GRO g Human, His

GRO-Gamma Human Recombinant (CXCL3), His Tag
Shipped with Ice Packs
In Stock

Description

Molecular Properties

ParameterValueSource
Molecular Mass7,902 Da
Amino Acid Length94 residues (includes a 20-residue His-Tag at the N-terminus)
Source OrganismEscherichia coli
Purification MethodProprietary chromatographic techniques
Formulation- Liquid: 20 mM Tris-HCl buffer (pH 8), 1 mM DTT, 20% glycerol
- Lyophilized: Sterile filtered white powder (no additives)

Key Features:

  • Non-glycosylated: Lacks post-translational modifications due to bacterial expression .

  • His-Tag: Facilitates affinity purification via nickel-chelate chromatography .

Functional and Regulatory Insights

GRO-g (CXCL3) belongs to the CXC chemokine family, which mediates neutrophil activation, inflammation, and tumor growth.

AspectDetailsSource
Gene FamilyShares 86–90% identity with GRO-α (CXCL1) and GRO-β (CXCL2)
mRNA Stability3' untranslated region (UTR) contains ATTTA repeats linked to instability
Regulatory SignalsInduced by IL-1, TNF-α, phorbol esters, and lipopolysaccharides (LPS)
Biological Role- Neutrophil chemotaxis and activation
- Tumor angiogenesis and metastasis

Conservation: A 122-bp region in the 3' UTR is conserved across species, suggesting regulatory importance .

Stability and Handling Guidelines

ParameterRecommendationSource
Storage- Short-term: 10°C for 1 week
- Long-term: ≤ -18°C; avoid freeze-thaw cycles
ReconstitutionDissolve in sterile 18 MΩ-cm H₂O (≥100 µg/mL)
AdditivesAdd 0.1% HSA/BSA to stabilize for prolonged storage

Critical Notes:

  • Lyophilized Form: Reconstitute immediately after thawing to prevent denaturation .

  • Buffer Compatibility: Avoid reducing agents like DTT in downstream applications .

Tissue-Specific Expression

GRO-g is expressed in epithelial cells, fibroblasts, and endothelial cells, with regulation tied to inflammatory stimuli . For example:

  • IL-1/TNF-α: Upregulates GRO-g mRNA within 2–4 hours of exposure.

  • LPS: Induces GRO-g in macrophages, amplifying neutrophil recruitment.

mRNA Stability and Gene Regulation

The 3' UTR of GRO-g contains ATTTA motifs, which recruit RNA-binding proteins (e.g., TTP) to degrade mRNA under stress conditions. This mechanism fine-tunes chemokine production during inflammation .

Therapeutic Implications

GRO-g is implicated in cancer progression and autoimmune diseases. Its His-tagged form enables:

  • Receptor Binding Studies: Elucidating CXCR2 interactions in neutrophil migration.

  • Drug Development: Screening small-molecule inhibitors targeting GRO-g/CXCL3 signaling.

Comparative Analysis of GRO Isoforms

FeatureGRO-α (CXCL1)GRO-β (CXCL2)GRO-γ (CXCL3)
Amino Acid Identity100% (Reference)90%86%
mRNA StabilityModerateLowHigh (ATTTA-rich)
Tumor RoleAngiogenesisMetastasisTumor growth

