Recombinant Mouse Fractalkine protein (Cx3cl1), partial (Active)

Shipped with Ice Packs
In Stock

Description

Ligand-Receptor Interactions

  • CX3CR1 Activation: Mediates leukocyte adhesion, chemotaxis, and immune cell recruitment .

  • Integrin Activation: Binds directly to integrins to enhance ligand affinity, independent of CX3CR1 .

Neuroprotective Role

  • Synergistic Toxicity Mitigation: Protects striatal neurons from combined HIV-1 Tat and morphine toxicity by normalizing microglial motility and reducing neuron death .

  • CX3CR1 Dependency: Neuroprotection requires functional CX3CR1 on microglia .

Immune Modulation

  • Soluble Form: Chemotactic for T cells and monocytes .

  • Membrane-Bound Form: Promotes leukocyte adhesion .

Neuroinflammation Studies

  • Key Finding: Exogenous fractalkine rescues neurons exposed to Tat and morphine, despite persistent TNF-α elevation .

  • Mechanism: Restores CX3CR1 levels on microglia, counteracting receptor downregulation caused by neurotoxic stimuli .

Cancer Immunotherapy

  • Antitumor Activity: Adenoviral delivery of CX3CL1 recruits cytotoxic lymphocytes (NK cells, CD8+ T cells) to tumors, enhancing innate and adaptive immunity .

Integrin Signaling

  • Dual Activation:

    • CX3CR1-Dependent: Binds classical integrin ligand-binding site (site 1) .

    • CX3CR1-Independent: Enhances ligand binding via integrin site 2 .

Functional Domains and Sequence

  • Post-Translational Modifications: Lacks glycosylation due to prokaryotic expression .

Clinical and Preclinical Relevance

  • Neurodegenerative Diseases: Potential therapeutic agent for Alzheimer’s and HIV-associated neurocognitive disorders .

  • Cardiovascular Disease: Soluble CX3CL1 correlates with atherosclerosis progression .

Product Specs

Buffer
Lyophilized from a 0.2 µm filtered PBS, pH 7.4
Form
Lyophilized powder
Lead Time
5-10 business days
Notes
Repeated freezing and thawing is not recommended. Store working aliquots at 4°C for up to one week.
Reconstitution
We recommend that this vial be briefly centrifuged prior to opening to bring the contents to the bottom. Please reconstitute the protein in deionized sterile water to a concentration of 0.1-1.0 mg/mL. We recommend adding 5-50% of glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our default final concentration of glycerol is 50%. Customers can use this as a reference.
Shelf Life
The shelf life is dependent on several factors, including storage state, buffer ingredients, storage 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 necessary for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag-Free
Synonyms
Cx3cl1; Cx3c; Fkn; Scyd1; Fractalkine; FK; C-X3-C motif chemokine 1; CX3C membrane-anchored chemokine; Neurotactin; Small-inducible cytokine D1
Datasheet & Coa
Please contact us to get it.
Expression Region
25-100aa
Mol. Weight
8.7 kDa
Protein Length
Partial
Purity
>97% as determined by SDS-PAGE.
Research Area
Immunology
Source
E.coli
Species
Mus musculus (Mouse)
Target Names
Uniprot No.

