Fractalkine Human, Sf9 denotes recombinant human fractalkine produced in Spodoptera frugiperda (Sf9) insect cells via baculovirus-mediated expression. This system allows for:
Potential mucin-like stalk retention (residues 1-336) for membrane-bound form studies
Post-translational modifications closer to mammalian systems compared to bacterial expression
Key production steps include:
cDNA cloning into baculovirus transfer vectors
Co-transfection with linearized baculovirus DNA in Sf9 cells
Sf9-derived fractalkine demonstrates dual functionality:
Adhesion Mechanism: Sf9-derived fractalkine enables integrin-independent adhesion - 60% of THP-1 cell binding persists despite β1/β2 inhibition .
Disease Relevance: Detected at 1560 pg/ml in inflammatory CSF samples , correlating with microglial activation .
Therapeutic Potential: Soluble fractalkine (10 nM) reduces ischemic brain injury by 40% in murine models .
Fractalkine, CX3CL1, Neurotactin, CX3C membrane-anchored chemokine, Small inducible cytokine D1, NTN, NTT, CXC3, CXC3C, SCYD1, ABCD-3, C3Xkine.
Sf9, Baculovirus cells.
QHHGVTKCNI TCSKMTSKIP VALLIHYQQN QASCGKRAII LETRQHRLFC ADPKEQWVKD
AMQHLDRQAA ALTRNGGTFE KQIGEVKPRT TPAAGGMDES VVLEPEATGE SSSLEPTPSS
QEAQRALGTS PELPTGVTGS SGTRLPPTPK AQDGGPVGTE LFRVPPVSTA ATWQSSAPHQ
PGPSLWAEAK TSEAPSTQDP STQASTASSP APEENAPSEG QRVWGQGQSP RPENSLEREE
MGPVPAHTDA FQDWGPGSMA HVSVVPVSSE GTPSREPVAS GSWTPKAEEP IHATMDPQRL GVLITPVPDA QAATRLEHHH HHH
Fractalkine (CX3CL1) is a unique chemokine that exhibits both cell adhesion and chemoattractive properties within the central nervous system (CNS). Unlike other chemokines, Fractalkine exists in both membrane-bound and soluble forms, allowing it to function as both an adhesion molecule and a traditional chemokine. In the CNS, Fractalkine is primarily expressed by neurons and astrocytes, as confirmed through reverse transcriptase-polymerase chain reaction (RT-PCR) analysis . The fractalkine receptor, CX3CR1, is expressed on neurons and microglia but notably absent on astrocytes . This differential expression pattern establishes a communication network where Fractalkine secreted by neurons and astrocytes can produce biological effects on neurons and microglia, creating a critical neuro-immune signaling pathway .
Functionally, Fractalkine plays a significant role in microglial regulation, with extracellularly applied Fractalkine shown to increase microglial proliferation three-fold over untreated controls, highlighting its importance in the neuro-inflammatory response . Additionally, Fractalkine acts as an adhesion molecule in NK cell interactions with endothelial cells, contributing to vascular processes and injury responses .
Fractalkine signaling through CX3CR1 receptors serves as a critical regulatory mechanism for microglial activation states in the CNS. The fractalkine-CX3CR1 axis appears to play a dual role depending on the neurological context and timing. In traumatic brain injury (TBI) models, CX3CR1 receptor deficiency has been associated with early protection against neurological damage . This suggests that disruption of regular fractalkine signaling can temporarily modulate the inflammatory response in acute brain injury.
Methodologically, researchers investigating microglial activation via fractalkine signaling should employ comprehensive approaches including:
Morphological analysis of microglial cells using immunohistochemistry with markers like CD11b, followed by detailed morphometric assessment of parameters including cell area, perimeter, Feret's diameter, circularity, and solidity
Assessment of microglial phenotypic markers (such as CD68) using confocal microscopy with Z-stack image acquisition (recommended 10 μm Z-axis with 0.23 μm step size)
Evaluation of TUNEL staining to correlate microglial activation with cellular injury and DNA damage
Analysis of cytokine/chemokine profiles using multiplex assays to understand the broader immune environment influenced by fractalkine signaling
Recent research suggests that local delivery of soluble fractalkine peptide can restore synaptic function after neuronal injury, highlighting its potential therapeutic applications in neuroinflammatory conditions .
