades compared to its soluble counterpart, potentially leading to distinct cellular responses . This functional dichotomy is critical to consider when designing experiments with recombinant CX3CL1.
The biological activity of recombinant human CX3CL1 is typically assessed using several established assays:
Chemotaxis assays: The gold standard for measuring CX3CL1 activity is its ability to induce chemotaxis in cells expressing CX3CR1. BaF3 mouse pro-B cells transfected with human CX3CR1 are commonly used, with the ED50 for chemotaxis ranging from 0.3-1.5 ng/mL for the chemokine domain alone and 2.5-10 ng/mL for full-length CX3CL1 .
Cell adhesion assays: For membrane-bound or full-length CX3CL1, adhesion assays using CX3CR1-expressing cells can demonstrate functionality.
Signal transduction assays: Reporter gene assays utilizing serum response element (SRE)-driven luciferase expression can measure CX3CR1 activation. These assays have demonstrated that CX3CR1 is activated by rhCX3CL1 in a concentration-dependent manner with an EC50 of 0.91nM .
Receptor binding assays: Competitive binding assays using labeled CX3CL1 can assess receptor binding affinity and specificity.
When validating a new lot of recombinant CX3CL1, researchers should consider performing multiple activity assays to ensure full functionality, particularly if the protein will be used for complex experiments.
Investigating the differential effects of membrane-bound versus soluble CX3CL1 requires specialized approaches:
Membrane Tethered Ligand (MTL) strategy: This approach has been effectively used to study various domains of CX3CL1. MTL constructs contain an extracellular linker, a transmembrane domain anchor, and the peptide ligand of interest . This strategy allows researchers to:
Compare different domains of CX3CL1 inexpensively
Study CX3CL1 in its endogenous membrane-anchored form
Examine how alterations in specific domains affect receptor activation
Domain swapping experiments: Creating chimeric constructs with various domains of CX3CL1 can help identify which regions are responsible for specific functions.
Serial stimulation experiments: Comparing cellular responses to sustained exposure to membrane-bound CX3CL1 versus pulsatile exposure to soluble CX3CL1 can reveal differences in signaling kinetics and downstream effects.
Co-culture systems: Establishing co-cultures of CX3CL1-expressing cells with CX3CR1-expressing cells allows for the study of direct cell-cell interactions mediated by membrane-bound CX3CL1.
Inhibitory approaches: Using domain-specific blocking antibodies or inhibitors to selectively interfere with either membrane-bound or soluble CX3CL1 function.
These methodologies have revealed that membrane-bound and soluble CX3CL1 can trigger distinct signaling pathways and cellular responses, which is particularly relevant in understanding CX3CL1's dual role in inflammation and tissue homeostasis .
Recent research has uncovered a previously unrecognized function of CX3CL1's intracellular domain (CX3CL1-ICD) that extends beyond its conventional role in CX3CR1 binding:
The CX3CL1-ICD is generated through sequential cleavage of membrane-bound CX3CL1 by α-, β-, and γ-secretases, similar to the Notch signaling pathway . Once cleaved, CX3CL1-ICD can translocate to the cell nucleus where it functions as a transcriptional regulator to alter gene expression .
This intrinsic "back-signaling" activity of CX3CL1-ICD has been demonstrated to exert neuroprotective effects in certain contexts. In the central nervous system, this mechanism represents a novel signaling pathway that operates independently of the canonical CX3CL1-CX3CR1 interaction .
For researchers studying CX3CL1, this finding suggests that:
Experiments using truncated forms of CX3CL1 lacking the intracellular domain may not capture the protein's full biological activity
Nuclear translocation assays may be necessary to fully characterize CX3CL1 signaling
Transcriptomic analyses following CX3CL1 stimulation should consider both CX3CR1-dependent and CX3CL1-ICD-dependent gene expression changes
This dual signaling capability (forward via CX3CR1 and backward via ICD) makes CX3CL1 an exceptional chemokine with multifaceted functions in cellular communication.
When investigating CX3CL1's role in disease pathogenesis, researchers should consider several context-specific factors:
Cell type-specific expression patterns:
Disease-specific regulation:
In brain disorders, CX3CL1 acts as a regulator of microglial activation during inflammation
In acute glomerulonephritis, both CX3CL1 and CX3CR1 expression are increased, with CX3CL1 prominently induced on the glomerular endothelium
In crescentic glomerulonephritis, CX3CL1 expression increases during acute disease and decreases following steroid treatment
Functional dichotomy:
Developmental considerations:
When designing experiments to study CX3CL1 in disease models, researchers should carefully select appropriate time points, cell types, and functional assays that reflect the complex and context-dependent roles of CX3CL1 signaling.
