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

Shipped with Ice Packs
In Stock

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 protein in deionized sterile water to a concentration of 0.1-1.0 mg/mL. We recommend adding 5-50% glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our default final concentration of glycerol is 50%. Customers may use this as a reference.
Shelf Life
The shelf life is influenced by various factors, including storage conditions, buffer composition, storage temperature, and the inherent stability of the protein. 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 use. Avoid repeated freeze-thaw cycles.
Tag Info
Tag-Free
Synonyms
Cx3cl1; Acc1; Fkn; Scyd1; Fractalkine; 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
8.8 kDa
Protein Length
Partial
Purity
>95% as determined by SDS-PAGE.
Research Area
Immunology
Source
E.coli
Species
Rattus norvegicus (Rat)
Target Names
Uniprot No.

Target Background

Function
Fractalkine (CX3CL1) is a chemokine that acts as a ligand for both CX3CR1 and integrins ITGAV:ITGB3 and ITGA4:ITGB1. CX3CR1-CX3CL1 signaling exerts distinct functions in different tissue compartments, including immune response, inflammation, cell adhesion, and chemotaxis. It regulates leukocyte adhesion and migration processes at the endothelium. CX3CL1 can activate integrins in a CX3CR1-dependent and CX3CR1-independent manner. In the presence of CX3CR1, it activates integrins by binding to the classical ligand-binding site (site 1) in integrins. In the absence of CX3CR1, it 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 of CX3CL1 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. Activation of STAT3 after muscle incision and retraction mediates the up-regulation of CX3CL1. PMID: 29313211
  2. Basal fractalkine signaling within the trigeminal ganglion plays an important role in mechanical sensitivity of masticatory muscle sensory afferent fibers. Inhibition of CX3CR1 signaling within the trigeminal ganglia may induce analgesia through a peripheral mechanism. PMID: 27209190
  3. Noradrenaline (NA) induces the production of CX3CL1 in neurons. The activation of beta2-adrenergic receptors is involved in this process. Increased mRNA levels and accumulation of CX3CL1 in the cells indicate that NA treatment stimulates neuronal expression and synthesis of CX3CL1. PMID: 27923568
  4. CX3CL1(+) apo-MPs released by apoptotic cells mediate the chemotactic transmigration of alveolar macrophages. PMID: 24603149
  5. CX3CL1/CX3CR1 signaling is involved in LTP of C-fiber-evoked field potentials in the rodent spinal dorsal horn. PMID: 25768734
  6. Up-regulation of CX3CL1 via NF-kappaB-dependent H4 acetylation might be critical for paclitaxel-induced mechanical allodynia. PMID: 25494456
  7. CX3CL1 activates Jak2-Stat5alpha-ERK1/2 signaling through CX3CR1, triggering integrin-dependent machinery reorganization to allow chemotactic migration of bone marrow-derived mesenchymal stem cells towards an ischemic cerebral lesion. PMID: 25559502
  8. CX3CR1 modulates cardiovascular function. PMID: 24036597
  9. This study identifies CX3CL1 signaling through CX3CR1 via the DRG/SC anatomical connection, representing a critical pathway involved in NPP induction in an EAE model of MS. PMID: 24175290
  10. Anti-T-lymphocyte globulin depletes fractalkine, inhibiting migration of dendritic cells and macrophages into the kidney, reducing ischemia-reperfusion injury. PMID: 23953563
  11. Neurotrophic factors expression in host retina receiving CX3CL1-MSCs was stronger than in the retina receiving normal MSCs. Conversely, the expression of proinflammatory factors was downregulated. PMID: 23718544
  12. Astrocytes are required for the neuroprotective activity of CX3CL1 against glucose-induced excitotoxicity. PMID: 23968561
  13. During peripheral inflammation, fractalkine is released in the dorsal root ganglion and contributes to the genesis of inflammatory hypernociception. PMID: 23776243
  14. The present study suggests that fractalkine expressed on endothelial cells and astrocytes are responsible for the recruitment of brain Iba1(+) /NG2(+) cells progenitors during the acute phase of stroke. PMID: 23400803
  15. Our study suggests that CX3CL1/CX3CR1 regulates nerve injury-induced pain hypersensitivity through the ERK5 signaling pathway. PMID: 23361876
  16. Studies examined the effects of cytokines, pathogen-associated molecular patterns, and ethanol on CX3CL1 secretion by PSCs. Ethanol increased CX3CL1 release from PSCs, at least in part through activation of ERK mitogen-activated protein kinase and ADAM17. PMID: 23147224
  17. Fractalkine (FKN) and CX3CR1 were involved in the inflammatory response in cerulein-stimulated AR42J cells. FKN upregulates inflammation through CX3CR1 and the Jak/Stat pathway in severe acute pancreatitis rat models. PMID: 22213034
  18. The fractalkine/CX3CR1 interaction may play an important role in the photoreceptor-microglia cross-talk in light-induced photoreceptor degeneration. PMID: 22536384
  19. Fractalkine and its receptor, CX3CR1, may modulate cardiovascular responses in the solitary nucleus of normal healthy rats via the phosphoinositide 3-kinase intracellular signaling pathway. PMID: 22387113
  20. FKN is involved in the progression of severe acute pancreatitis and might be a valuable marker for the assessment of the pancreatitis. PMID: 22435508
  21. Autologous implantation of nucleus pulposus induces hyperalgesia and allodynia, leading to upregulation of fractalkine and its receptor in spinal neurons and glia, implicating fractalkine in association with radicular pain. PMID: 21224760
  22. Fractalkine promotes myocardial injury and accelerates the progress of heart failure, which is associated with the activation of MAPKs. PMID: 21840883
  23. The results suggest that fractalkine/CX(3)CR1 signaling has a regulatory role in modulating hippocampal neurogenesis via mechanisms that involve indirect modification of the niche environment. PMID: 20018408
  24. Chronic hypoxia stimulates the synthesis and release of fractalkine. PMID: 17953351
  25. A transitory increased expression of fractalkine and its receptor CX3CR1 occurred at the crest time of endotoxin-induced uveitis. This change in expression may play a role in the turnover of lipopolysaccharide-induced acute retina inflammation. PMID: 19648777
  26. TNF-alpha activation of PKCzeta/iota, p42/44 MAPK, c-Jun/AP-1, and p65/NF-kappaB are involved in TNF-alpha-stimulated monocyte fractalkine expression, with soluble fractalkine mediating in part TNF-alpha-induced monocyte transmigration. PMID: 12631113
  27. Data show that Leydig cell expression of fractalkine may be involved in inflammation, whereas fractalkine expressed by germ cells may participate in the cellular interactions between germ cells and other seminiferous tubule cell types. PMID: 12837921
  28. Our data demonstrate that activated mesangial cells are a source for the augmented glomerular CX3CL1/fractalkine expression during the proliferative phase of acute anti-Thy1 glomerulonephritis. PMID: 14605272
  29. Fractalkine expression is induced by TNFalpha in arterial endothelial cells. PMID: 15111313
  30. Fractalkine and CX3CR1 upregulation were demonstrated in an early stage of diabetic kidney. Findings suggest that fractalkine expression and CX3CR1-positive cell infiltration in diabetic kidneys might play a role in the progression of diabetic nephropathy. PMID: 15153757
  31. Production by human airway smooth muscle cells in vitro is regulated by inflammatory and anti-inflammatory factors. PMID: 15321787
  32. Cellular distribution of fractalkine and its receptor in the spinal circuit associated with nociceptive transmission supports a potential role in the mechanisms that contribute to the exaggerated pain state in models of neuropathy. PMID: 15341587
  33. Taken together, these initial investigations of spinal fractalkine effects suggest that exogenous and endogenous fractalkine are involved in spinal sensitization, including that induced by peripheral neuropathy. PMID: 15525271
  34. This selective regulation of CX3CL1 and its receptor, CX3CR1, suggests that these chemokines may represent new targets for the treatment of neuropathic pain. PMID: 15993821
  35. Fractalkine reduces excitotoxicity through activation of the ERK1/2 and PI3K/Akt pathways. It reduces Glu-activated whole-cell current through mechanisms dependent on intracellular Ca2+ and is released from the hippocampus after excitotoxic insult. PMID: 16019082
  36. The sequential expression of fractalkine may direct distinct CX3CR1 receptor-expressing mononuclear cell subsets to inflammatory sites. Fractalkine expression is modulated, at least in part, through the NF-kappaB signaling pathway. PMID: 16030495
  37. High glucose levels, AGE formation, and cytokine activation in diabetes may induce fractalkine upregulation in the kidneys, leading to progression of diabetic nephropathy. PMID: 16234287
  38. ALA, which inhibits the NF-kappaB/fractalkine pathway, may be used to prevent neointimal hyperplasia after angioplasty or stenting. PMID: 16413026
  39. Assessment of fractalkine expression in chronic allograft nephropathy. PMID: 16980051
  40. Data suggest that fractalkine cleavage (release) after nerve injury may play an important role in neural-glial interaction, and microglial CX3CR1/p38 MAPK pathway is critical for the development of neuropathic pain. PMID: 17174525
  41. A genome-wide screen implicates WT1 in the transcriptional regulation of the EGF-family of growth factors as well as the CX3CL1 chemokine during nephrogenesis. PMID: 17430890
  42. Rat hindlimb ischemia, which rapidly stimulated fractalkine expression, was significantly alleviated by the injection of whole-length Fkn protein. PMID: 18006432
  43. The fractalkine/CX3CR1 signaling system plays a potential paracrine/autocrine role in preovulatory follicles. PMID: 18292196
  44. There is greater fractalkine expression in mesenteric arteries of female spontaneously hypertensive rats compared with males. PMID: 19201996
  45. In an experimental model of atrial inflammation, macrophages infiltrated the myocardium mainly through the atrial endocardium with the contribution of fractalkine. PMID: 19282612
  46. Gene expression profiling of Cx3cl1 in bone marrow mesenchymal cells by osteogenic induction is reported. PMID: 19422292
  47. Both soluble and membrane-bound fractalkine attenuate lipopolysaccharide (LPS)-induced microglial activation. PMID: 19627440