References and Diverse Sources

  1. Prospec Bio : Detailed production and stability data.

  2. PMC : Gene family analysis, mRNA regulation, and tissue expression.

  3. ScienceDirect : Principles for credible health information (indirect relevance).

Product Specs

Introduction
Chemokine (C-X-C motif) ligand 3 (CXCL3) is a small cytokine known as GRO3 oncogene (GRO3), GRO protein gamma (GROg), and macrophage inflammatory protein-2-beta (MIP2b). It belongs to the CXC chemokine family. CXCL3 regulates monocyte migration and adhesion and exerts its effects by interacting with the CXCR2 chemokine receptor on target cells. The gene encoding CXCL3 is found on chromosome 4, clustered with other CXC chemokine genes.
Description
Recombinant human GRO-Gamma, produced in E. coli, is a single, non-glycosylated polypeptide chain comprising 94 amino acids (35-107). It has a molecular weight of 7902 Daltons. A 20 amino acid His-Tag is fused to the N-terminus of GRO-g, which is purified using proprietary chromatographic methods.
Physical Appearance
A sterile, colorless, and clear solution.
Formulation
The CXCL3 protein is supplied in a buffer consisting of 20mM Tris-HCl (pH 8), 1mM DTT, and 20% glycerol.
Stability
While liquid CXCL3 remains stable at 10°C for one week, it is recommended to store it below -18°C. For long-term storage, adding a carrier protein (0.1% HSA or BSA) is advised. Avoid repeated freeze-thaw cycles.
Purity
Purity is determined to be greater than 95.0% using SDS-PAGE analysis.
Synonyms
Macrophage inflammatory protein 2-beta, MIP2-beta, CXCL3, Growth-regulated protein gamma, GRO-gamma, GRO-gamma(1-73), GRO3, GROg, MIP2B, SCYB3, MIP-2b, CINC-2b, MGSA gamma.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MASVVTELRC QCLQTLQGIH LKNIQSVNVR SPGPHCAQTE VIATLKNGKK ACLNPASPMV QKIIEKILNKGSTN.

Q&A

What is human GRO gamma and how does it relate to other GRO proteins?

Human GRO gamma (also known as CXCL3, MIP-2β, or DCIP-1) is a member of the alpha (C-X-C) subfamily of chemokines. It is one of three distinct GRO proteins (alpha, beta, and gamma) encoded by separate non-allelic genes in humans. GRO gamma shares 86% amino acid sequence homology with GRO alpha, while GRO beta shares 90% homology with GRO alpha . All three human GRO proteins are synthesized as 107 amino acid precursor proteins, from which the N-terminal 34 amino acid residues are cleaved to generate the mature forms . These chemokines serve as potent neutrophil attractants and activators, and can also stimulate basophil activity .

What is the significance of the histidine tag in recombinant GRO γ human protein?

The histidine tag (His-tag) is a sequence of typically six histidine residues added to recombinant GRO gamma protein during expression. This tag serves several crucial research functions:

  • Purification efficiency: The His-tag enables single-step purification using immobilized metal affinity chromatography (IMAC), allowing researchers to isolate the recombinant protein with high purity (>97%) .

  • Detection capability: The tag provides an epitope for antibody recognition, facilitating detection in Western blots and other immunoassays without requiring protein-specific antibodies.

  • Structural studies: For researchers investigating protein structure, the His-tag provides a defined anchor point that can be utilized in various structural biology techniques.

The tag generally has minimal impact on protein folding and biological activity when placed at the N- or C-terminus, making it invaluable for research applications requiring purified protein.

How are GRO genes regulated in human tissues?

GRO gene expression demonstrates complex regulation patterns across different tissues and in response to various stimuli. Expression studies reveal:

  • Tissue-specific regulation: Different tissues show unique baseline and inducible expression patterns for GRO gamma .

  • Induction by inflammatory mediators: GRO expression is inducible by serum, platelet-derived growth factor (PDGF), and various inflammatory mediators including interleukin-1 (IL-1) and tumor necrosis factor (TNF) in monocytes, fibroblasts, melanocytes, and epithelial cells .

  • Regulation via mRNA stability: The 3' untranslated regions of GRO genes contain different numbers of ATTTA repeats associated with mRNA instability, suggesting post-transcriptional regulation differences between the three forms .

  • Conserved regulatory regions: A 122-base-pair region in the 3' region is conserved among all three GRO genes, with part of it also conserved in the Chinese hamster genome, suggesting an important regulatory role .

  • Constitutive expression in tumors: In certain tumor cell lines, GRO proteins are expressed constitutively, independent of inflammatory stimulation .

What experimental approaches are recommended for studying GRO γ signaling pathways?

Investigating GRO gamma signaling requires sophisticated methodological approaches:

  • Receptor binding assays: Use labeled recombinant His-tagged GRO gamma to measure binding kinetics to IL-8 receptor type B (CXCR2) through surface plasmon resonance or radioligand binding assays.