Human Fractalkine possesses a unique structure among chemokines that directly influences its diverse biological functions. The protein consists of:
A chemokine domain (amino acids 25-105) - This 80 amino acid region contains the core signaling functionality
A mucin-like stalk - This region allows the protein to extend from the cell surface when membrane-bound
A transmembrane domain - Present only in the membrane-bound form
A cytoplasmic domain - Involved in intracellular signaling
Research has demonstrated that while the full-length fractalkine efficiently mediates NK cell adhesion, truncated forms containing only the chemokine domain or mucin domain fail to facilitate this adhesion . This structural requirement highlights the importance of the complete protein architecture for proper function.
Various recombinant forms of fractalkine are used in research, including:
E. coli-derived mouse CX3CL1/Fractalkine peptide (9.3 kDa, aa 25-105)
Baculovirus-derived mouse CX3CL1/Fractalkine protein (34 kDa, 312 amino acids)
When designing experiments, researchers must carefully consider which form is appropriate for their specific research questions, as different forms may elicit different biological responses.
Sf9 cells, derived from Spodoptera frugiperda, offer several advantages for the expression of complex proteins like human Fractalkine:
Post-translational modification capability - While not identical to mammalian cells, Sf9 cells can perform many post-translational modifications necessary for proper protein folding and function
High expression yields - Sf9 cells infected with baculovirus can dedicate significant cellular machinery to recombinant protein production
Scalability - Sf9 cultures can be scaled up efficiently compared to mammalian expression systems
Safety profile - Insect cell systems eliminate concerns related to mammalian pathogens
When expressing Fractalkine in Sf9 cells, researchers typically use the baculovirus expression system (BES) . Recent developments include transgenic Sf9 cell lines like Sf9-QE that allow for rapid virus quantification, significantly reducing the time required for complete virus quantification to approximately 5-6 days, which is 4-6 days faster than conventional methods . These advances improve workflow efficiency in Fractalkine production protocols.
For optimal results, researchers should note that Sf9-QE cells are typically smaller (average diameter around 16 μm compared to 18 μm for standard Sf9 cells) and proliferate approximately 1.6 times faster than standard Sf9 cells .
Detecting Fractalkine expression across various cell types requires a multifaceted approach that combines molecular and cellular techniques:
mRNA Detection:
RT-PCR remains the gold standard for initial assessment of fractalkine gene expression. In human CNS studies, this technique successfully distinguished expression patterns between neurons, astrocytes, and microglia .
Real-time quantitative PCR (RT-qPCR) on sorted cell populations (e.g., CD11b+ cells isolated via MACS technology) provides quantitative assessment of expression levels and temporal changes .
Protein Detection:
Immunohistochemistry/immunofluorescence using specific anti-fractalkine antibodies can visualize cellular distribution
Confocal microscopy with sequential scanning mode prevents bleed-through effects when co-staining with other markers
Recommended image acquisition parameters: 10 μm Z-axis imaging with 0.23 μm step size for optimal resolution
Secreted Fractalkine:
When analyzing fractalkine expression in complex tissues (e.g., brain), cell sorting prior to analysis is recommended to avoid misleading results from heterogeneous cell populations. For example, isolation of CD11b+ cells from brain tissue before RT-PCR analysis provides clearer cell-specific expression profiles .
Evaluating Fractalkine's effects on microglia requires methods that address both functional and phenotypic changes:
Proliferation Assessment:
Morphological Analysis:
CD11b immunostaining followed by quantitative morphometric analysis using software like Fiji
Key parameters to measure include cell area, perimeter, Feret's diameter, circularity, and solidity
Standardized image processing: scale images to microns (pixel size = 0.172 × 0.172 μm), subtract background, apply thresholding, and analyze cells with area > 25 μm²
Activation State Characterization:
Migration Assays:
Transwell migration assays to quantify chemotactic responses
Real-time cell migration tracking using live-cell imaging systems
For temporal studies, researchers should establish appropriate timepoints (e.g., 1 day, 2 days, 4 days, 7 days, and 5 weeks post-intervention) to capture both acute and chronic effects of fractalkine signaling on microglial populations .