Target Background

Function
Fractalkine is a chemokine that acts as a ligand for both CX3CR1 and integrins ITGAV:ITGB3 and ITGA4:ITGB1. The CX3CR1-CX3CL1 signaling pathway exerts distinct functions in different tissue compartments, such as immune response, inflammation, cell adhesion, and chemotaxis. Fractalkine regulates leukocyte adhesion and migration processes at the endothelium. It can activate integrins in both a CX3CR1-dependent and CX3CR1-independent manner. In the presence of CX3CR1, fractalkine activates integrins by binding to the classical ligand-binding site (site 1) in integrins. In the absence of CX3CR1, fractalkine binds to a second site (site 2) in integrins, which is distinct from site 1 and enhances the binding of other integrin ligands to site 1. The soluble form of fractalkine is chemotactic for T-cells and monocytes, but not for neutrophils. The membrane-bound form promotes adhesion of those leukocytes to endothelial cells.
Gene References Into Functions
  1. This study suggests that overexpression of only the chemokine domain of CX3CL1 does not protect against tau pathology. PMID: 30253780
  2. The CX3CL1/CX3CR1 axis contributes to the proliferative and pro-inflammatory effects of Ang II in VSMCs. PMID: 29356931
  3. CX3CL1-CX3CR1 signaling is a molecular mechanism capable of modulating microglial-mediated degeneration. PMID: 27314452
  4. CXCR4(+) CD45(-) bone marrow cells are niche forming for osteoclastogenesis via the SDF-1, CXCL7, and CX3CL1 signaling pathways in bone marrow. PMID: 27339271
  5. Increased fractalkine and its receptor CX3CR1 may cause a cross-talk between activated glial cells and neurons, playing an important role in the development of neuroinflammation in fructose-fed mice. PMID: 26765996
  6. miR-223 controls the expression of CX3CL1 by targeting HDAC2 in chronic obstructive pulmonary disease patients and mouse models of the disease. PMID: 26864305
  7. This study identified CX3CL1 as a novel substrate of MMP-19. PMID: 26555704
  8. The CX3CL1/CX3CR1 system is essential for restricting coxsackievirus B3-induced myocarditis. PMID: 28800592
  9. 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 other neurological disorders. PMID: 27832289
  10. Medial ganglionic eminence (MGE) interneurons secrete fractalkine that promotes genesis of oligodendrocytes from glially biased cortical precursors in culture. Moreover, when MGE interneurons are genetically ablated in vivo prior to their migration, this causes a deficit in cortical oligodendrogenesis. PMID: 28472653
  11. These findings reveal a previously unknown regulatory role for LRRK2 in CX3CR1 signaling and suggest that an increase of CX3CR1 activity contributes to the attenuated inflammatory responses in Lrrk2-null microglia. PMID: 27378696
  12. This study reports a crucial role of CX3CL1-CX3CR1 in experimental colitis, in particular for intestinal leukocyte recruitment during murine colitis. PMID: 27942903
  13. These results strongly suggest the involvement of CX3CL1 in the migration of osteoclast precursors and osteoclastogenesis. PMID: 27579490
  14. Authors provide evidence that interactions between CX3CL1 and CX3CR1 play crucial roles in determining the number of M1 macrophages within the skin of mice, which in turn can have dramatic effects on psoriasis-like inflammation. PMID: 26976687
  15. In the absence of the rd8 allele, deficiency of CCR2 and CX3CL1 in mice leads to a mild form of retinal degeneration which is associated with the recruitment of macrophages, particularly to the subretinal space. This model enables to assess consequences of perturbed chemokine signaling, but it does not recapitulate cardinal age-related macular degeneration features. PMID: 26670885
  16. Also, icariin reduced CX3CR1 and CX3CL1 protein levels in the artery wall. In conclusion, icariin could be a potential anti-atherosclerosis agent by downregulating the expression of CX3CR1. PMID: 26802470
  17. The biological activity of CX3CL1 is regulated by conversion of a membrane integrated to a soluble form during neurogenesis and in response to pathologic changes in the adult retinal milieu. PMID: 25191897
  18. CX3CL1/CX3CR1 signaling is involved in LTP of C-fiber-evoked field potentials in the rodent spinal dorsal horn. PMID: 25768734
  19. CX3CL1/CX3CR1-mediated microglial activation plays a detrimental role in ischemic brain via p38MAPK/PKC signaling. PMID: 25966946
  20. Cx3cl1 overexpression suppresses alpha-synuclein-mediated neurodegeneration. PMID: 25195598
  21. IFN-gamma induces aberrant CD49b+ NK cell recruitment and pregnancy failure through regulating CX3CL1. PMID: 25375377
  22. Insulin resistance increases plaque vulnerability by augmenting the CX3CL1/CX3CR1 axis, which is mechanistically linked to reduced vascular smooth muscle cell survival. PMID: 24788416
