Recombinant Mouse C-X-C motif chemokine 5 protein (Cxcl5), partial (Active) is a truncated form of CXCL5, a chemokine belonging to the CXC family. It is engineered to retain biological activity while excluding non-essential regions. CXCL5 plays dual roles in inflammation, acting as both a neutrophil chemoattractant and a regulator of chemokine scavenging. This recombinant protein is widely used in research to study immune regulation, cancer progression, and infectious disease models .
CXCL5 modulates neutrophil trafficking via CXCR2 and DARC (Duffy antigen receptor for chemokines):
Chemokine scavenging: Competes with CXCL1/CXCL2 for DARC binding on erythrocytes, increasing plasma chemokine levels and impairing neutrophil recruitment during severe bacterial infections (e.g., E. coli) .
Neutrophil activation: Truncated forms (e.g., CXCL5(9–78)) enhance chemotaxis in vivo compared to full-length CXCL5 .
CXCL5 is secreted by tumor-associated fibroblasts, macrophages, and mesothelial cells, promoting:
Angiogenesis: via CXCR2-mediated endothelial cell migration .
EMT (Epithelial-Mesenchymal Transition): Activates PI3K/AKT signaling in tumor cells, enhancing metastasis .
In H1N1-infected mice, CXCL5 deficiency:
Reduces early neutrophil influx (innate immunity phase).
Enhances pulmonary antibody production (adaptive immunity phase), with increased local B cell activation .
For optimal reconstitution, the protein vial should be briefly centrifuged prior to opening to bring the contents to the bottom. The lyophilized protein should be reconstituted in deionized sterile water to a concentration of 0.1-1.0 mg/mL . To enhance stability for long-term storage, it is recommended to add glycerol to a final concentration of 5-50%, with 50% being the default concentration used by many manufacturers .
Storage recommendations include:
Store the unopened vial at -20°C/-80°C upon receipt
For multiple use applications, create working aliquots to avoid repeated freeze-thaw cycles
Working aliquots can be stored at 4°C for up to one week
Avoid repeated freeze-thaw cycles as these can damage protein integrity
CXCL5 exhibits a complex and seemingly paradoxical role in neutrophil recruitment during inflammation. Despite being a neutrophil chemoattractant itself, CXCL5 can actually impair neutrophil recruitment to inflammatory sites through its interactions with erythrocyte Duffy Antigen Receptor for Chemokines (DARC) .
The underlying mechanism involves:
CXCL5 binding to erythrocyte DARC, which impairs DARC's ability to scavenge other chemokines from circulation
This results in elevated plasma concentrations of other neutrophil chemoattractants like CXCL1 and CXCL2
The elevated systemic levels of these chemokines leads to:
In a model of E. coli pneumonia, CXCL5-deficient mice demonstrated increased neutrophil influx to the lung and decreased bacterial burden, despite CXCL5's known role as a neutrophil chemoattractant . This counterintuitive finding highlights CXCL5's role in regulating chemokine gradients necessary for efficient neutrophil trafficking to sites of infection.
Calcium flux assays confirmed that neutrophils from CXCL5-deficient mice maintain better CXCR2 responsiveness (with indo-1 violet/blue ratio peaking at 541±27) compared to wild-type mice, supporting the model of CXCR2 desensitization by elevated ligand concentrations in normal conditions .
CXCL5 exerts its biological effects primarily through binding to its receptor CXCR2, activating multiple downstream signaling pathways that regulate various cellular processes:
STAT3 Signaling Pathway:
AKT/PI3K Pathway:
MMP2/9 Regulation:
PXN-AKT-PD-L1 Signaling Cascade:
Functional assays have demonstrated that disruption of CXCL5/CXCR2 signaling significantly suppresses malignant cellular phenotypes including proliferation, clonogenesis, apoptosis resistance, migration, and invasion .
The biological activity of Recombinant Mouse CXCL5 is commonly assessed through neutrophil chemotaxis assays, which provide quantifiable measurements of its functional potency. Below is a methodological approach for conducting these assays:
Neutrophil Chemotaxis Assay Protocol:
Neutrophil Isolation:
Isolate human or mouse neutrophils from peripheral blood using density gradient centrifugation
Verify neutrophil purity (>95%) using flow cytometry with neutrophil markers
Transwell Migration Setup:
Use a modified Boyden chamber with 3-5μm pore size
Add CXCL5 at various concentrations (typically 1-100 ng/ml) to the lower chamber
Add neutrophils (1-5×10^5 cells) to the upper chamber
Activity Measurement:
Incubate for 1-2 hours at 37°C, 5% CO2
Count migrated cells by flow cytometry or microscopy after fixation and staining
Calculate migration index as the ratio of cells migrating toward CXCL5 versus medium control
Verification of Receptor Specificity:
The biological activity of CXCL5 is typically observed in the concentration range of 10-100 ng/ml, with maximal activity often seen at 50 ng/ml . When analyzing results, a dose-response curve should be constructed to determine the EC50 value, which can serve as a measure of potency.