  • Calcium flux measurement: Monitor intracellular calcium mobilization in neutrophils or receptor-transfected cells using fluorescent calcium indicators like Fluo-4 AM after GRO gamma stimulation.

  • Phosphorylation cascade analysis: Employ phospho-specific antibodies to track activation of downstream signaling molecules including MAPKs, PI3K/Akt, and JAK/STAT pathways using Western blot or phospho-flow cytometry.

  • Chemotaxis assays: Utilize Boyden chambers or microfluidic devices to quantify neutrophil migration in response to GRO gamma concentration gradients.

  • Gene expression profiling: Apply RNA-seq or microarray analysis to identify transcriptional changes following GRO gamma treatment, with validation by RT-qPCR for specific targets.

These methodologies should incorporate appropriate controls including heat-inactivated protein, receptor-blocking antibodies, and specific signaling pathway inhibitors.

How can researchers address challenges in differentiating between biological effects of GRO α, β, and γ?

Distinguishing the specific biological effects of highly homologous GRO proteins presents significant challenges. Recommended methodological approaches include:

  • Selective receptor modulation: Utilize receptor-specific antagonists or gene silencing approaches targeting CXCR1 versus CXCR2 to distinguish receptor-mediated effects.

  • Custom antibodies: Develop antibodies targeting unique epitopes within the variable regions of each GRO protein, focusing on the proline/leucine substitution that creates conformational differences between GRO alpha versus beta/gamma .

  • Recombinant protein comparisons: Conduct side-by-side experiments with highly purified recombinant proteins (>97% purity) at equimolar concentrations to directly compare biological activities.

  • Domain swapping: Generate chimeric proteins containing domains from different GRO proteins to map structure-function relationships.

  • Single-cell analysis: Apply single-cell RNA-seq or CyTOF to identify differential cellular responses to each GRO protein in heterogeneous cell populations.

Table 1: Key Structural and Functional Differences Between Human GRO Proteins

FeatureGRO α (CXCL1)GRO β (CXCL2)GRO γ (CXCL3)
Amino acid homology to GRO α100%90%86%
Key amino acid differencesProline at position XLeucine at position XLeucine at position X
Predicted conformational impactReference structureSignificant changeSignificant change
ATTTA repeats in 3' UTRMultiple (highest)IntermediateFewest
Predicted mRNA stabilityLowestIntermediateHighest
Primary receptor bindingCXCR2 > CXCR1CXCR2 > CXCR1CXCR2 > CXCR1
Neutrophil chemotaxis potency+++++++

What are the optimal conditions for expression and purification of biologically active His-tagged GRO γ?

Producing high-quality His-tagged GRO gamma requires careful optimization of expression and purification protocols:

  • Expression system selection:

    • E. coli systems: BL21(DE3) strains with pET vectors typically yield high protein levels, but require refolding protocols for proper disulfide bond formation

    • Mammalian expression: HEK293 or CHO cells provide proper post-translational modifications but at lower yields

    • Insect cell systems: Intermediate option balancing yield with proper folding

  • Induction optimization:

    • For bacterial systems: IPTG concentration (0.1-1.0 mM), temperature (16-37°C), and duration (4-24 hours) should be systematically optimized

    • Lower temperatures (16-25°C) often improve soluble protein yield

  • Purification strategy:

    • Initial capture: Ni-NTA or TALON resin chromatography with imidazole gradient elution

    • Secondary purification: Size exclusion chromatography to remove aggregates

    • Endotoxin removal: Critical for immunological applications, using specialized resins or phase separation techniques

  • Protein refolding considerations:

    • Disulfide bond formation is essential for chemokine activity

    • Controlled oxidation using glutathione redox systems (reduced:oxidized ratio of 10:1)

    • Refolding by dialysis against decreasing concentrations of denaturants

  • Quality control metrics:

    • SDS-PAGE analysis: >97% purity standard

    • Endotoxin levels: <0.1 EU/μg protein

    • Biological activity: Neutrophil chemotaxis assay with EC50 values

    • Mass spectrometry to confirm molecular weight and modifications

How should researchers design experiments to investigate GRO γ differential expression in disease models?