  23. Together, these studies challenge the "frustrated phagocytosis" concept and suggest that neuronal-microglial communication link the two central AD pathologies. PMID: 25209291
  24. CX3CL1 may contribute to the regulation of toxigenic C. difficile infection. PMID: 24362517
  25. These results demonstrate that the de novo CX3CL1-CX3CR1 axis plays a pivotal role in osteoclast recruitment and subsequent bone resorption. PMID: 24401612
  26. Our data suggest that the CX3CR1/ CX3CL1 pathway is involved in the recruitment of circulating CD16 thorn CX3CR1 thorn monocytes to the periprosthetic tissues. PMID: 24700421
  27. Data indicate that atopic dermatitis (AD) pathology and immune responses were profoundly decreased in CX3CR1-deficient mice and upon blocking CX3CL1-CX3CR1 interactions in wild-type mice. PMID: 24821910
  28. Loss of ACE2 exacerbates AngII-mediated inflammation, myocardial injury, and dysfunction in ACE2-deficient hearts via activation of the CTGF-FKN-ERK and MMP signaling. PMID: 24161906
  29. CX3CL1 transiently potentiates NMDAR function though mechanisms involving A2AR activity and the release of D-serine. PMID: 23981568
  30. Interactions between CX3CL1 and CX3CR1 may contribute to the development of leukocytoclastic vasculitis. PMID: 23470165
  31. HDACs and NF-kB signaling coordinate epithelial expression of CX3CL1 to promote mucosal antimicrobial defense through suppression of the mir-424-503 gene. PMID: 23724129
  32. Carotid artery injury was associated with greater chemokine 1 CX3C expression in the acute phase followed by greater CX3C receptor 1 coexpressing smooth muscle-like cell content in later lesions and less neointima formation than in femoral arteries. PMID: 23653073
  33. Our data indicate an upregulation of fractalkine and downregulation of CX3CR1 in sepsis, which seems to be mediated by the transcripting factor NF-KappaB likely via reduced liberation of proinflammatory cytokines in the whole murine organism. PMID: 23026294
  34. Extracellular adenosine is an endogenous modulator of neuroinflammation that induces CX3CL1 at the chorid plexus. PMID: 22883932
  35. Fractalkine's essential role in the formation of atherosclerotic lesions and atherosclerosis progression has been impressively described in mouse models. Review. PMID: 22739755
  36. Suggest that Ang-II induces functional CX(3)CL1 expression in arterial but not in venous endothelial cells. PMID: 23117657
  37. H(2)S hampers the progression of atherosclerosis in fat-fed apoE(-/-) mice and downregulates CX3CR1 and CX3CL1 expression on macrophages and in lesion plaques. PMID: 22815945
  38. Hypoxic release of endothelial CX3CL1 induced SMC phenotypic switching from the contractile to the proliferative state. Inhibition of CX3CR1 prevented CX3CL1 stimulation of SMC proliferation and monolayer expansion. PMID: 23002075
  39. Fractalkine is expressed in early and advanced atherosclerotic lesions and supports monocyte recruitment via CX3CR1. PMID: 22916279
  40. Fractalkine induced cellular reactive oxygen species production and activation of ERK1/2 and p38 MAPK in mesangial cells, stimulating cell proliferation. PMID: 22564616
  41. Lipopolysaccharide induces monocyte-mesangial cell binding through the fractalkine/CX3CR1 system. PMID: 22564617
  42. Findings that exogenous fractalkine reduces microglial motility and fails to protect neurons co-cultured with Cx3cr1-/- mixed glia suggest that fractalkine may act by interfering with toxic microglial-neuron interactions. PMID: 22093090
  43. Fractalkine promotes myocardial injury and accelerates the progress of heart failure, which is associated with the activation of MAPKs. PMID: 21840883
  44. Structure/function and expression analysis of the CX3C chemokine fractalkine. PMID: 21951685
  45. The fractalkine-CX3CR1 axis contributes to kidney fibrosis in a hypertensive mouse model. PMID: 21451526
  46. Syk mediated chemotaxis toward CX3CL1 by regulating both Rac1/WAVE2 and Cdc42/WASP pathways, whereas Src family kinases were required for proper WASP tyrosine phosphorylation. PMID: 21388954
  47. Fractalkine attenuates excito-neurotoxicity via microglial clearance of damaged neurons and antioxidant enzyme heme oxygenase-1 expression. PMID: 21071446
  48. CX3CR1-expressing macrophages are induced by CX3CL1/fractalkine to express heme oxygenase-1, thereby ameliorating Clostridium difficile toxin A-induced enteritis. PMID: 21131421
  49. Fractalkine-induced depression of excitatory postsynaptic current is absent in cultures from adenosine A3 receptor-deficient mice but not in tissue from adenosine A1- or A2-receptor-deficient mice. PMID: 20570369
  50. The CX3CL1-CX3CR1 interaction inhibits inflammatory properties in Kupffer cells/macrophages and results in decreased liver inflammation and fibrosis. PMID: 20683935