Purifying functional human Fractalkine from Sf9 expression systems requires careful consideration of protein structure and bioactivity preservation:
Initial Harvest:
For secreted forms, collect culture supernatant 72-96 hours post-infection
For membrane-bound forms, cell lysis using detergent-based buffers containing protease inhibitors is recommended
Affinity Chromatography Options:
Histidine-tagged constructs: Ni-NTA or IMAC purification
Antibody-based purification: Anti-fractalkine monoclonal antibodies coupled to resin
Heparin affinity chromatography: Exploits the natural heparin-binding properties of chemokines
Secondary Purification:
Size exclusion chromatography to separate monomeric from aggregated forms
Ion exchange chromatography for charge-based separation of fractalkine from contaminants
Activity Preservation Considerations:
Maintain physiological pH (7.2-7.4) throughout purification
Include stabilizing agents such as glycerol (10-20%) in buffers
Minimize freeze-thaw cycles; single-use aliquots are recommended
Consider carrier-free formulations for downstream applications sensitive to additives
Quality Control:
Of particular note, researchers should be aware that commercially available recombinant fractalkine peptides vary significantly in source and structure. For example, baculovirus-derived mouse CX3CL1/Fractalkine (34 kDa, 312 amino acids) differs substantially from E. coli-derived peptides (9.3 kDa, aa 25-105) . The choice between these forms should be guided by specific experimental requirements.
Quantifying Fractalkine-CX3CR1 binding interactions requires methods that capture both affinity and functional consequences:
Receptor Expression Analysis:
Binding Affinity Determination:
Surface Plasmon Resonance (SPR) for label-free measurement of binding kinetics
Radioligand binding assays using 125I-labeled fractalkine
Competitive binding assays with unlabeled fractalkine to determine IC50 values
Functional Binding Assessment:
Cell adhesion assays measuring binding of CX3CR1-expressing cells to immobilized full-length fractalkine
Comparative studies using truncated forms (chemokine domain only or mucin domain only) to determine structural requirements for binding
Inhibition studies using soluble fractalkine or anti-CX3CR1 antibodies to confirm specificity
Signal Transduction Analysis:
G-protein activation can be assessed using pertussis toxin inhibition studies, as G-protein inhibition has been shown to completely block fractalkine-induced granular exocytosis from NK cells
Calcium flux assays to measure immediate receptor activation
Phosphorylation of downstream signaling molecules (ERK, Akt) via Western blotting
When designing binding studies, researchers should note that membrane-bound and soluble fractalkine may exhibit different binding characteristics. Studies have shown that while adhesion to full-length fractalkine is efficient, truncated forms fail to support this interaction .
The dual nature of Fractalkine as both a membrane-bound adhesion molecule and a soluble chemokine creates distinct functional outcomes in biological systems:
Membrane-Bound Fractalkine:
Primarily functions as an adhesion molecule, facilitating direct cell-cell contact
Studies demonstrate that full-length membrane-bound fractalkine effectively mediates NK cell adhesion to endothelial cells, while truncated forms fail to support this interaction
Contributes to sustained cellular interactions, potentially facilitating immune surveillance
May induce bidirectional signaling, affecting both the fractalkine-expressing cell and the CX3CR1-bearing cell
Soluble Fractalkine (sFKN):
Acts as a classical chemokine, creating concentration gradients that direct cell migration
Enhances NK cell cytolytic activity against target cells in a dose- and time-dependent manner
Increases granular exocytosis from NK cells through G protein-dependent pathways (inhibited by pertussis toxin)
Recent therapeutic applications show sFKN can restore ribbon synapses and hearing loss after neuronal injury
Regulatory Mechanisms:
Methodological Considerations:
When studying membrane-bound forms, cell surface expression should be confirmed via flow cytometry or immunofluorescence
For soluble fractalkine studies, ELISA or multiplex cytokine arrays can quantify levels in biological samples
Functional comparisons require parallel testing of both forms in the same experimental system
Recent therapeutic approaches highlight the potential of soluble fractalkine peptides (80 amino acids, aa 25-105) as interventional agents in neuroinflammatory conditions, suggesting unique functional properties of this form that merit further investigation .