Robust experimental designs for studying GRO gamma expression in disease models should incorporate:

  • Comprehensive sampling approach:

    • Tissue microenvironments: Sample multiple anatomical locations due to heterogeneous expression

    • Temporal dynamics: Include multiple time points to capture expression kinetics

    • Cell type specificity: Use flow cytometry or single-cell approaches to identify cellular sources

  • Appropriate controls:

    • Matched healthy tissues from the same subjects when possible

    • Age/sex-matched controls for human studies

    • Vehicle-treated controls for intervention studies

    • Isotype controls for antibody-based detection methods

  • Quantification methodology:

    • mRNA quantification: RT-qPCR with validated reference genes specific to tissue/condition

    • Protein quantification: ELISA or Western blot with recombinant protein standard curves

    • In situ detection: IHC or RNAscope for spatial distribution analysis

  • Data normalization strategies:

    • For RT-qPCR: Multiple reference gene normalization using geometric averaging

    • For protein quantification: Total protein normalization or housekeeping proteins validated for stability in the disease model

  • Statistical analysis considerations:

    • Power analysis to determine sample size

    • Non-parametric tests for small sample sizes

    • Multiple testing correction for high-dimensional data

    • Correlation with clinical parameters for translational relevance

What strategies can resolve contradictory findings regarding GRO γ function in different experimental systems?

When faced with contradictory findings about GRO gamma function across different experimental systems, researchers should implement these resolution strategies:

  • System-specific variable identification:

    • Systematically document differences in experimental conditions, cell types, animal models, and reagents

    • Create a comprehensive comparison table highlighting key methodological differences

  • Cross-validation approaches:

    • Test hypotheses across multiple model systems (cell lines, primary cells, animal models)

    • Employ both gain-of-function and loss-of-function approaches

    • Utilize complementary methodologies (genetic manipulation, pharmacological inhibition)

  • Receptor competition analysis:

    • Evaluate receptor expression levels in different systems using quantitative approaches

    • Assess competition between GRO proteins for receptor binding

    • Investigate receptor heterodimerization patterns that may differ between systems

  • Contextual signaling evaluation:

    • Analyze signaling pathway activation in a context-dependent manner

    • Measure effects in presence/absence of other inflammatory mediators

    • Evaluate cell-state dependency of responses (naive vs. primed)

  • Collaborative verification:

    • Establish collaborations to test key findings in independent laboratories

    • Exchange reagents, protocols, and cell lines to identify sources of variation

    • Develop consensus protocols for standardized assessment

Table 2: Troubleshooting Guide for Common Discrepancies in GRO γ Research

Discrepancy TypePotential CausesVerification ApproachResolution Strategy
Activity differences between sourcesVariations in protein folding or purityCompare commercial vs. lab-produced proteins in same assayUse proteins with verified bioactivity based on neutrophil chemotaxis
Contradictory signaling resultsCell type-specific signaling networksTest in multiple relevant cell typesMap complete signaling networks with phosphoproteomics
Species-specific differencesEvolutionary divergence in receptor bindingCompare human and mouse systems directlyUse humanized mouse models or human primary cells
Concentration-dependent effectsNon-linear dose responsesPerform detailed dose-response curvesStandardize to physiologically relevant concentrations
Context-dependent outcomesPresence of other inflammatory mediatorsTest in defined cytokine backgroundsDevelop multiparameter models accounting for cytokine networks

How can researchers effectively study the evolutionary conservation and diversification of GRO genes in primate models?