Researchers have employed various experimental approaches to investigate CXCL5's role in disease pathophysiology:
Genetic Manipulation Models:
Therapeutic Administration Studies:
Cellular Functional Assays:
Molecular Signaling Analysis:
Combination Therapy Approaches:
For example, in lupus studies, researchers administered CXCL5 to Fas^lpr mice and monitored various disease parameters including survival, autoantibody levels, proteinuria, nephritis indices, complement deposition, and neutrophil extracellular trap formation .
CXCL5 demonstrates significant therapeutic potential in autoimmune diseases, particularly systemic lupus erythematosus (SLE), through its immunomodulatory effects on multiple pathways:
CXCL5 Levels in Autoimmune Disease:
SLE patients show significantly lower serum CXCL5 levels compared to healthy individuals (p<0.0001)
CXCL5 levels negatively correlate with disease activity (p=0.004)
In lupus-prone Fas^lpr mice, disease severity progression inversely correlates with plasma CXCL5 levels
Therapeutic Effects of CXCL5 Administration:
Intravenous administration of CXCL5 to Fas^lpr mice resulted in:
Restored endogenous circulatory CXCL5 levels
Improved survival rates
Reduced anti-dsDNA antibody titers
Decreased proteinuria
Improved lupus nephritis activity and chronicity indices
Reduced renal complement deposition
Immunomodulatory Mechanisms:
CXCL5 treatment modulates immune responses through:
Regulation of neutrophil trafficking and suppression of neutrophil activation
Reduction of neutrophil degranulation, proliferation, and renal infiltration
Promotion of energy production in renal-infiltrated immune cells
Activation of certain T cell populations
Reduction of tissue fibrosis, granulocyte extravasation, complement components, and interferons
Interestingly, factorial design experiments indicated that CXCL5 may enhance host tolerability to cyclophosphamide in vulnerable individuals, suggesting potential benefits as an adjunctive therapy .
CXCL5 plays a multifaceted role in cancer biology and presents as both a biomarker and potential therapeutic target:
CXCL5 as a Cancer Biomarker:
Elevated serum CXCL5 levels are associated with tumor progression in multiple cancer types
Preoperative serum CXCL5 levels are markedly higher in cancer patients compared to healthy individuals (p=0.001)
CXCL5 expression correlates with its receptor CXCR2 in cancer tissues
Oncogenic Functions of CXCL5/CXCR2 Signaling:
Experimental data from knockdown studies demonstrate that CXCL5/CXCR2 signaling promotes:
Cancer cell proliferation and clonogenic capacity
Resistance to apoptosis
Enhanced migration (measured by wound healing assays)
Increased invasion (measured by transwell invasion assays)
Activation of oncogenic signaling pathways including STAT3 and AKT
Increased MMP2/9 secretion facilitating extracellular matrix degradation
Immune Evasion Mechanisms:
Recent research has uncovered a critical role for CXCL5 in immune evasion through:
Upregulation of PD-L1 expression in cancer cells
Creation of an immunosuppressive tumor microenvironment
Impairment of CD8+ T cell anti-tumor immunity
Mobilization of immunosuppressive neutrophils
Therapeutic Targeting Approaches:
Preclinical studies have shown promising results with:
Anti-CXCL5 antibody therapy
Combination therapy with anti-PD-L1 immune checkpoint inhibitors
Significant tumor growth inhibition in vivo with combination approaches
The dual targeting of CXCL5 and PD-L1 represents a potentially synergistic approach that addresses both tumor cell-intrinsic growth pathways and immune evasion mechanisms.
The structural nuances between CXCL5 and other CXC chemokines contribute significantly to their functional specialization:
Structural Comparison Analysis:
While CXCL5 maintains the canonical chemokine fold similar to CXCL1, CXCL2, CXCL7, and CXCL8, several key structural differences exist:
The organization of β-sheets and α-helices remains conserved across these chemokines
Higher structural variability is observed in the N-terminal residues, N-terminal loop, and 30s turn
These variable regions coincide with functionally important domains and show the largest sequence differences
When superimposing backbone residues (positions 11-76) of CXCL5 with other CXC chemokines:
These structural variations likely contribute to:
Differential receptor binding affinities and specificities
Distinct protein-protein interaction profiles
Varied susceptibility to proteolytic processing
Different oligomerization properties
Specialized functional roles in neutrophil recruitment versus retention
Understanding these structure-function relationships is crucial for rational drug design targeting specific chemokine functions while preserving others.
When designing experiments to investigate CXCL5 in disease models, researchers should address several critical considerations:
1. Proteolytic Processing and Isoform Specificity:
CXCL5 undergoes proteolytic processing after secretion from fibroblasts and epithelial cells
Multiple N-terminal (processed between positions 41-48) and C-terminal (processed between positions 118-132) forms exist
GCP-2(1-78) and GCP-2(9-78) are the most prominent forms, with GCP-2(9-78) showing higher potency
Experimental design should specify which isoform is being studied or measured
2. Expression System Considerations:
CXCL5 produced in different expression systems (E. coli vs. HEK 293) may exhibit different post-translational modifications
These differences can affect biological activity and should be considered when interpreting results
Endotoxin contamination must be minimized (<0.005 EU/μg) to prevent confounding inflammatory effects
3. Receptor Complexity:
While CXCR2 is the primary receptor, CXCL5 may interact with other receptors including atypical chemokine receptors
DARC (Duffy Antigen Receptor for Chemokines) interactions critically affect CXCL5 bioavailability
Experiments should account for receptor expression patterns in the model system studied
4. Context-Dependent Effects:
CXCL5 exhibits apparently contradictory functions in different disease contexts (pro-inflammatory in some, anti-inflammatory in others)
Time-dependent effects should be assessed with both short-term (10 weeks) and long-term (2 years) endpoints
Dose-response relationships should be thoroughly characterized
5. Methodological Approaches:
Combined in vitro and in vivo approaches provide complementary insights
Genetic approaches (knockdown, knockout, overexpression) should be coupled with pharmacological interventions
Appropriate controls for specificity, including receptor antagonists and neutralizing antibodies
Advanced analytical methods including RNA sequencing, mass spectrometry, and multiplexed cytokine analysis provide comprehensive assessment
By addressing these considerations, researchers can design robust experiments that advance our understanding of CXCL5's complex roles in health and disease.
The therapeutic potential of CXCL5 in inflammatory and autoimmune diseases opens several promising research directions:
1. Selective CXCL5 Modulation Strategies:
Development of isoform-specific CXCL5 modulators that target pathogenic functions while preserving beneficial immune surveillance
Design of selective CXCR2 agonists/antagonists that specifically modulate CXCL5 signaling without affecting other ligands
Exploration of biased signaling modulators that activate beneficial downstream pathways while inhibiting harmful ones
2. Precision Medicine Approaches:
Identification of biomarker profiles that predict patient responsiveness to CXCL5-targeted therapies
Development of circulating CXCL5 level assays as companion diagnostics
Patient stratification strategies based on CXCL5/CXCR2 expression patterns
3. Novel Delivery Systems:
Targeted delivery of recombinant CXCL5 to specific tissues using nanoparticle formulations
Cell-based delivery systems using engineered cells programmed to secrete CXCL5 in response to inflammatory cues
Sustained-release formulations to maintain therapeutic CXCL5 levels while minimizing systemic exposure
4. Combination Therapy Optimization:
Further investigation of CXCL5 as an adjunct to standard immunosuppressive therapies
Factorial design studies to identify optimal dosing regimens and drug combinations
Exploration of synergistic combinations targeting multiple aspects of autoimmune pathogenesis
Research priorities should include detailed mechanistic studies to fully elucidate CXCL5's immunomodulatory effects in specific disease contexts, development of more predictive preclinical models, and early-phase clinical studies to establish safety and preliminary efficacy.
Advanced structural biology approaches offer transformative potential for understanding CXCL5-receptor interactions at molecular resolution:
1. Cryo-Electron Microscopy (Cryo-EM) Applications:
Determination of CXCL5-CXCR2 complex structures in different activation states
Visualization of oligomeric assemblies and higher-order signaling complexes
Elucidation of conformational changes during receptor activation and signaling
2. Hydrogen-Deuterium Exchange Mass Spectrometry (HDX-MS):
Mapping protein dynamics and conformational changes upon binding
Identification of allosteric communication networks within the CXCL5-CXCR2 complex
Characterization of binding interfaces with other interaction partners
3. Single-Molecule Fluorescence Resonance Energy Transfer (smFRET):
Real-time monitoring of CXCL5-induced conformational changes in CXCR2
Investigation of the dynamics of receptor activation and signaling
Determination of the kinetics of ligand binding and dissociation
4. Molecular Dynamics Simulations:
In silico modeling of CXCL5-receptor interactions in membrane environments
Prediction of binding energetics and identification of key interaction residues
Virtual screening of potential modulators targeting specific interaction interfaces
5. Structure-Based Drug Design:
Rational design of small molecules targeting specific CXCL5-CXCR2 interaction sites
Development of peptide mimetics based on critical binding epitopes
Creation of bispecific molecules targeting both CXCL5 and complementary pathways