Show More

Hide All

Database Links
Protein Families
Intercrine delta family
Subcellular Location
Cell membrane; Single-pass type I membrane protein.; [Processed fractalkine]: Secreted.
Tissue Specificity
Highest levels in brain. Lower levels in kidney, heart and lung. Also found in skeletal muscle and testis. Highly expressed in lesional smooth muscle cells, but not macrophages.

Q&A

What is the molecular structure of Recombinant Mouse Fractalkine (Cx3cl1) protein, and how does it compare to the native form?

Recombinant Mouse Fractalkine protein typically comprises only the chemokine domain (approximately amino acids 22-105 or 25-105) of the native protein. While native Fractalkine exists in two forms—a membrane-bound protein (~95 kDa) tethered by a mucine-like stalk and a soluble factor (~70 kDa) released upon cleavage—the recombinant protein represents just the chemokine domain with a molecular mass of approximately 8.7-9.5 kDa . This non-glycosylated polypeptide chain contains 76-84 amino acids and lacks the mucin-like stalk present in the membrane-bound form .

The amino acid sequence of commercially available Recombinant Mouse Cx3cl1 is typically:
QHLGMTKCEIMCGKMTSRIPVALLIRYQLNQESCGKRAIVLETTQHRRFCADPKEKWVQDAMKHLDHQAAALTKNG

How does the recombinant partial Mouse Cx3cl1 maintain biological activity despite lacking the mucin-like stalk?

The chemokine domain alone is sufficient for receptor binding and activation. Functional assays demonstrate that the E. coli-derived recombinant chemokine domain maintains high biological activity, with ED50 values typically less than 0.5 ng/mL in cell proliferation assays using human peripheral blood lymphocytes (PBL), corresponding to a specific activity greater than 2000 IU/mg . This indicates that while the mucin-like stalk is important for membrane tethering in native contexts, the chemokine domain independently retains receptor-binding properties essential for biological function .

What are the optimal methodologies for using Recombinant Mouse Cx3cl1 in chemotaxis assays?

For chemotaxis assays, the recommended methodology includes:

  • Cell preparation: Use CX3CR1-expressing cells such as microglia, monocytes, or CX3CR1-transfected cell lines (e.g., BaF3 mouse pro-B cells transfected with human CX3CR1) .

  • Assay setup: A standard transwell migration system (e.g., millicell PCF 8 μm insert) with Recombinant Mouse Cx3cl1 added to the lower chamber at concentrations ranging from 1-100 ng/mL. The optimal concentration for chemotaxis is typically around 30 ng/mL .

  • Quantification: After 3-4 hours incubation, migrated cells can be quantified by fluorescence (if cells express fluorescent proteins), by DAPI nuclear staining, or by metabolic assays such as Resazurin .