Optimizing experimental conditions is crucial for obtaining reliable and reproducible results when studying CX3CL1-CX3CR1 interactions:
Concentration considerations:
For soluble recombinant human CX3CL1 (chemokine domain), maximum efficacy in cell-based assays is typically reached at 10nM
The ED50 for chemotaxis ranges from 0.3-1.5 ng/mL for the chemokine domain alone and 2.5-10 ng/mL for full-length CX3CL1
Concentration-response curves should be generated for each experimental system to determine the optimal working concentration
Time course optimization:
Acute responses to CX3CL1 (e.g., calcium flux) occur within seconds to minutes
Chemotaxis assays typically require 3-4 hours of stimulation
Gene expression changes may require 4-24 hours of stimulation
For studies comparing soluble and membrane-bound CX3CL1, time courses should be carefully matched
Buffer and media considerations:
Serum components can affect CX3CL1 activity and stability
pH changes can alter CX3CL1-CX3CR1 binding kinetics
Carrier proteins may be necessary to prevent non-specific adsorption to plastic surfaces
Receptor expression levels:
Overexpression systems may show different kinetics compared to endogenous receptor levels
The level of CX3CR1 expression should be quantified and standardized across experiments
For transfection-based systems, optimal DNA concentrations should be determined empirically
Signaling pathway detection:
CX3CR1 couples to multiple G proteins and can activate diverse signaling pathways
Reporter gene assays using serum response element (SRE)-driven luciferase expression provide a reliable readout of receptor activation
For more complex signaling analyses, chimeric G proteins like Gq5i66V can be employed to channel signaling through specific pathways
Standardizing these conditions across experiments is essential for meaningful comparisons and reproducible results in CX3CL1 research.
Generating and interpreting dose-response curves for CX3CL1 requires careful consideration of several methodological aspects:
Proper concentration range selection:
Experimental setup:
Use serial dilutions prepared fresh for each experiment
Include appropriate vehicle controls for each concentration
Perform experiments in triplicate with at least three independent experiments
Data analysis approaches:
Plot data using non-linear regression to generate sigmoidal curves
Calculate EC50 values using appropriate software (e.g., GraphPad Prism)
Report both potency (EC50) and efficacy (maximum response) metrics
Interpretation considerations:
A typical dose-response curve for soluble rhCX3CL1 shows a sigmoidal pattern with an EC50 around 0.91nM
Different forms of CX3CL1 (chemokine domain alone vs. full-length) may yield different EC50 values
Right-shifted curves may indicate reduced potency, while reduced maximum response suggests reduced efficacy
Bell-shaped curves may indicate receptor desensitization at high concentrations
Common pitfalls to avoid:
Using degraded protein (check activity before experiments)
Inadequate equilibration time (allow sufficient time for binding)
Inappropriate statistical analysis (use non-linear rather than linear regression)
Failing to account for receptor desensitization in prolonged assays
By following these guidelines, researchers can generate reliable dose-response data that accurately reflects CX3CL1's biological activity and enables meaningful comparisons across different experimental conditions.
Inconsistent results when working with recombinant CX3CL1 can stem from several factors:
Protein stability issues:
CX3CL1 can lose activity through freeze-thaw cycles
Adhesion to plastic surfaces may reduce effective concentration
Oxidation of critical cysteine residues can impair activity
Receptor expression variability:
Inconsistent CX3CR1 expression levels between experiments
Receptor desensitization due to endogenous CX3CL1 production
Heterogeneity in receptor coupling to downstream signaling pathways
Experimental variables:
Changes in cell density affecting receptor numbers per cell
Variations in incubation times between experiments
Inconsistent buffer compositions or serum lot variability
Technical considerations:
For chemotaxis assays, minor variations in gradient formation can significantly impact results
For reporter gene assays, transfection efficiency variations between experiments
For binding assays, non-specific binding can obscure specific interactions
To minimize these issues, researchers should implement strict standardization protocols, including:
Aliquoting recombinant proteins to avoid multiple freeze-thaw cycles
Including carrier proteins to prevent adsorption to surfaces
Careful monitoring of cell density and passage number
Implementing positive controls in each experiment to normalize responses
Using consistent lot numbers of reagents when possible
Comparing results across studies using different forms of recombinant CX3CL1 requires careful consideration of several factors:
Standardization approaches:
Normalize activity to molar concentration rather than weight