Resolving contradictory findings in Fractalkine-CX3CR1 signaling research requires systematic consideration of several experimental variables:
Temporal Dynamics:
The timing of assessment is critical, as CX3CR1 deficiency shows contrasting effects depending on the phase of neurological injury
Implement comprehensive temporal analysis with multiple timepoints (e.g., 1 day, 2 days, 4 days, 7 days, and 5 weeks) to capture both acute and chronic effects
Studies should clearly distinguish between immediate effects and long-term consequences of fractalkine signaling
Context-Specific Effects:
The same signaling pathway may yield opposite outcomes in different disease models
Directly compare fractalkine effects across multiple experimental models using identical assessment methods
Control for variables such as inflammation status, tissue type, and age of the experimental system
Methodological Standardization:
Employ rigorous morphological analysis with standardized parameters (area, perimeter, Feret's diameter, circularity, solidity)
Use consistent cell isolation techniques; for example, MACS technology for CD11b+ cell sorting prior to gene expression analysis
Standardize image acquisition parameters (e.g., 10 μm Z-axis with 0.23 μm step size) for microscopy studies
Molecular Forms Consideration:
Different forms of fractalkine (full-length vs. truncated, E. coli-derived vs. baculovirus-derived) may elicit different responses
Clearly document the exact molecular form used, including size (e.g., 9.3 kDa vs. 34 kDa) and amino acid composition
Conduct side-by-side comparisons of different fractalkine forms to determine form-specific effects
Comprehensive Readouts:
When contradictory findings emerge, researchers should specifically examine differences in the microglial activation states being studied, as fractalkine may differentially affect specific microglial subpopulations or activation states.
The Sf9 baculovirus expression system offers powerful capabilities for structure-function analyses of human Fractalkine:
Mutational Analysis:
Generate systematic mutations in key domains (chemokine domain, mucin stalk, transmembrane region) to determine their contribution to function
Create chimeric proteins by swapping domains with other chemokines to identify unique structural determinants of fractalkine activity
Introduce site-specific mutations at predicted receptor interaction sites based on structural modeling
Truncation Studies:
Express series of truncated variants to determine minimal functional domains
Compare full-length fractalkine with isolated chemokine domain (aa 25-105) to distinguish adhesion versus chemokine functions
Evaluate differences in bioactivity between full-length (34 kDa) and truncated (9.3 kDa) forms in parallel assays
Post-Translational Modification Analysis:
Investigate the role of glycosylation by comparing Sf9-expressed fractalkine (with insect glycosylation patterns) to mammalian-expressed forms
Use site-directed mutagenesis to eliminate specific glycosylation sites and assess functional consequences
Analyze disulfide bonding patterns critical for chemokine domain structure
Advanced Expression Strategies:
Utilize novel transgenic Sf9 cell lines like Sf9-QE that enable rapid virus quantification (5-6 days vs. 9-12 days with conventional methods)
Implement fluorescent tagging strategies to monitor expression and localization without compromising function
Consider co-expression of human processing enzymes to better mimic human post-translational processing
Structural Biology Applications:
Optimize expression conditions for structural studies requiring high protein yields
Produce isotopically labeled fractalkine for NMR structural analysis
Generate crystallization-grade protein for X-ray crystallography studies
The smaller average diameter of optimized Sf9 cell lines (16 μm for Sf9-QE versus 18 μm for standard Sf9) and their faster proliferation rates (approximately 1.6× higher) can be advantageous for production efficiency in structure-function studies requiring multiple protein variants .
Translating Fractalkine research from in vitro systems to in vivo models presents several specific challenges:
Delivery and Bioavailability Issues:
Fractalkine's size and structure create challenges for effective delivery to target tissues
Local delivery approaches may be necessary, as demonstrated in studies using soluble fractalkine peptide to restore synaptic function after neuronal injury
Researchers must consider blood-brain barrier permeability when targeting CNS applications
Stability and half-life of recombinant fractalkine in vivo may differ significantly from in vitro conditions
Physiological Complexity:
The dual functionality of fractalkine as both adhesion molecule and soluble chemokine creates complex in vivo dynamics
Context-dependent effects seen in CX3CR1 knockout models highlight the importance of timing and disease state in determining outcomes
Comprehensive assessment requires multiple readouts, including behavioral testing, histological analysis, and molecular profiling
Source Material Considerations:
Different recombinant forms (e.g., E. coli-derived 9.3 kDa peptide vs. baculovirus-derived 34 kDa protein) may exhibit different in vivo activities
Species-specific differences in fractalkine signaling must be considered when translating between model organisms
Purification methods that remove endotoxin and other contaminants are essential for valid in vivo results
Experimental Design Complexity:
Analytical Approaches:
Multiplex analysis of inflammatory mediators provides broader context for understanding fractalkine effects in complex in vivo environments
Cell-specific responses require isolation techniques (e.g., MACS technology for CD11b+ cells) before molecular analysis
Image analysis should follow standardized protocols with clearly defined parameters for cell morphology assessment
Recent studies demonstrating therapeutic potential of locally delivered soluble fractalkine peptide highlight the possibility of overcoming these translation challenges through careful experimental design and delivery optimization .