  • Controls: Include both negative controls (media without Cx3cl1) and positive controls (known chemoattractants like SDF-1 or MCP-1) to validate the assay system .

In validation studies, dose-dependent migration responses typically show a bell-shaped curve, with maximal migration at 10-50 ng/mL of Recombinant Mouse Cx3cl1 .

How can researchers effectively validate the cross-species reactivity between Mouse Cx3cl1 and human CX3CR1?

Cross-species reactivity between Mouse Cx3cl1 and human CX3CR1 can be validated using the following approaches:

  • Transmigration assays: Culture bone marrow-derived macrophages from CX3CR1-knockout mice and transfect them with expression vectors containing human CX3CR1 variants (reference V249/T280 or variant I249/M280). Expose these cells to different concentrations of soluble recombinant Mouse Fractalkine (1-100 ng/mL) in a transwell system and quantify migrated cells .

  • Calcium mobilization assays: Measure intracellular calcium flux in human CX3CR1-expressing cells after stimulation with Mouse Cx3cl1 .

  • Binding assays: Use radiolabeled or fluorescently-labeled Mouse Cx3cl1 to measure direct binding to human CX3CR1-expressing cells .

Research demonstrates that human CX3CR1 receptors can respond to Mouse Fractalkine, but with varying efficiency depending on the human receptor variant. The reference human receptor (V249/T280) signals effectively in response to Mouse Fractalkine, while the variant human receptor (I249/M280) shows blunted responses .

How can Recombinant Mouse Cx3cl1 be utilized to investigate microglial physiology in neuroinflammation models?

Recombinant Mouse Cx3cl1 serves as a valuable tool for dissecting microglial responses in neuroinflammation through several methodological approaches:

  • Microglia isolation and stimulation: Primary microglia can be isolated from mouse brain tissue and stimulated with Recombinant Mouse Cx3cl1 to study:

    • Calcium mobilization (within minutes of application)

    • Cytoskeletal rearrangement

    • Migratory behavior

    • Cytokine production profiles

    • Gene expression changes

  • Ex vivo slice cultures: Brain slices can be treated with Recombinant Mouse Cx3cl1 to observe microglial dynamics in a preserved tissue architecture using time-lapse microscopy .

  • Comparative studies with CX3CR1-deficient models: Experiments comparing wild-type microglia with CX3CR1-knockout microglia can evaluate receptor-dependent responses to Recombinant Mouse Cx3cl1 .

  • EAE (Experimental Autoimmune Encephalomyelitis) models: Researchers can manipulate the Fractalkine/CX3CR1 axis using Recombinant Mouse Cx3cl1 in combination with MOG35-55 immunization to study microglial contributions to neuroinflammation .

Research findings indicate that Fractalkine signaling critically regulates microglial properties during normal physiological conditions. In neuroinflammatory conditions, basal fractalkine levels in wild-type mice show approximately fivefold increase during EAE-induced disease, while CX3CR1-deficient mice show elevated baseline levels that remain unchanged during disease progression .

What methodological approaches can be employed to study the role of Cx3cl1-CX3CR1 signaling in osteoclastogenesis?

To investigate Cx3cl1-CX3CR1 signaling in osteoclastogenesis, researchers can employ the following methodologies:

  • Osteoclast precursor (OCP) culture systems:

    • Isolate CD11bhighCD115+ OCPs from bone marrow

    • Culture OCPs on immobilized Recombinant Mouse Cx3cl1 prior to RANKL stimulation

    • Assess osteoclast formation through TRAP staining and counting multinucleated cells

  • Gene expression analysis:

    • Monitor expression of osteoclast differentiation markers (Nfatc1, Calcr, Ctsk)

    • Track dynamic changes in Cx3cr1 and Rank (Tnfrsf11a) expression during differentiation using qRT-PCR