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 (neurons). Significant levels in kidney, heart, lung and adrenal gland.

Q&A

What is the molecular structure of rat CX3CL1/Fractalkine?

Rat CX3CL1/Fractalkine is a type 1 membrane protein belonging to the delta chemokine subfamily, containing a novel C-X3-C motif. The rat CX3CL1 cDNA encodes a 393 amino acid residue precursor protein with two alternative (21 aa or 24 aa residue) putative signal peptides, a 74-76 aa residue globular chemokine domain, a 238 aa residue stalk region rich in Gly, Pro, Ser, and Thr (containing degenerate mucin-like repeats), a 19 aa residue transmembrane segment, and a 36 aa residue cytoplasmic domain. This structure distinguishes CX3CL1 from other chemokines, as it features a chemokine domain tethered on a long mucin-like stalk .

Where is CX3CL1 primarily expressed in rat tissues?

CX3CL1 is expressed in various rat tissues including heart, brain, lung, kidney, skeletal muscle, and testis. In rat brain specifically, CX3CL1 expression is localized principally to neurons. Its expression can also be upregulated on activated endothelial cells. Immunofluorescence studies confirm that fractalkine is primarily expressed by neurons, although subsets of other cell types, including astrocytes, occasionally display fractalkine immunoreactivity .

What are the functional forms of CX3CL1/Fractalkine?

CX3CL1/Fractalkine exists in two functional forms:

  • Membrane-bound form: Acts as an adhesion molecule that promotes leukocyte adhesion

  • Soluble form: Generated by proteolytic cleavage at the conserved dibasic motif proximal to the transmembrane region. The soluble chemokine domain functions as a chemoattractant for T cells, monocytes, and in mouse models, neutrophils and T-lymphocytes

How can I assess the biological activity of recombinant rat CX3CL1 in vitro?

The biological activity of recombinant rat CX3CL1 can be assessed through chemotaxis assays. A validated approach uses the BaF3 mouse pro-B cell line transfected with human CX3CR1. In this assay:

  • Plate transfected BaF3 cells in the upper chamber of a chemotaxis system

  • Add recombinant rat CX3CL1 at varying concentrations (e.g., 0-100 ng/mL) to the lower chamber

  • Allow migration for 2-4 hours at 37°C

  • Quantify migrated cells using Resazurin or other cell viability indicators

  • Plot dose-dependent chemotactic response

The activity can be confirmed by neutralization with anti-CX3CL1 antibodies, where the neutralizing dose (ND50) is typically 0.3-1.2 μg/mL for specific antibodies like Goat Anti-Rat CX3CL1/Fractalkine Chemokine Domain Antigen Affinity-purified Polyclonal Antibody .

What methods are recommended for detecting CX3CL1 expression in brain tissue?

Multiple complementary techniques can be employed for detecting CX3CL1 expression in brain tissue:

  • Immunohistochemistry/Immunofluorescence:

    • Use specific antibodies (e.g., Goat Anti-Rat CX3CL1/Fractalkine)

    • For frozen sections, fix with paraformaldehyde, incubate with primary antibody (1-2 μg/mL) overnight at 4°C

    • Visualize using appropriate secondary antibodies coupled with fluorochromes or HRP-DAB detection systems

    • Counterstain with nuclear markers like DAPI or hematoxylin

  • Western Blot Analysis:

    • Extract proteins from brain tissue using standard protocols

    • Separate proteins by SDS-PAGE and transfer to membranes

    • Probe with anti-CX3CL1 antibodies

    • Quantify relative expression normalized to housekeeping proteins

  • ELISA:

    • Homogenize brain tissue samples

    • Measure soluble CX3CL1 concentrations using sandwich ELISA

    • Compare levels between experimental groups or brain regions

How can I establish a CX3CL1-expressing experimental model?

To establish an experimental model expressing CX3CL1, consider these approaches:

  • Cell line-based models:

    • Select appropriate cell lines that either naturally express CX3CL1 (like NCI-H630) or can be transfected (like RKO-CX3CL1)

    • Confirm expression using ELISA of cell culture supernatants or cell lysates

    • Validate functional activity using chemotaxis assays

  • Animal models:

    • Utilize transgenic approaches to modify CX3CL1 expression

    • Consider conditional knockout or overexpression models to study tissue-specific effects

    • For tumor models, implant CX3CL1-expressing cells and confirm expression using ELISA of tumor lysates

Note that established cancer cell lines frequently lose the ability to produce specific chemokines including Fractalkine, so expression validation is essential .

How does fractalkine signaling affect microglial physiology in the CNS?

Fractalkine signaling through the CX3CL1-CX3CR1 axis regulates multiple aspects of microglial physiology:

  • Microglial Motility and Migration:

    • Fractalkine exerts a strong, dose-dependent migratory effect on microglia

    • Application triggers calcium mobilization within minutes, leading to cytoskeletal rearrangement and migration

    • This response depends on CX3CR1 and G-protein coupled signaling (inhibited by G-protein inhibitors)

  • Surveillance Behavior:

    • Fractalkine normalizes microglial motility that is typically elevated by inflammatory stimuli

    • Properly regulated motility limits excessive microglial surveillance that may lead to neurotoxic effects

  • Inflammatory Response Regulation:

    • Serves as a critical communication link between neurons and microglia

    • Helps maintain microglial homeostasis under physiological conditions

    • Disruption of fractalkine signaling can alter microglial phenotype and responsiveness

What roles does CX3CL1-CX3CR1 signaling play in synaptic plasticity?

CX3CL1-CX3CR1 signaling modulates several aspects of synaptic plasticity:

These effects highlight the complex role of fractalkine signaling in regulating synaptic strength and plasticity in an age-dependent manner .

How can recombinant CX3CL1 be used to improve T cell recruitment in immunotherapy models?

Recombinant CX3CL1 can enhance T cell recruitment in immunotherapy through several strategies:

  • Engineering T cells to express CX3CR1:

    • Ectopic expression of CX3CR1 enhances the homing of adoptively transferred T cells toward CX3CL1-producing tumors

    • This approach capitalizes on the natural chemotactic properties of CX3CL1

  • Tumor microenvironment targeting:

    • Select or engineer tumor models that express CX3CL1 (either naturally or through genetic modification)

    • Compare tumor lysates from CX3CL1-expressing tumors with normal tissue lysates using ELISA to confirm expression levels

    • RKO-CX3CL1 engineered cells, for example, produce significantly higher amounts of Fractalkine compared to control tissues

  • Experimental validation:

    • Measure T cell infiltration using flow cytometry or immunohistochemistry

    • Assess functional responses including tumor growth inhibition and T cell activation markers

    • Compare recruitment efficiency between CX3CR1-positive and CX3CR1-negative T cells

How does fractalkine signaling interact with opioid systems in neuroinflammatory conditions?

Fractalkine signaling demonstrates complex interactions with opioid systems during neuroinflammatory conditions:

  • Neuroprotective Effects:

    • Fractalkine can protect striatal neurons from synergistic morphine and HIV-1 Tat toxicity

    • Disruption of fractalkine-CX3CR1 signaling mimics the toxic effects of morphine plus Tat exposure

  • Microglial Regulation:

    • Exogenous fractalkine normalizes microglial motility that is elevated by Tat and morphine co-exposure

    • This normalized motility may limit excessive microglial surveillance that can lead to neurotoxic effects

  • Cellular Dependency:

    • The protective effects of fractalkine require proper CX3CR1 expression on microglia

    • Fractalkine fails to protect wild-type neurons co-cultured with CX3CR1-null glia against morphine and Tat toxicity

These findings suggest a critical role for fractalkine-CX3CR1 signaling in modulating the toxic effects of opioids during neuroinflammatory conditions, with potential implications for understanding HIV-associated neurocognitive disorders in opioid users .

What are the differences in fractalkine signaling between species and their implications for translational research?

Important species differences in fractalkine signaling include:

  • Structural Homology:

    • Rat CX3CL1 shares a high degree of amino acid sequence homology (83% sequence identity) with human and mouse CX3CL1, with the exception of the stalk region

    • This conservation suggests core functional preservation across species

  • Chemotactic Specificity:

    • Human soluble fractalkine is chemotactic for T cells and monocytes

    • Mouse soluble fractalkine attracts neutrophils and T-lymphocytes but not monocytes

    • Rat fractalkine can chemoattract BaF3 cells transfected with human CX3CR1, indicating cross-species receptor activation

  • Experimental Design Implications:

    • When designing experiments, consider species-specific differences in target cell populations

    • For neutralization studies, confirm cross-reactivity of antibodies between species

    • In translational research, validate findings across species before extrapolating to human applications

  • Transgenic Model Considerations:

    • CX3CR1-GFP reporter mice and CX3CR1 knockout models have been instrumental in revealing fractalkine functions

    • When using these models, consider that complete ablation may not reflect the more nuanced alterations in human pathologies

How do changes in CX3CL1-CX3CR1 expression correlate with neurological disorders?

Recent research provides insights into CX3CL1-CX3CR1 expression changes in neurological conditions:

  • Vascular Cognitive Impairment:

    • In 2VO rat models (bilateral common carotid artery occlusion), there is upregulation of both CX3CL1 and CX3CR1 in the hippocampus

    • This upregulation is detectable by both immunofluorescence staining and western blot analysis

    • Expression changes are quantifiable, showing statistically significant increases (P < 0.05) compared to sham controls

  • MicroRNA Regulation:

    • Loss-of-function of miR-195 upregulates the expression of both CX3CL1 and CX3CR1 in rat hippocampus

    • This regulatory relationship suggests potential therapeutic approaches targeting microRNA pathways

  • Relationship to Inflammation:

    • CX3CL1-CX3CR1 expression changes often correlate with altered inflammatory states

    • These changes may be either compensatory (neuroprotective) or contributory to pathology, depending on the context and timing

  • Quantitative Analysis:

    • Expression changes can be quantified using western blot followed by densitometry

    • Results should be normalized to appropriate housekeeping proteins and analyzed using appropriate statistical tests (e.g., Student's t-test for two-group comparisons or ANOVA followed by Tukey test for multiple comparisons)

What are common challenges in detecting CX3CL1 in experimental samples and how can they be addressed?