Perform side-by-side activity comparisons using standardized assays
Calculate relative potency ratios between different forms
Form-specific considerations:
Chemokine domain alone (Gln25-Gly100) typically shows higher potency in chemotaxis assays (ED50: 0.3-1.5 ng/mL) compared to full-length protein (ED50: 2.5-10 ng/mL)
Full-length CX3CL1 (Gln25-Arg339) may engage additional binding sites or exhibit different signaling kinetics
Membrane-tethered constructs may show qualitatively different responses compared to soluble forms
Analytical framework:
Develop comparative EC50 tables across different assays and CX3CL1 forms
Consider both kinetic and equilibrium parameters when available
Distinguish between affinity, potency, and efficacy differences
Contextual interpretation:
Different forms may be more relevant to specific physiological or pathological contexts
Membrane-bound forms better represent cell-cell contact scenarios
Soluble forms better represent paracrine/endocrine signaling
Several cutting-edge methodologies are expanding our understanding of CX3CL1 signaling mechanisms:
Advanced imaging techniques:
Single-molecule imaging to track individual CX3CL1-CX3CR1 interactions
FRET-based biosensors to monitor real-time signaling events
Super-resolution microscopy to visualize CX3CL1 clustering and distribution
Genetic approaches:
CRISPR-Cas9 engineering to create domain-specific mutations in endogenous CX3CL1
Conditional knockout models to study tissue-specific CX3CL1 functions
Single-cell RNA sequencing to characterize heterogeneous cellular responses
Structural biology:
Cryo-EM studies of CX3CL1-CX3CR1 complexes in different activation states
Hydrogen-deuterium exchange mass spectrometry to map dynamic interaction interfaces
Computational modeling of the full-length CX3CL1 structure including the mucin stalk
Systems biology approaches:
Multi-omics integration to comprehensively map CX3CL1 signaling networks
Mathematical modeling of CX3CL1 gradient formation and receptor dynamics
Network analysis to identify key nodes in CX3CL1-regulated pathways
Translational methods:
Patient-derived organoids to study CX3CL1 function in disease-relevant contexts
Biomarker development based on soluble CX3CL1 levels in various conditions
High-throughput screening for selective modulators of CX3CL1-CX3CR1 signaling
These emerging methodologies are likely to resolve current contradictions in the literature and provide deeper insights into the complex biology of CX3CL1 signaling.
Based on current literature, several research areas show particular promise for expanding our understanding of CX3CL1's role in disease:
Neurological disorders:
Kidney diseases:
Viral infections:
Cancer biology:
Examining CX3CL1's dual role in tumor progression versus anti-tumor immunity
Investigating CX3CL1 as a potential biomarker for cancer prognosis
Exploring therapeutic targeting of CX3CL1-CX3CR1 in cancer immunotherapy
Developmental biology:
These research directions hold promise for translating our molecular understanding of CX3CL1 into clinically relevant insights and potentially novel therapeutic approaches for various diseases.
Selecting the appropriate experimental model is crucial for relevant CX3CL1 research:
Cell culture systems:
Ex vivo preparations:
Fresh tissue slices maintaining cellular architecture and local connections
Isolated glomeruli for studying CX3CL1 in kidney microcirculation
Brain organoids for developmental and neuroinflammatory studies
In vivo models:
Human samples:
Hybrid approaches:
Patient-derived cells in controlled experimental settings
Humanized mouse models expressing human CX3CL1/CX3CR1
The optimal model depends on the specific research question, with simpler systems (cell lines) being appropriate for mechanistic studies and more complex models (in vivo, human samples) necessary for understanding disease relevance and potential therapeutic applications.
Studying the regulated cleavage and shedding of CX3CL1 requires specialized methodological approaches:
Quantification techniques:
ELISA assays for measuring soluble CX3CL1 in culture supernatants or body fluids
Western blotting with domain-specific antibodies to detect full-length protein versus cleaved fragments
Flow cytometry to quantify cell surface CX3CL1 levels before and after stimulation
Cleavage inhibition approaches:
Pharmacological inhibitors of specific proteases (e.g., ADAM10, ADAM17)
Genetically engineered CX3CL1 with mutated cleavage sites
siRNA knockdown of specific proteases to determine their contribution
Real-time monitoring:
FRET-based biosensors to detect CX3CL1 cleavage in living cells
Live-cell imaging with fluorescently tagged CX3CL1 to track shedding events
Surface plasmon resonance to monitor cleavage kinetics in real-time
Identification of cleavage products:
Functional assessment:
Understanding the regulated cleavage of CX3CL1 is particularly important given the distinct biological activities of membrane-bound versus soluble forms, as well as the newly discovered signaling functions of the intracellular domain .