Recombinant Rat Growth-regulated alpha protein (Cxcl1) (Active)

Shipped with Ice Packs
In Stock

Description

Cxcl1 exhibits chemotactic activity through CXCR2 binding, with ED₅₀ values of 0.1–0.3 µg/mL for neutrophil migration and 0.2–1 ng/mL for CXCR2 activation . Key functions include:

ActivityMechanismConcentration Range
Neutrophil ChemotaxisCXCR2-mediated migration 10–50 ng/mL
AngiogenesisCXCR2 interaction with endothelial cells N/A
Pain ModulationPotentiates NMDA neurotransmission in astrocytes N/A
Cancer ProgressionHB-EGF cleavage → EGF R transactivation N/A

Research Applications

Cxcl1 is used in diverse experimental models:

ApplicationExperimental ModelKey FindingsSource
BioassaysNeutrophil chemotaxis (BaF3 cells) ED₅₀ = 0.2–1 ng/mL for CXCR2 binding
In Vivo StudiesSpinal cord inflammation (rat) CXCL1 upregulation exacerbates pain
OncologyOvarian cancer cell proliferation CXCL1 → CXCR2 → HB-EGF cleavage
Infectious DiseaseAcinetobacter baumannii pneumonia (rat) CXCL1 as ELISA standard

Regulation and Pathways

Cxcl1 expression is tightly regulated by:

RegulatorMechanismImpactSource
NF-κBBinds promoter region (-94 bp to -84 bp) Induces transcription
IL-17/TNF-αStabilizes CXCL1 mRNA via TRAF6/TAK1 pathway Enhances expression in cancer
Syndecan-1Binds and sequesters CXCL1 until MMP-7 cleavage Controls bioavailability

Key Research Findings

  1. Otitis Media Model: CXCL1 mediates mucosal proliferation in Acinetobacter baumannii infection .

  2. Pain Pathways: Astrocytic CXCL1 enhances NMDA signaling, contributing to chronic pain .

  3. Cancer Metastasis: CXCL1 recruits neutrophils and stromal cells to tumors, promoting angiogenesis .

Product Specs

Buffer
Lyophilized from a 0.2 µm filtered PBS solution, pH 7.4.
Form
Lyophilized powder
Lead Time
5-10 business days
Notes
Repeated freezing and thawing is not recommended. For optimal preservation, store working aliquots at 4°C for up to one week.
Reconstitution
We recommend briefly centrifuging this vial prior to opening to ensure the contents are collected at the bottom. Reconstitute the protein in deionized sterile water to a concentration of 0.1-1.0 mg/mL. For long-term storage, we recommend adding 5-50% glycerol (final concentration) and aliquoting the solution at -20°C/-80°C. Our default final glycerol concentration is 50%, which can serve as a reference for your experiments.
Shelf Life
The shelf life of this product is influenced by various factors, including storage conditions, buffer composition, temperature, and the inherent stability of the protein. Generally, the shelf life of the liquid form is 6 months at -20°C/-80°C. For the lyophilized form, the shelf life is 12 months at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquoting is recommended for multiple use. Avoid repeated freeze-thaw cycles.
Tag Info
Tag-Free
Synonyms
Cxcl1; Cinc1; Gro; Scyb1Growth-regulated alpha protein; C-X-C motif chemokine 1; Cytokine-induced neutrophil chemoattractant 1; CINC-1; Platelet-derived growth factor-inducible protein KC
Datasheet & Coa
Please contact us to get it.
Expression Region
25-96aa
Mol. Weight
7.8 kDa
Protein Length
Full Length of Mature Protein
Purity
>97% as determined by SDS-PAGE.
Research Area
Immunology
Source
E.coli
Species
Rattus norvegicus (Rat)
Target Names
Uniprot No.