Recent advances in insect cell engineering are opening new possibilities for enhanced Fractalkine production and research:
Transgenic Sf9 Cell Line Development:
Novel cell lines like Sf9-QE integrate fluorescent reporter systems for rapid virus quantification, reducing the time required from 9-12 days to just 5-6 days
These transgenic lines demonstrate altered cellular characteristics, including smaller average diameter (16 μm vs. 18 μm) and approximately 1.6× faster proliferation rates compared to standard Sf9 cells
Future engineering could target:
Humanized glycosylation pathways for more authentic post-translational modifications
Enhanced secretion mechanisms for improved yields of soluble fractalkine
Stable integration of fractalkine expression constructs for continuous production
CRISPR/Cas9 Applications:
Precise genome editing could eliminate proteases that degrade fractalkine during expression
Knockout of insect-specific glycosylation enzymes might reduce immunogenicity of recombinant fractalkine
Integration of mammalian chaperones could improve folding of complex domains
Expression System Optimization:
Development of promoter-enhancer combinations specifically optimized for glycoprotein expression
Engineering of customized signal sequences for improved secretion efficiency
Implementation of inducible expression systems for tight regulation of toxic proteins
Analytical Integration:
Next-generation transgenic lines could incorporate biosensors that monitor protein folding in real-time
Integration of secretion-coupled reporters to quantify fractalkine release dynamically
Development of cell lines expressing tagged CX3CR1 for direct binding studies
Scale-Up Technologies:
Adaptation of high-density culture systems specifically optimized for engineered Sf9 variants
Development of serum-free media formulations that enhance both cell growth and recombinant protein quality
Implementation of continuous processing technologies for sustained fractalkine production
These emerging approaches build upon current systems like the hr3 promoter-driven expression constructs used in generating the Sf9-QE cell line , moving toward more sophisticated platforms that combine high productivity with enhanced protein quality.
Emerging research highlights several promising therapeutic applications of Fractalkine in neurological disorders:
Traumatic Brain Injury (TBI):
CX3CR1 deficiency is associated with early protection after TBI, suggesting targeted modulation of the fractalkine pathway could provide therapeutic benefits
Temporal dynamics are critical, as fractalkine signaling may have different effects in acute versus chronic phases of injury
Research approaches should include comprehensive assessment of:
Hearing Loss and Synaptic Repair:
Local delivery of soluble fractalkine (sFKN) peptide has shown promise in restoring ribbon synapses and hearing function after noise-induced cochlear synaptopathy
The therapeutic sFKN (E. coli-derived mouse CX3CL1/Fractalkine peptide of 80 amino acids, aa 25-105) represents a specific molecular form with demonstrated efficacy
This approach provides proof-of-principle that targeted immune modulation can restore synaptic function after neuronal injury
Neuroinflammatory Conditions:
Fractalkine-CX3CR1 signaling regulates microglial activation states, suggesting potential applications in conditions characterized by microglial dysfunction
Comprehensive cytokine/chemokine profiling using multiplex analysis helps identify broader immune effects of fractalkine intervention
Careful consideration of delivery methods is essential, as demonstrated by successful local delivery approaches in cochlear models
Research Design Considerations:
Temporal analysis at multiple timepoints (e.g., 1 day, 4 days, 5 weeks) is crucial for capturing both immediate and long-term effects
Cell-specific responses should be evaluated through isolation of relevant populations (e.g., CD11b+ cells) prior to molecular analysis
Standardized morphological analysis parameters (area, perimeter, Feret's diameter, circularity, solidity) provide consistent assessment of microglial responses
As these therapeutic applications advance, researchers must carefully consider the specific molecular form of fractalkine being utilized, as functional differences exist between full-length (34 kDa) and truncated (9.3 kDa) variants, as well as between different expression systems (E. coli vs. baculovirus/Sf9) .