Researchers frequently encounter several challenges when detecting CX3CL1:

  • Low Expression Levels:

    • Challenge: Baseline CX3CL1 expression can be low in certain tissues or cell types

    • Solution: Use sensitive detection methods such as amplified ELISA systems or chemiluminescent western blots; consider concentrating samples through immunoprecipitation before analysis

  • Proteolytic Processing:

    • Challenge: CX3CL1 exists in both membrane-bound and soluble forms due to proteolytic cleavage

    • Solution: Include protease inhibitors in all extraction buffers; design detection strategies that can distinguish between full-length and cleaved forms; consider using domain-specific antibodies

  • Cross-Reactivity:

    • Challenge: Antibodies may cross-react with related chemokines

    • Solution: Validate antibody specificity using positive and negative controls; consider using preabsorption controls to confirm specificity as mentioned in the research literature

  • Cell Culture Considerations:

    • Challenge: Established cell lines often lose chemokine expression during passaging

    • Solution: Regularly verify expression levels; consider using early passage cells or primary cultures; validate findings using multiple detection methods

How can I optimize the functional analysis of CX3CL1-CX3CR1 interactions in neuronal-microglial co-cultures?

For optimal analysis of CX3CL1-CX3CR1 interactions in co-culture systems:

  • Co-culture System Establishment:

    • Use primary neurons from wild-type animals co-cultured with microglia from either wild-type or CX3CR1-knockout mice

    • Alternatively, use transgenic reporter systems like CX3CR1-GFP to facilitate visualization

    • Control the ratio of microglia to neurons (typically 1:5 to 1:10) to mimic physiological conditions

  • Experimental Manipulations:

    • Apply recombinant CX3CL1 at physiologically relevant concentrations (10-100 ng/mL)

    • Use CX3CL1 neutralizing antibodies as negative controls

    • Consider using recombinant CX3CL1 with mutations in key functional domains to identify structure-function relationships

  • Multi-parameter Analysis:

    • Combine live-cell imaging to assess microglial dynamics and morphology

    • Use calcium imaging to assess real-time responses to CX3CL1

    • Collect conditioned media for cytokine/chemokine profiling

    • Perform post-experiment immunocytochemistry to assess marker expression

  • Controls and Validation:

    • Include genotype-matched controls (wild-type vs. CX3CR1-knockout)

    • Use receptor antagonists or function-blocking antibodies as pharmacological controls

    • Validate key findings with in vivo experiments when possible

What emerging applications of CX3CL1 are being explored in neurodegenerative disease models?

Emerging applications of CX3CL1 in neurodegenerative disease research include:

  • Biomarker Development:

    • Soluble CX3CL1 levels in cerebrospinal fluid or plasma may serve as biomarkers for neuroinflammatory states

    • Changes in the CX3CL1/CX3CR1 axis could potentially predict disease progression or treatment response

  • Therapeutic Modulation:

    • Recombinant CX3CL1 or mimetic peptides might serve as neuroprotective agents

    • Targeting the proteolytic processing of CX3CL1 could regulate its availability and function

    • Age-dependent effects of CX3CL1 signaling suggest potential for stage-specific interventions

  • Neurogenesis and Neural Circuit Repair:

    • CX3CL1's role in hippocampal neurogenesis could be leveraged for promoting recovery after injury

    • The influence on synaptic pruning, maturation, and plasticity makes it a potential target for enhancing neural circuit repair

  • Combined Therapeutic Approaches:

    • Integration with other neuroimmune modulators or growth factors might provide synergistic effects

    • Understanding interactions with other signaling systems (like opioid pathways) could lead to novel treatment strategies for complex neurological conditions

How might advances in protein engineering enhance the therapeutic potential of modified CX3CL1 proteins?

Protein engineering approaches offer several avenues to enhance CX3CL1's therapeutic potential:

  • Domain-Specific Modifications:

    • Engineering the chemokine domain for enhanced receptor binding or specificity

    • Modifying the mucin-like stalk for altered proteolytic processing or half-life

    • Creating chimeric proteins combining functional domains from different chemokines

  • Delivery System Integration:

    • Fusion with antibody fragments for targeted delivery to specific brain regions

    • Incorporation into nanoparticles or exosomes for enhanced blood-brain barrier penetration

    • Development of controlled-release formulations for sustained local effects

  • Functional Optimization:

    • Creating variants with enhanced stability in inflammatory environments

    • Developing mutants that selectively activate beneficial signaling pathways while minimizing detrimental ones

    • Engineering pH-sensitive variants that activate only under specific pathological conditions

  • Translational Considerations:

    • Humanized variants of rat CX3CL1 preserving key epitopes but reducing immunogenicity

    • Species-optimized versions accounting for the documented differences in cellular targeting between species

    • Simplified structural analogs retaining essential functional elements while reducing manufacturing complexity

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.