Target Background

Function
CXCL1, also known as Growth-regulated alpha protein (GRO), exhibits chemotactic activity for neutrophils. It plays a significant role in neutrophil activation during inflammatory responses.
Gene References Into Functions
  1. Studies have shown that short-term incubation with both CXCL1 and CXCL2 can activate TRPV1+/IB4+ DRG neurons by significantly reducing TRPV1 desensitization. At acute application, both chemokines induce an increase in [Ca2+]i, although with different amplitudes, in a subset of neurons. Notably, CXCL1 partially activated itch-sensitive neurons, while CXCL2 primarily activated non-itch-sensitive neurons. PMID: 28820395
  2. Remote ischemic preconditioning has been shown to protect hepatic microcirculation by inducing HO and modulating cytokine-induced neutrophil chemoattractant (CINC) in hepatic ischemia-reperfusion injury. PMID: 27672274
  3. Results demonstrate that sphingosine-1-phosphate induces Ca(2+) signaling in astrocytes via Gq-coupled receptors S1P2 and S1P3, followed by Ca(2+) influx through TRPC6. This activation pathway potentially leads to MAPK signaling and an increase in the secretion of the proinflammatory or neuroprotective chemokine CXCL1. PMID: 28300348
  4. Aging can reduce the expression of CINC and MCP-1 mRNA in lung tissues, consequently decreasing the infiltration of neutrophils and monocyte-macrophages induced by CINC and MCP-1. This reduction might contribute to an increased risk of pneumonia in elderly patients. PMID: 24464584
  5. In opioid-tolerant rats and patients, CXCL1 is upregulated in the cerebrospinal fluid (CSF). PMID: 25383568
  6. The function of the transient receptor potential vanilloid (TRPV)1 channel is enhanced by dorsal root ganglia inflammation. This effect, characterized by decreased tachyphylaxis, is mimicked by incubation with GRO/KC. PMID: 22466126
  7. CXCL1 inhibition of oligodendrocyte progenitor cell migration is regulated through changes in intracellular calcium flux. PMID: 22554866
  8. Hyperoxia exposure has been found to suppress the expression and activity of HDAC1/HDAC2, along with the overall HDAC activity, in newborn rats. This suppression is accompanied by arrested alveolarization and an elevated expression of CINC-1 in the lungs. PMID: 21905265
  9. Advanced glycation end products (AGEs) promote the secretion of MCP-1 and IL-8 in vascular smooth muscle cells. PMID: 21418993
  10. Severe burn injury activates pulmonary NF-kappaB, leading to the secretion of IL8 and TNF alpha in the lung. PMID: 17959522
  11. In rats with chronic nonbacterial prostatitis, the levels of TNF-alpha and IL-8 in the serum and prostate tissues were significantly lower in the curcumin-treated group compared to the positive control group. PMID: 20180411
  12. ATP-stimulated C6 glioma cells exhibited enhanced expression of MCP-1 & IL-8. Elevated levels of MCP-1 & IL-8 are predicted to enhance tumor cell mobility and promote the recruitment of microglia into developing tumors, thereby supporting tumor growth. PMID: 20003523
  13. Research suggests that IL-6 and CINC-1 contribute to alpha2M production in rats only when they act synergistically. PMID: 21030786
  14. The decreased serum concentration of CINC-1, as a neuroprotective agent, and increased CINC-2alpha in the late stage of Morris hepatoma may be considered for their contribution to cerebellar degeneration. PMID: 20602290
  15. Rats treated with fullerenes showed a transient significant increase in neutrophils and expression of CINC-1,-2alphabeta, and -3. PMID: 20226088
  16. CINC-1, released at sites of inflammation, mediates inflammatory hyperalgesia in rats via the release of sympathomimetic amines. PMID: 12517731
  17. CINC-1 also functions as an early acute-phase protein following injury to the brain or peripheral tissues. PMID: 12709409
  18. Neutrophil migration induced by IL-8 is dependent on the release of CINC-1 from mast cells. PMID: 12824006
  19. Data show that TII alveolar epithelial cells produce three of the major proinflammatory CXC chemokines (GRO, CINC-2alpha, and MIP-2) and their cognate receptor CXCR2. PMID: 12829448
  20. In contrast to MIP-2, CINC is selectively transported from the lung to the systemic circulation, where it promotes neutrophil migration into the lung in response to a chemotactic stimulus. PMID: 14617513
  21. Reactive oxygen species and lipid peroxidation primarily originate from neutrophils, which are stimulated and mobilized by TNF-alpha and cytokine-induced neutrophil chemoattractant-1 (CINC-1). PMID: 15942680
  22. CINC-1 may contribute to liver repair and regeneration. PMID: 16271365
  23. Studies have demonstrated that hyperalgesia, local PGE2 production, and spinal c-Fos expression occur following CFA-induced inflammation, but not after CXCL1- or CXCL2/3-induced, selective PMN recruitment. PMID: 16522746
  24. Results indicate that ascorbic acid deficiency elevates the serum, liver, and spleen concentrations of cytokine-induced neutrophil chemoattractant-1 (CINC-1), as seen in acute inflammation. PMID: 16637227
  25. Following cecal ligation and puncture, CINC-1 production increases in the lung, setting the stage for neutrophil accumulation in the lung during sepsis. PMID: 16818791
  26. Activation of PAR-2, as well as PAR-1, can rescue astrocytes from ceramide-induced apoptosis via regulating chemokine GRO/CINC-1 release. PMID: 16942465
  27. Berberine has been shown to dose-dependently inhibit the expression of CINC-1 induced by LPS and diminish anterior uveitis. PMID: 17164575
  28. TNFalpha plays a role in different forms of arthritis, suggesting that CINC-1 (the homologue for human IL-8) may be a promising target for reactive arthritis management. PMID: 17166735
  29. A basal level of calcium is a prerequisite for GRO/CINC-1 protein synthesis and secretion. PMID: 17666044
  30. Extracellular matrix induces CXCL1, and this induction is further enhanced autocrine by interleukin-1 in rat pancreatic beta-cells. PMID: 17702850
  31. GGA effectively suppressed the up-regulation of GRO1 messenger ribonucleic acid (mRNA), as validated by Northern hybridization. PMID: 18683011
  32. This study provides the first evidence that centrally injected CINC-1 acts directly on the pyrogen-sensitive neurons of the preoptic area, promoting a thermoregulatory response that appears to depend on the synthesis of other endogenous pyrogens and PGE2. PMID: 18694739
  33. Incubation with CXCL1 (1.5 nM, overnight) caused a marked upregulation of Na+ currents in acutely isolated small diameter rat (adult) sensory neurons in vitro. PMID: 18816377
  34. The findings suggest that GRO/KC has important effects in inflammatory processes via its direct actions on sensory neurons. PMID: 19476648
  35. CINC-1 is a neutrophil chemoattractant; studies have established that CINC-1 also functions as an early acute-phase protein following injury to the brain or peripheral tissues. PMID: 12709409