Systems biology offers powerful frameworks for unraveling the complex signaling networks associated with Fractalkine:
Multi-omics Integration:
Combine transcriptomics, proteomics, and metabolomics data from fractalkine-stimulated cells to construct comprehensive signaling maps
Analyze temporal dynamics using time-series experiments to capture both immediate and delayed responses
Compare system-wide responses across different cell types (neurons, microglia, NK cells) to identify cell-specific signaling nodes
Network Analysis of Inflammatory Mediators:
Multiplex cytokine/chemokine profiling can identify broader immune networks influenced by fractalkine signaling
Key analytes to include in multiplex panels: IL-1α, IL-1β, IL-2, IL-4, IL-6, IL-10, IL-12, IL-13, IL-15, IL-17, IL-18, IL-22, IL-23, IL-33, M-CSF, IFNα, IFNβ, IFNγ, TNF-α, CXCL1, CXCL2, CXCL10, CCL2, CCL3, CCL4, and CCL7
Network visualization techniques can identify hub molecules and feedback loops in fractalkine-responsive pathways
Computational Modeling:
Develop mathematical models of fractalkine signaling that incorporate both membrane-bound and soluble forms
Simulate perturbations to predict therapeutic targets and potential side effects
Validate model predictions using experimental approaches across multiple cell types
Single-Cell Analysis:
Apply single-cell RNA sequencing to identify heterogeneous responses within seemingly uniform populations
Correlate transcriptional changes with morphological parameters in microglia responding to fractalkine
Integrate spatial transcriptomics to map fractalkine signaling effects within complex tissue environments
Methodological Recommendations:
For protein interaction networks, use standardized isolation and analysis protocols
Process multiplex data using appropriate statistical approaches (e.g., 5PL algorithm) to accurately calculate concentrations
Employ comprehensive experimental designs that capture multiple timepoints and conditions to build robust models
By applying these systems biology approaches, researchers can move beyond studying isolated components of fractalkine signaling to understand the integrated networks that determine biological outcomes in complex tissues such as the brain.
Fractalkine pathway research is revealing potential biomarkers with significant implications for disease monitoring:
Soluble Fractalkine as a Biomarker:
Changes in circulating sFKN levels may reflect specific neurological conditions
Consider form-specific detection methods, as different molecular forms (e.g., 9.3 kDa vs. 34 kDa) may have distinct clinical implications
Develop standardized ELISA or multiplex assays optimized for detecting specific fractalkine forms in biological fluids
Receptor Expression Patterns:
CX3CR1 expression on circulating immune cells could serve as an accessible surrogate for CNS inflammation
Flow cytometric analysis can quantify receptor density and distribution on specific cell populations
Monitor changes in receptor expression following treatment interventions as a potential response biomarker
Downstream Signaling Molecules:
Identify stable and measurable products of fractalkine signaling in accessible biofluids
Develop multiplex panels that simultaneously measure fractalkine alongside related inflammatory mediators for a more comprehensive profile
Establish normative ranges across different demographic groups to enable clinical application
Microglia Morphological Signatures:
Advanced image analysis of microglial morphology using standardized parameters (area, perimeter, Feret's diameter, circularity, solidity) may serve as a tissue-based biomarker
These parameters reflect activation states that correlate with disease progression or treatment response
Development of AI-assisted image analysis could enable higher throughput assessment of these complex morphological features
Methodological Considerations:
For multiplex biomarker panels, standardized sample processing is critical to minimize pre-analytical variability
Comprehensive discovery approaches should include multiple analytes beyond just fractalkine pathway components
Longitudinal sampling is essential to establish temporal patterns and individual baselines
These emerging biomarker approaches build upon established methodologies such as MACS technology for cell isolation, standardized image analysis protocols, and multiplex cytokine/chemokine profiling that have proven valuable in fractalkine research .
Fractalkine, also known as CX3CL1, is a unique chemokine that plays a crucial role in the immune system. It is the only member of the CX3C chemokine family and is known for its dual functions: acting as both a potent chemotactic factor in its soluble form and an efficient cell adhesion molecule in its membrane-bound form .
The recombinant human Fractalkine (CX3CL1) produced in Sf9 Baculovirus cells is a single, glycosylated polypeptide chain containing 323 amino acids (25-339aa) and has a molecular mass of 34.3 kDa. It is fused to an 8 amino acid His tag at the C-terminus and purified using proprietary chromatographic techniques .
Fractalkine plays a significant role in the immune system by mediating the adhesion and migration of leukocytes. It is involved in various physiological and pathological processes, including:
Fractalkine is associated with the development of various diseases, including: