Recombinant Human Fractalkine protein (CX3CL1), partial (Active)

Shipped with Ice Packs
In Stock

Description

Production and Quality Control

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 .

Immune Modulation

  • 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 .

Neuroprotection

  • 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 .

Disease Models

  • 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 .

Mechanistic Studies

  • Receptor Signaling: Used to study CX3CR1 downregulation under inflammatory conditions .

  • Structural Analysis: Soluble chemokine domain facilitates crystallography and binding assays .

Key Research Findings

  1. NeuroAIDS: Exogenous fractalkine rescues neurons from Tat-morphine toxicity by normalizing microglial motility (Figure 3E, ).

  2. Chemokine Stability: N-terminal pyroglutamate modification enhances receptor interaction stability .

  3. Dual Signaling: Functions as both chemoattractant (soluble form) and adhesion molecule (membrane-bound form) .

Product Specs

Buffer
Lyophilized from a 0.2 µm filtered concentrated solution in 20 mM phosphate buffer (PB), pH 7.4, 50 mM sodium chloride (NaCl)
Description

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.

Form
Liquid or Lyophilized powder
Lead Time
5-10 business days
Shelf Life
The shelf life is influenced by various factors, including storage conditions, buffer composition, temperature, and the protein's inherent stability.
Generally, the shelf life of the liquid form is 6 months at -20°C/-80°C. The shelf life of the lyophilized form is 12 months at -20°C/-80°C.
Storage Condition
Store at -20°C/-80°C upon receipt. Aliquoting is recommended for multiple use. Avoid repeated freeze-thaw cycles.
Tag Info
Tag-Free
Synonyms
A 152E5.2; AB030188; ABCD 3; ABCD3; AI848747; C-X3-C motif chemokine 1; C3Xkine; Chemokine (C-X3-C motif) ligand 1; Chemokine C X3 C motif ligand 1; Chemokine CX3C Motif Ligand 1; CX3C membrane anchored chemokine; CX3C membrane-anchored chemokine; Cx3cl1; CXC 3; CXC3; CXC3C; D8Bwg0439e; FKN; Fractalkine; Neurotactin; NTN; NTT; Processed fractalkine; SCYD 1; SCYD1; Small inducible cytokine D1; Small inducible cytokine subfamily D (Cys X3 Cys) member 1; small inducible cytokine subfamily D (Cys-X3-Cys), member 1 (fractalkine, neurotactin); Small inducible cytokine subfamily D member 1; Small-inducible cytokine D1; X3CL1_HUMAN
Datasheet & Coa
Please contact us to get it.
Expression Region
25-100aa
Mol. Weight
8.6 kDa
Protein Length
Partial
Purity
>97% as determined by SDS-PAGE.
Research Area
Immunology
Source
E.Coli
Species
Homo sapiens (Human)
Target Names
Uniprot No.

Target Background

Function
CX3CL1 is a chemokine that functions as a ligand for both CX3CR1 and integrins ITGAV:ITGB3 and ITGA4:ITGB1. The CX3CR1-CX3CL1 signaling pathway exerts distinct functions in various tissue compartments, encompassing immune response, inflammation, cell adhesion, and chemotaxis. It regulates leukocyte adhesion and migration processes at the endothelium. CX3CL1 can activate integrins in both a CX3CR1-dependent and CX3CR1-independent manner. In the presence of CX3CR1, CX3CL1 activates integrins by binding to the classical ligand-binding site (site 1) in integrins. In the absence of CX3CR1, CX3CL1 binds to a second site (site 2) in integrins, distinct from site 1, and enhances the binding of other integrin ligands to site 1. The soluble form is chemotactic for T-cells and monocytes, but not for neutrophils. The membrane-bound form promotes adhesion of these leukocytes to endothelial cells.
Gene References Into Functions
  1. This study demonstrates that allergenic proteases directly cleave the chemokine CX3CL1 from the surface of airway epithelium, amplifying the effect of rhinovirus. PMID: 28677664
  2. The study revealed that CX3CL1/CX3CR1 was overexpressed in prostate cancer tissues with spinal metastasis compared to primary tumors. Overexpression of CX3CR1 promoted cell proliferation, migration, and invasion. Additionally, the study observed that the EGFR/Src/FAK pathway was activated by CX3CL1/CX3CR1. PMID: 30066854
  3. The chemokine CX3CL1 was identified as a central NF-kappaB target gene mediating therapy resistance. While no direct impact of CX3CL1 expression on cancer cell apoptosis was observed in co-culture assays, it became evident that CX3CL1 exerts a paracrine effect, leading to increased recruitment of inflammatory cells. PMID: 29867042
  4. TRAF1, CTGF, and CX3CL1 genes exhibit hypomethylation in osteoarthritis. PMID: 28470428
  5. Low shear stress (approximately 4.58 dyne/cm) for more than 1 hour promoted Fractalkine expression and activated the extracellular signal-regulated kinase (ERK)1/2, p38, and Jun N-terminal kinase (JNK) mitogen-activated protein kinases signaling pathways through phosphorylation. PMID: 29406386
  6. The findings strongly suggest that glutaminyl cyclase-catalyzed N-terminal pyroglutamate formation of CX3CL1 is essential for stability or interaction with its receptor, providing new insights into the function of glutaminyl cyclase in inflammation. PMID: 28739588
  7. Serum fractalkine levels were significantly elevated in the impaired glucose tolerance and type 2 diabetes groups compared to the normal glucose tolerance group. PMID: 29455547
  8. Reduced fractalkine levels were observed in follicular fluid and granulosa cells, accompanied by decreased progesterone production and reduced steroidogenic acute regulatory protein (StAR) expression in the granulosa cells of patients with polycystic ovary syndrome. Administration of fractalkine reversed the inhibition of progesterone and StAR expression. PMID: 27386819
  9. The US28 gene product has retained the function of the ancestral gene and possesses the ability to bind and signal in response to human CX3CL1, the natural ligand for CX3CR1. PMID: 28315475
  10. The findings indicate that fractalkine may be involved in the vulnerability of human carotid plaque. PMID: 28677375
  11. FKN concentrations are attenuated in girls with anorexia nervosa compared with healthy adolescents and are positively correlated with nutritional status. PMID: 27658415
  12. The CX3CL1/CX3CR1 axis plays a crucial role in the development of ischemia-induced oligodendrocyte injury via the p38MAPK signaling pathway. PMID: 26189830
  13. miR-223 regulates the expression of CX3CL1 by targeting HDAC2 in chronic obstructive pulmonary disease patients and mouse models of the disease. PMID: 26864305
  14. High levels of fractalkine in ectopic endometrium obtained from patients with endometriosis promoted proliferation and invasion of endometrial stromal cells through activation of the AKT and p38 signaling pathways. PMID: 27553970
  15. Modification of the cytokine profile in macrophages subsequent to their interaction with smooth muscle cells: Differential modulation by fractalkine and resistin. PMID: 27180200
  16. Soluble FKN, efficiently shed from the surface of LPS-activated ECs in response to binding of CD16(+) monocytes to ECs, diminished monocyte adhesion by down-regulating CX3CR1 expression on the surface of CD16(+) monocytes, resulting in decreased TNF-secretion. PMID: 27031442
  17. Data indicate that patients with systemic sclerosis (SSc) displayed higher serum levels of VEGF, endothelin-1, and s-Fractalkine. Moreover, s-Fractalkine levels positively correlated with CD34(+)CD45(-) endothelial progenitor cell (EPC) numbers. PMID: 28320472
  18. FKN and CX3CR1 expression was significantly elevated in pancreatic ductal adenocarcinoma (PDAC) tissues, particularly in metastatic samples, and was highly correlated with the severity of PDAC. Ectopic expression of FKN promoted proliferation and migration of PDAC, while knockdown of CX3CR1 reversed the effect of FKN. PMID: 28986258
  19. Fractalkine may be involved in both immunopathological and anti-viral immune responses to rhinovirus infection in asthma. PMID: 28859129
  20. High CX3CL1 expression is associated with spinal metastases. PMID: 28122354
  21. High expression of CX3CR1 correlates with significantly shorter survival, specifically in post-menopausal patients with advanced and terminal stages of the disease. Collectively, this supports a key regulatory role for the fractalkine axis in advanced and relapsed peritoneal metastasis in epithelial ovarian carcinoma. PMID: 27941884
  22. Changes in GSK-3beta activity and/or levels regulate the production and subsequent secretion of fractalkine, a chemokine involved in the immune response that has been linked to AD and various neurological disorders. PMID: 27832289
  23. In conclusion, leukoplakia-associated fibroblasts produced and secreted less CX3CL1 by inhibiting the ERK signaling pathway, thereby contributing to impaired cell resistance to Candida albicans. PMID: 27891323
  24. Fractalkine-CX3CR1 signaling has been shown to protect neurons. PMID: 27814376
  25. CX3CL1 exerts numerous effects on pathophysiological conditions, resulting in both negative and positive consequences on pathogenesis and outcome [review]. PMID: 27098399
  26. XCL2 and CX3CL1 expression in lung cancers and adjacent non-cancerous tissues was detected by quantitative PCR and ELISA. The expression of XCL2 and CX3CL1 increases with increasing degree of malignancy, indicating that both chemokines might be important targets in gene therapy for lung cancer. PMID: 27156946
  27. Recent studies demonstrate 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 at the inflammatory site. [review] PMID: 27011244
  28. Serum FKN may serve as a novel biomarker for assessing disease progression and a new potential therapeutic target for anti-resorptive treatment in osteoporosis patients. PMID: 27476376
  29. FKN may serve as a reliable biomarker for evaluating disease severity in atopic dermatitis patients. PMID: 27098623
  30. Post-transcriptional suppression of KSRP destabilizes CX3CL1 mRNA in liver epithelial cells. PMID: 26631623
  31. CX3CL1 increased the migration and invasiveness of DU145 and PC-3, causing epithelial-to-mesenchymal transition. TACE/TGF-alpha/EGFR pathway activation and Slug upregulation were responsible for this. PMID: 26718770
  32. Cell proliferation enhancement and anti-apoptosis activity require the intracellular domain and, apparently, the dimerization of the transmembrane chemokine ligand. PMID: 26796342
  33. CX3CL1 was expressed exclusively by the normal and cancer-adjacent normal fallopian tube epithelium; its expression was largely lost in the malignant fallopian epithelium. PMID: 26633537
  34. Skin and serum CX3CL1 elevated expression was associated with psoriasis severity. PMID: 26586708
  35. FKN enhances cell proliferation by promoting cell cycle progression in hypoxic prostate cancer cells. PMID: 26496926
  36. CX3CL1(+) apo-MPs released by apoptotic cells mediate the chemotactic transmigration of alveolar macrophages. PMID: 24603149
  37. This study suggests that CX3CL1 participates in cross-talk mechanisms between endothelium and other muscle tissue cells and may promote a shift in the microenvironment toward a more regenerative milieu after exercise. PMID: 26632602
  38. The study reports increased circulating fractalkine in STEMI patients, which was rapidly decreased after PCI. PMID: 26049921
  39. This study found that CX3CL1 and TREM2, two genes related to neuroinflammation, were expressed at higher levels in brain regions with pronounced vulnerability to Alzheimer disease-related changes. PMID: 25596843
  40. The interactions of CX3CL1, LEPR, and IL-6 genes might increase the risk of coronary artery disease in the Chinese Han population. PMID: 26191329
  41. Fractalkine levels in synovial fluid and serum reflect symptomatic severity in knee osteoarthritis. PMID: 25692263
  42. CX3CL1 and CX3CR1 may contribute to the formation of coronary atherosclerotic plaque in coronary artery disease. PMID: 25845619
  43. Forskolin-induced differentiation and syncytialization of the trophoblast cell line BeWo was accompanied by a substantial upregulation in fractalkine expression and led to increased adhesion of the monocyte cell line THP-1, which bound to syncytia. PMID: 25566740
  44. Fractalkine signaling regulates macrophage recruitment into the cochlea and promotes survival of spiral ganglion neurons. PMID: 26558776
  45. The results from the present study support the concept that CX3CL1-mediated activation contributes to the progression of multiple myeloma via CX3CR1. PMID: 25962684
  46. This study suggests that increased maternal TNF-alpha may up-regulate the expression and release of placental fractalkine, which, in turn, may contribute to an exaggerated systemic inflammatory response in preeclampsia. PMID: 25769431
  47. Data show that the US28 receptor binds with high selectivity and improved binding for the CX3C chemokine, CX3CL1. PMID: 26080445
  48. The data suggest that fractalkine contributes to lymphocyte shifts, which may influence the development of MVO through the action of effector T cells. PMID: 26168217
  49. In a CKD cohort, CX3CL1 levels were associated positively with several CVD risk factors and metabolic traits, a lower estimated glomerular filtration rate, and higher levels of inflammatory cytokines, as well as prevalent CVD and diabetes. PMID: 25795074
  50. Fractalkine levels are elevated in the first 12 hours after percutaneous coronary intervention in patients with acute myocardial infarction, however, are not correlated to infarct size. PMID: 24930044

Show More

Hide All

Database Links

HGNC: 10647

OMIM: 601880

KEGG: hsa:6376

STRING: 9606.ENSP00000006053

UniGene: Hs.531668

Protein Families
Intercrine delta family
Subcellular Location
Cell membrane; Single-pass type I membrane protein.; [Processed fractalkine]: Secreted.
Tissue Specificity
Expressed in the seminal plasma, endometrial fluid and follicular fluid (at protein level). Small intestine, colon, testis, prostate, heart, brain, lung, skeletal muscle, kidney and pancreas. Most abundant in the brain and heart.

Q&A

What is CX3CL1 and what makes it structurally unique among chemokines?

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.

How does soluble CX3CL1 differ functionally from membrane-bound CX3CL1?

Membrane-bound and soluble CX3CL1 exhibit different functional properties despite sharing the same chemokine domain:

FormPrimary FunctionSignal TransductionCellular Response
Membrane-boundCell adhesionSustained signalingAnti-inflammatory effects in some contexts
SolubleChemotaxisTransient signalingPro-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.

What are the standard assays to confirm the biological activity of 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.

What methodological approaches can effectively investigate the differential effects of membrane-bound versus soluble CX3CL1?

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 .

How does the intracellular domain of CX3CL1 contribute to its biological functions?

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.

What considerations should be taken when studying CX3CL1's role in specific disease contexts?

When investigating CX3CL1's role in disease pathogenesis, researchers should consider several context-specific factors:

  • Cell type-specific expression patterns:

    • In the central nervous system, CX3CL1 is primarily expressed by neurons while CX3CR1 is predominantly expressed by microglia

    • In kidney inflammation, CX3CL1 expression is induced on glomerular endothelium, while CX3CR1 is found on infiltrating T-cells and macrophages

  • 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:

    • The CX3CL1-CX3CR1 axis can exert either neurotoxic or neuroprotective effects depending on the neurological disease context

    • In the kidneys, blocking CX3CR1 with antibodies can attenuate disease severity in experimental glomerulonephritis

  • Developmental considerations:

    • CX3CL1-CX3CR1 signaling may play roles in embryonic development, including nephrogenesis, as both are expressed in the developing kidney

    • In the CNS, this signaling pathway contributes to neurodevelopment

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.

What are the optimal experimental conditions for studying CX3CL1-CX3CR1 interactions?

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.

How can dose-response curves for CX3CL1 be properly generated and interpreted?

Generating and interpreting dose-response curves for CX3CL1 requires careful consideration of several methodological aspects:

  • Proper concentration range selection:

    • For chemotaxis assays with recombinant human CX3CL1 chemokine domain, concentrations typically range from 0.01-10 ng/mL

    • For full-length CX3CL1, a wider range (0.1-100 ng/mL) may be necessary

    • Include concentrations spanning at least two logs below and one log above the expected EC50

  • 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.

What are common causes of inconsistent results when working with recombinant CX3CL1?

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

How can researchers effectively compare results from studies using different forms of recombinant CX3CL1?

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

What emerging methodologies are advancing our understanding of CX3CL1 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.

What are promising research areas for understanding CX3CL1's role in disease pathogenesis?

Based on current literature, several research areas show particular promise for expanding our understanding of CX3CL1's role in disease:

  • Neurological disorders:

    • Investigating the dual role of CX3CL1-CX3CR1 in neuroinflammation across different disease states

    • Exploring the neuroprotective mechanisms of CX3CL1-ICD nuclear signaling

    • Examining how CX3CL1 regulates microglial phenotype and function in neurodegenerative diseases

  • Kidney diseases:

    • Further characterizing CX3CL1's role in acute and chronic kidney diseases

    • Investigating therapeutic targeting of CX3CL1-CX3CR1 in glomerulonephritis

    • Understanding how CX3CL1 expression changes correlate with disease progression and treatment response

  • Viral infections:

    • Exploring CX3CL1's role in regulating immune responses during viral infections

    • Investigating viral strategies for modulating CX3CL1-CX3CR1 signaling

    • Developing CX3CL1-based approaches for enhancing antiviral immunity

  • 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:

    • Understanding CX3CL1's role in organ development, particularly in the kidney and brain

    • Investigating potential developmental origins of CX3CL1-related pathologies

    • Exploring the role of CX3CL1 in tissue regeneration and repair

These research directions hold promise for translating our molecular understanding of CX3CL1 into clinically relevant insights and potentially novel therapeutic approaches for various diseases.

What experimental models are most appropriate for studying CX3CL1 in different research contexts?

Selecting the appropriate experimental model is crucial for relevant CX3CL1 research:

  • Cell culture systems:

    • Transfected cell lines (e.g., BaF3 cells expressing human CX3CR1) for basic signaling studies

    • Primary microglia cultures for neuroinflammation studies

    • Primary kidney cells (podocytes, mesangial cells) for renal disease models

    • Co-culture systems to study cell-cell interactions mediated by membrane-bound CX3CL1

  • 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:

    • CX3CR1^gfp/+ reporter mice for tracking receptor-expressing cells in vivo

    • Conditional knockout models for tissue-specific deletion of CX3CL1 or CX3CR1

    • Disease-specific models (e.g., experimental autoimmune encephalitis, anti-GBM glomerulonephritis)

  • Human samples:

    • Renal biopsies from patients with various kidney diseases

    • Post-mortem brain tissue from patients with neurological disorders

    • Clinical blood samples for measuring soluble CX3CL1 levels

  • 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.

What are effective strategies for studying the cleavage and shedding of CX3CL1?

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:

    • Mass spectrometry to precisely identify cleavage sites and fragment composition

    • N-terminal sequencing of shed fragments to confirm cleavage points

    • Detection of intracellular domain (CX3CL1-ICD) using specific antibodies or tags

  • Functional assessment:

    • Comparing biological activities of shed fragments versus recombinant fragments

    • Nuclear translocation assays for the CX3CL1-ICD

    • Transcriptional profiling to assess gene expression changes induced by CX3CL1-ICD

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 .

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 2024 Thebiotek. All Rights Reserved.