Show More

Hide All

Database Links
Protein Families
Intercrine alpha (chemokine CxC) family
Subcellular Location
Secreted.
Tissue Specificity
At least expressed in the lung and trachea.

Q&A

What is the molecular structure of Recombinant Rat CXCL1 protein?

Recombinant Rat CXCL1/GRO alpha protein is a full-length protein spanning amino acids 25 to 96. The protein belongs to the intercrine alpha (chemokine CxC) family and has the following amino acid sequence: APVANELRCQCLQTVAGIHFKNIQSLKVMPPGPHCTQTEVIATLKNGREACLDPEAPMVQKIVQKMLKGVPK . This sequence is critical for its biological activity and receptor binding. The protein contains conserved CXC motifs characteristic of this chemokine family, with disulfide bonds that are essential for maintaining its tertiary structure and biological function.

What are the primary biological functions of CXCL1 in rat models?

CXCL1 functions primarily as a potent neutrophil chemoattractant in rat models. It has significant chemotactic activity for neutrophils and directly contributes to neutrophil activation during inflammatory processes . CXCL1 mediates the directional immigration of inflammatory cells and is critical in recruiting neutrophils and monocytes/macrophages to target positions such as injured myocardium and arterial walls in cardiovascular disease . This protein can be secreted by various cell types including macrophages, fibroblasts, keratinocytes, and epithelial cells, making it an important mediator in diverse tissue inflammations .