    • Studies show that growth on immobilized Cx3cl1 increases expression of both Cx3cr1 and Rank transcripts, but following RANKL stimulation, OCPs rapidly downregulate Cx3cr1 expression

  • Survival and differentiation assays:

    • Measure OCP survival using Annexin V/PI staining

    • Quantify CD11bhighCD115+ OCP numbers using flow cytometry

    • Research indicates that growth on immobilized Cx3cl1 prior to RANKL stimulation increases CD11bhighCD115+ OCP number and enhances their survival and differentiation potential

  • In vivo models of bone loss:

    • RANKL-induced bone loss model with anti-Cx3cl1 monoclonal antibody pretreatment

    • µCT analysis of bone parameters

    • Studies demonstrate that anti-Cx3cl1 mAb pretreatment significantly inhibits RANKL-dependent bone loss

Experimental GroupOsteoclast FormationCx3cr1 ExpressionRank ExpressionOCP Survival
Control (no Cx3cl1)BaselineBaselineBaselineBaseline
Immobilized Cx3cl1EnhancedIncreasedIncreasedEnhanced
Cx3cl1 + anti-Cx3cl1 mAbAttenuated--Reduced
Cx3cl1 → RANKLStrongly enhancedRapidly downregulatedMaintained-

What are critical quality control parameters for ensuring reproducible results with Recombinant Mouse Cx3cl1?

To ensure reproducible results when working with Recombinant Mouse Cx3cl1, researchers should verify the following critical parameters:

  • Purity assessment: Confirm protein purity is >97% by SDS-PAGE and/or HPLC analysis to avoid contamination with bacterial proteins that may cause confounding effects .

  • Endotoxin testing: Verify that endotoxin levels are below 0.1 EU/µg of protein using LAL method to prevent non-specific immune activation in cellular assays .

  • Biological activity validation: Confirm specific activity using standardized assays such as:

    • Cell proliferation assays with human peripheral blood lymphocytes (expected ED50 <0.5 µg/ml)

    • Chemotaxis assays using CX3CR1-expressing cells (expected dose-dependent response)

  • Reconstitution protocol adherence: Reconstitute lyophilized protein according to manufacturer specifications to maintain activity:

    • Use sterile buffers (typically PBS, pH 7.4)

    • Avoid excessive vortexing

    • Allow complete reconstitution before use

  • Storage conditions: Maintain proper storage to prevent activity loss:

    • Store lyophilized protein at -20°C to -70°C

    • Store reconstituted protein at 2-8°C for short-term (1 month) or -20°C to -70°C (3 months) for long-term use

    • Avoid repeated freeze-thaw cycles

How can researchers address experimental variability when comparing studies using different Recombinant Mouse Cx3cl1 preparations?

When comparing studies using different Recombinant Mouse Cx3cl1 preparations, researchers should implement the following strategies to address experimental variability:

  • Standardization of activity units: Convert protein concentrations to activity units (IU/mg) based on standardized bioassays to normalize for preparation differences between manufacturers .

  • Internal calibration curves: Generate dose-response curves with each new lot of Recombinant Mouse Cx3cl1 to identify the optimal working concentration for specific experimental systems .

  • Reference standards: Include a well-characterized laboratory reference standard across experiments to normalize results from different commercial preparations .

  • Comprehensive reporting: Document and report the following parameters in publications:

    • Manufacturer and catalog number

    • Lot number and production date

    • Expression system (typically E. coli)

    • Amino acid range included in the recombinant protein

    • Reconstitution buffer composition

    • Storage conditions and duration before use

  • Validation with neutralizing antibodies: Confirm specificity of observed effects by using anti-Cx3cl1 neutralizing antibodies such as Goat Anti-Mouse CX3CL1/Fractalkine Chemokine Domain Antigen Affinity-purified Polyclonal Antibody to block activity in parallel experiments .

Research has shown that the ND50 (neutralization dose) of anti-Cx3cl1 antibodies is typically 0.3-1.5 µg/mL in the presence of 30 ng/mL Recombinant Mouse Cx3cl1 .