How does recombinant CXCL1 differ from native CXCL1 in experimental applications?

Recombinant Rat CXCL1/GRO alpha protein is produced in Escherichia coli expression systems with high purity (>97%) and low endotoxin levels (<1 EU/μg) . This contrasts with native CXCL1, which may contain varying post-translational modifications depending on the cellular source. The recombinant version offers experimental advantages including:

  • Consistent protein quality and activity across experiments

  • Defined purity levels suitable for in vitro and in vivo applications

  • Well-characterized biological activity for standardized assays

  • Known endotoxin levels to prevent confounding inflammatory responses

What are the recommended protocols for using recombinant CXCL1 in neutrophil chemotaxis assays?

When conducting neutrophil chemotaxis assays with recombinant CXCL1, researchers should follow these methodological considerations:

  • Concentration range: Establish a dose-response curve typically between 1-100 ng/mL of CXCL1

  • Assay format: Use Transwell migration chambers with 3-5μm pore size membranes

  • Cell preparation: Isolate fresh neutrophils from rat blood using density gradient centrifugation

  • Controls: Include both positive controls (fMLP or IL-8) and negative controls (buffer alone)

  • Incubation time: Optimize between 30-120 minutes depending on experimental goals

  • Quantification: Count migrated cells using flow cytometry or microscopy with appropriate staining

The recombinant protein should be reconstituted in sterile buffers without serum to prevent interference, and researchers should verify protein activity using functional assays before experimentation .

How can CXCL1 be utilized in cardiac fibrosis research models?

CXCL1 plays a significant role in cardiac fibrosis research through several experimental approaches:

  • In vivo administration: Recombinant CXCL1 can be administered to induce or exacerbate cardiac inflammation models

  • Neutralization studies: Anti-CXCL1 antibodies can be used to assess the protective effects of CXCL1 inhibition in cardiac remodeling models

  • CXCR2 receptor blockade: CXCR2-specific inhibitors can help distinguish CXCL1-specific effects from other chemokines

  • Transgenic approaches: Genetic models with CXCL1 overexpression or deletion help establish causality in fibrosis development

Research indicates that CXCL1 inhibition protects the heart from angiotensin II-induced inflammation, hypertrophy, and fibrosis. Experimental models using CXCL1 neutralizing antibodies show improved cardiac function, lower levels of brain natriuretic peptide, and reduced hypertrophy compared to control groups . The role of CXCL1 in cardiac fibrosis can be assessed through histological analysis of collagen deposition, echocardiographic measurements of cardiac function, and molecular analysis of fibrotic markers.

What analytical techniques are most appropriate for measuring CXCL1 levels in experimental samples?

Several analytical techniques are appropriate for measuring CXCL1 levels with varying sensitivity and sample requirements:

TechniqueSensitivitySample TypesAdvantagesLimitations
ELISA5-10 pg/mLSerum, plasma, tissue lysatesHigh-throughput, quantitativeLimited dynamic range
Western Blot~50-100 pg/mLTissue/cell lysatesProtein size confirmationSemi-quantitative only
qRT-PCRN/A (mRNA)Tissues, cellsHighly sensitive for gene expressionMeasures mRNA, not protein
SearchLight Proteome Array1-5 pg/mLSerum, plasmaMultiplex detectionSpecialized equipment needed
ImmunohistochemistryQualitativeTissue sectionsSpatial localizationSemi-quantitative
Cytokine Array10-50 pg/mLSerum, culture mediaMultiple cytokine detectionLess quantitative than ELISA

For clinical samples, ELISA remains the gold standard due to its quantitative nature and established reference ranges . For mechanistic studies examining CXCL1 regulation, combined approaches measuring both protein (ELISA/Western) and mRNA (qRT-PCR) provide complementary data on expression control .

How does CXCL1 contribute to cardiac remodeling and fibrosis mechanisms?

CXCL1 contributes to cardiac remodeling and fibrosis through multiple mechanisms:

  • Inflammatory cell recruitment: CXCL1 mediates the infiltration of neutrophils and monocytes/macrophages into injured myocardium, initiating inflammatory cascades

  • Pro-inflammatory signaling: CXCL1 activates CXCR2 receptors on inflammatory and resident cardiac cells, amplifying inflammatory cytokine production

  • Fibroblast activation: CXCL1 can directly or indirectly stimulate cardiac fibroblasts to proliferate and produce extracellular matrix components

  • Oxidative stress induction: Research suggests that CXCL1 may promote cardiac fibrosis through oxidative stress-related pathways

Studies have shown that CXCL1 levels are significantly elevated in patients with cardiac remodeling and fibrosis, particularly in heart failure associated with hypertension . The inhibition of CXCL1 or its receptor CXCR2 results in improved cardiac function and reduced fibrosis in experimental models, suggesting that CXCL1 actively promotes these pathological processes rather than merely serving as a biomarker .

What are the key considerations when designing CXCL1 inhibition studies for cardiovascular disease models?

When designing CXCL1 inhibition studies for cardiovascular disease models, researchers should consider:

  • Inhibition approach selection:

    • Neutralizing antibodies: Specific but may have limited tissue penetration

    • CXCR2 receptor antagonists: Broader effects as they block multiple chemokines

    • Gene silencing approaches: More sustained inhibition but delivery challenges

    • Small molecule inhibitors: Potential for oral administration but may have off-target effects

  • Timing of intervention:

    • Preventive protocols: Administration before disease induction

    • Treatment protocols: Administration after established disease

    • Regression studies: Testing ability to reverse existing fibrosis

  • Appropriate controls and readouts:

    • Cardiac function: Echocardiography, pressure-volume measurements

    • Histological analysis: Collagen deposition, inflammatory cell infiltration

    • Molecular markers: Fibrosis genes, inflammatory cytokines

    • Systemic effects: Monitoring for potential immunosuppression

  • Mechanistic assessments:

    • Signaling pathway analysis: NF-κB activation, MAP kinase activation

    • Cell-specific responses: Effects on cardiomyocytes, fibroblasts, immune cells

    • Temporal dynamics: Time course of CXCL1 expression and inhibition effects

How do CXCL1 levels correlate with clinical outcomes in cardiovascular patients?

Clinical studies have established several important correlations between CXCL1 levels and cardiovascular outcomes:

  • Heart Failure Progression: Elevated serum levels of CXCL1 are observed in patients with cardiac remodeling and fibrosis, particularly in those with hypertension-induced heart failure

  • Diagnostic Value: CXCL1 levels may serve as a biomarker for ongoing cardiac inflammation and fibrosis in various cardiovascular conditions

  • Prognostic Significance: Higher CXCL1 levels appear to correlate with:

    • More severe cardiac dysfunction

    • Greater extent of cardiac remodeling

    • Worse clinical outcomes in heart failure patients

  • Treatment Response: Changes in CXCL1 levels might potentially indicate response to therapies targeting cardiac remodeling

What is the significance of CXCL1 in lung cancer progression and metastasis?

CXCL1 plays significant roles in lung cancer progression and metastasis as evidenced by multiple studies:

How does CXCL1 interact with DACH1 in tumor suppression pathways?

Research has revealed a significant negative correlation between CXCL1 and DACH1, which appears to function as a tumor suppressor:

This reciprocal relationship suggests that DACH1 may exert its tumor suppressive effects partially through the negative regulation of CXCL1, identifying a potential therapeutic approach through targeting this pathway .

How do researchers reconcile contradictory findings regarding CXCL1 regulation by matrix metalloproteinases?

The literature contains apparent contradictions regarding CXCL1 regulation by matrix metalloproteinases (MMPs) that require careful consideration:

  • Contradictory observations:

    • Some studies indicate that MMP-12 inhibits infiltration of neutrophils by suppressing the CXCL1/CXCL2/CXCL5-CXCR2 axis, with MMP12−/− mice showing higher mRNA levels of CXCL1 in infarct areas compared to wild-type controls

    • Conversely, other research suggests that silencing MMP12 leads to decreased CXCL1 levels, implying that MMP12 may actually contribute to CXCL1 secretion

  • Methodological approaches to resolve contradictions:

    • Context specificity: Researchers should carefully control for tissue type, disease model, and experimental timing when evaluating MMP-CXCL1 interactions

    • Comprehensive proteolytic profiling: Examine multiple MMPs simultaneously (MMP-9, MMP-12) to understand compensatory mechanisms

    • Direct vs. indirect effects: Distinguish between direct proteolytic processing of CXCL1 and indirect effects on CXCL1 expression

    • In vitro validation: Conduct enzyme assays with purified components to confirm direct proteolytic activity

  • Experimental design considerations:

    • Use multiple experimental models with appropriate controls

    • Employ both genetic approaches (knockouts) and pharmacological inhibitors

    • Analyze both transcriptional regulation and post-translational processing

    • Conduct time-course studies to capture dynamic regulation

These contradictions highlight the complex regulation of CXCL1 and emphasize the need for comprehensive experimental approaches that consider both direct and indirect regulatory mechanisms .

What are the emerging methodologies for studying CXCL1 in three-dimensional tissue culture models?

Advanced three-dimensional (3D) tissue culture models provide new opportunities for studying CXCL1 biology in more physiologically relevant contexts:

  • Organoid systems:

    • Cardiac organoids can be used to study CXCL1's role in heart development and pathology

    • Lung organoids allow investigation of CXCL1 in respiratory epithelial responses

    • These systems capture cellular heterogeneity and spatial organization lacking in 2D cultures

  • Microfluidic organ-on-chip platforms:

    • Enable real-time observation of neutrophil recruitment in response to CXCL1 gradients

    • Allow co-culture of multiple cell types (e.g., endothelial cells, fibroblasts, immune cells)

    • Can incorporate fluid flow to mimic physiological conditions

  • Bioprinted tissue constructs:

    • 3D bioprinting allows precise spatial positioning of multiple cell types

    • Can incorporate CXCL1-releasing hydrogels to study chemotactic gradients

    • Enables research on CXCL1's role in tissue architecture and remodeling

  • Analytical considerations for 3D models:

    • Employ confocal microscopy for spatial analysis of CXCL1 expression and cellular responses

    • Use microdissection techniques for region-specific analysis of CXCL1 signaling

    • Implement computational modeling to understand CXCL1 gradient formation

These advanced models bridge the gap between traditional cell culture and animal models, providing new insights into CXCL1 biology in complex tissue environments while potentially reducing animal experimentation.

What are the current limitations in translating CXCL1 research from rat models to human applications?

Several important limitations exist when translating CXCL1 research from rat models to human applications:

  • Species-specific differences:

    • Sequence homology: While rat and human CXCL1 share functional similarities, there are structural differences that may affect receptor binding and downstream signaling

    • Expression patterns: The cell types and tissues expressing CXCL1 may differ between species

    • Receptor affinity: Rat CXCL1 may interact differently with CXCR2 receptors compared to human CXCL1

  • Disease model limitations:

    • Cardiac fibrosis models in rats may not fully recapitulate human pathophysiology

    • Cancer models may lack the genetic complexity of human tumors

    • The inflammatory response timeline differs between rats and humans

  • Methodological challenges:

    • Cross-reactivity of antibodies and detection methods between species

    • Differences in pharmacokinetics and pharmacodynamics of CXCL1-targeting therapeutics

    • Limited availability of humanized models for testing CXCL1-targeted therapies

  • Translational strategies:

    • Use of multiple species models (rat, mouse, larger animals) for comparative studies

    • Validation in human tissues and primary cells whenever possible

    • Employment of humanized animal models

    • Correlation of findings with human biospecimens and clinical data

What are the common technical challenges when working with recombinant CXCL1 in experimental systems?

Researchers commonly encounter several technical challenges when working with recombinant CXCL1:

  • Protein stability issues:

    • CXCL1 may lose activity during freeze-thaw cycles

    • Recommendation: Aliquot upon first thaw and avoid repeated freeze-thaw cycles

    • Store at recommended temperatures (-20°C or -80°C) with appropriate protease inhibitors

  • Aggregation problems:

    • CXCL1 can form aggregates under certain buffer conditions

    • Solution: Optimize buffer composition (pH, salt concentration) and include appropriate additives

    • Use filtration or centrifugation to remove aggregates before experiments

  • Activity assay variability:

    • Functional assays for CXCL1 (chemotaxis) can show high variability

    • Standardize neutrophil isolation protocols and use internal controls

    • Include positive controls (known chemotactic factors) for comparison

  • Endotoxin contamination:

    • Even low levels of endotoxin can confound inflammatory experiments

    • Verify endotoxin levels (<1 EU/μg) before in vivo or sensitive in vitro experiments

    • Consider endotoxin removal procedures if necessary

  • Receptor desensitization:

    • Prolonged or high-dose CXCL1 exposure can lead to CXCR2 receptor desensitization

    • Design time-course experiments to account for this phenomenon

    • Consider pulse rather than continuous exposure designs

These challenges can significantly impact experimental outcomes and should be addressed through appropriate controls and optimization steps.

How can researchers differentiate between direct and indirect effects of CXCL1 in complex disease models?

Differentiating between direct and indirect effects of CXCL1 requires systematic experimental approaches:

  • Cell-specific conditional models:

    • Use cell-type specific knockout or overexpression of CXCL1 or CXCR2

    • Employ Cre-lox systems for temporal control of gene expression

    • Compare phenotypes between global and cell-specific manipulations

  • Sequential inhibition studies:

    • Block CXCL1 at different time points in disease progression

    • Analyze effects on downstream pathways to establish causality chains

    • Use combinatorial blocking approaches (e.g., CXCL1 + TNF-α inhibition)

  • Ex vivo approaches:

    • Isolate primary cells from disease models for direct stimulation studies

    • Compare responses in the presence/absence of other cell types

    • Use conditioned media experiments to identify secreted mediators

  • Pathway validation techniques:

    • Pharmacological inhibition of suspected downstream pathways

    • CRISPR-based genome editing to modify specific pathway components

    • Phosphoproteomic analysis to identify direct signaling events after CXCL1 stimulation

  • Advanced imaging approaches:

    • Use intravital microscopy to visualize CXCL1-induced cell recruitment in real-time

    • Implement FRET-based biosensors to detect immediate CXCL1-receptor interactions

    • Apply spatial transcriptomics to map expression patterns in complex tissues

These methodologies help establish causality and mechanistic understanding beyond simple correlations between CXCL1 levels and disease phenotypes .

What are the optimal experimental controls for CXCL1 neutralization studies?

CXCL1 neutralization studies require carefully designed controls to ensure valid interpretation of results:

  • Antibody controls:

    • Isotype-matched control antibodies (same species, isotype, concentration)

    • Heat-inactivated anti-CXCL1 antibodies to control for non-specific effects

    • Fab fragments to eliminate Fc receptor-mediated effects

  • Genetic approach controls:

    • Empty vector controls for overexpression studies

    • Scrambled siRNA/shRNA for knockdown experiments

    • Rescue experiments with exogenous CXCL1 to confirm specificity

  • Pharmacological controls:

    • Vehicle controls matching solvent composition and concentration

    • Dose-response studies to establish specific vs. off-target effects

    • Structurally related but inactive compounds as negative controls

  • Biological validation controls:

    • Alternative methods of CXCL1 inhibition (receptor antagonists, gene silencing)

    • Measurement of known CXCL1-dependent biomarkers to confirm neutralization

    • Analysis of multiple CXCL1-producing and CXCL1-responsive cell types

  • Experimental design controls:

    • Temporal controls (pre-treatment vs. treatment after disease establishment)

    • Administration route controls (systemic vs. local delivery)

    • Random assignment and blinded analysis to reduce bias

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.