How can Recombinant Mouse Cx3cl1 and anti-Cx3cl1 antibodies be used to investigate therapeutic potential in fibrotic disorders?

Recent research reveals promising applications of Recombinant Mouse Cx3cl1 and anti-Cx3cl1 antibodies in investigating potential therapies for fibrotic disorders:

  • Fibroblast response studies:

    • Treat human dermal fibroblasts with Recombinant Mouse Cx3cl1 with or without anti-Cx3cl1 mAb

    • Measure expression of fibrosis markers (type I collagen, fibronectin 1)

    • Studies show that anti-Cx3cl1 mAb treatment significantly inhibits TGF-β1-induced expression of type I collagen and fibronectin 1 in human dermal fibroblasts

  • Multi-model validation approach:

    • Test anti-Cx3cl1 mAb therapy in multiple mouse models of fibrosis:

      • Bleomycin-induced fibrosis

      • Growth factor-induced fibrosis

      • Scleroderma-chronic graft-versus-host disease (Scl-cGVHD)

    • Research demonstrates anti-mouse Cx3cl1 mAb efficiently suppresses skin inflammation and fibrosis across these models

  • Organ-specific effects assessment:

    • Investigate effects on both skin and lung fibrosis

    • Measure inflammatory cell infiltration, collagen deposition, and fibrosis-related gene expression

    • Recent findings indicate anti-Cx3cl1 mAb therapy could be a rational therapeutic approach for fibrotic disorders such as human systemic sclerosis (SSc) and Scl-cGVHD

  • Mechanisms of action studies:

    • Investigate Cx3cl1-induced signaling in target cells using Recombinant Mouse Cx3cl1

    • Determine how anti-Cx3cl1 antibodies interrupt pathological signaling cascades

    • Measure fractalkine levels in tissue extracts to correlate with disease severity

What are the methodological considerations when using transgenic mouse models expressing human CX3CR1 variants in combination with Recombinant Mouse Cx3cl1?

When working with transgenic mouse models expressing human CX3CR1 variants and Recombinant Mouse Cx3cl1, researchers should consider these methodological approaches:

  • Cross-species signaling validation:

    • Confirm responsiveness of human CX3CR1 receptors to mouse fractalkine in vitro using bone marrow-derived macrophages electroporated with human CX3CR1 expression vectors

    • Perform dose-response transmigration assays (1-100 ng/mL mouse Cx3cl1)

    • Research shows the reference human receptor (V249/T280) signals effectively in response to mouse Cx3cl1, while the variant receptor (I249/M280) displays blunted responses

  • Receptor expression confirmation:

    • Validate human CX3CR1 expression at transcript levels in sorted microglial cells from transgenic mice

    • Compare expression levels to wild-type and CX3CR1-knockout controls

    • Studies confirm that mice expressing the human CX3CR1 I249/M280 variant exhibit CX3CR1 mRNA levels comparable to wild-type mice

  • Functional response assessment:

    • Compare cellular responses to Recombinant Mouse Cx3cl1 between wild-type mice, CX3CR1-knockout mice, and human CX3CR1 variant mice

    • Measure parameters such as cell migration, calcium mobilization, and cytokine production

    • Research indicates that human CX3CR1 I249/M280 variant mice show intermediate fractalkine levels in naïve conditions that appear sustained upon EAE induction

  • Disease model phenotyping:

    • Characterize how human CX3CR1 variants respond to mouse Cx3cl1 in various disease models

    • Measure fractalkine levels, inflammatory markers, and tissue-specific pathology

    • Studies show that mice expressing the human CX3CR1 I249/M280 variant exhibit similar defects in CNTF production and neuronal cell loss as CX3CR1-knockout mice during chronic EAE

These methodological considerations enable researchers to use transgenic models to uniquely define the role of human CX3CR1 variants in various disease contexts while leveraging the research utility of Recombinant Mouse Cx3cl1.

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.