Studying GRO gene evolution in primates requires specialized methodological approaches:

  • Comparative genomic analysis:

    • Utilize dot-plot analysis to visualize sequence homology across primate species

    • Generate phylogenetic trees to establish evolutionary relationships between GRO gene variants

    • Apply bootstrapping analysis to assess statistical support for evolutionary relationships

  • Selective pressure analysis:

    • Calculate dN/dS ratios (non-synonymous to synonymous substitution rates) across gene regions

    • Identify signatures of purifying selection versus diversifying selection

    • Apply PAML or HyPhy software packages for codon-based selection analysis

  • Structure-function correlation:

    • Map sequence variations to protein structural elements

    • Focus on the proline/leucine substitution that creates significant predicted conformational changes between GRO variants

    • Use homology modeling to predict functional impacts of sequence variations

  • Expression pattern comparison:

    • Compare tissue-specific expression patterns across primate species

    • Analyze conservation of regulatory elements, particularly the 122-bp conserved 3' region

    • Investigate conservation of ATTTA repeats that influence mRNA stability

  • Receptor-ligand co-evolution:

    • Study evolutionary patterns of both GRO genes and their cognate receptors

    • Test cross-species receptor activation to identify functional conservation

    • Evaluate whether receptor polymorphisms correlate with ligand variations

The comparative genomic analysis reveals interesting evolutionary patterns: while the δ locus shows high conservation across primate species, the γ locus displays significant divergence, particularly in the group 1 Vγ genes, suggesting different selective pressures between these loci .

What are the methodological considerations for using His-tagged GRO γ in high-throughput drug discovery platforms?

Implementing His-tagged GRO gamma in drug discovery platforms requires careful methodological planning:

  • Assay development considerations:

    • Optimize protein immobilization strategies (direct coupling vs. antibody capture)

    • Validate that His-tag doesn't interfere with binding site accessibility

    • Develop assays with Z' factors >0.5 for screening robustness

    • Include both binding and functional readouts in screening cascades

  • Screening library design:

    • Focus on compound classes known to interact with chemokine-receptor interfaces

    • Include peptidomimetics targeting the chemokine N-terminal region

    • Consider allosteric modulators that may stabilize specific receptor conformations

  • Detection methodology selection:

    • For binding assays: Fluorescence polarization, TR-FRET, or SPR

    • For functional assays: BRET-based receptor activation, β-arrestin recruitment, or calcium flux

    • For cellular assays: High-content imaging of neutrophil migration

  • Data analysis pipeline:

    • Implement machine learning algorithms to identify structure-activity relationships

    • Cluster hits based on chemical scaffolds and mechanism of action

    • Integrate molecular docking to prioritize compounds for follow-up

  • Confirmation strategies:

    • Counter-screen against related chemokines to assess selectivity

    • Evaluate binding to both GRO gamma and receptor

    • Test effects in physiologically relevant cell-based systems

How can single-cell technologies advance our understanding of GRO γ-mediated inflammatory responses?

Single-cell technologies offer powerful approaches to dissect GRO gamma functions:

  • Single-cell RNA sequencing applications:

    • Map cell type-specific responses to GRO gamma stimulation

    • Identify previously unknown target cell populations

    • Characterize transcriptional heterogeneity in responding cells

    • Discover novel GRO gamma-induced gene modules

  • Mass cytometry approaches:

    • Develop CyTOF panels incorporating phospho-specific antibodies for GRO gamma signaling

    • Simultaneously measure 30+ parameters to connect receptor expression with signaling outputs

    • Identify differential responses in neutrophil subpopulations

  • Spatial transcriptomics integration:

    • Map GRO gamma expression and responding cells within tissue microenvironments

    • Correlate with tissue pathology in disease models

    • Identify spatial relationships between GRO gamma-producing and responding cells

  • Technical considerations:

    • Cell fixation/permeabilization protocols must be optimized for chemokine receptor detection

    • Stimulation times require careful optimization due to rapid and transient nature of chemokine responses

    • Data normalization approaches must account for technical variation between single-cell platforms

  • Computational analysis approaches:

    • Trajectory inference to map temporal dynamics of GRO gamma responses

    • Gene regulatory network reconstruction to identify master regulators

    • Integration with spatial data using mathematical modeling approaches

What experimental designs best address the therapeutic potential of targeting GRO γ in inflammatory diseases?

Investigating GRO gamma as a therapeutic target requires systematic experimental approaches:

  • Target validation strategies:

    • Genetic approaches: Conditional knockout models, inducible systems, or CRISPR-mediated deletion

    • Pharmacological approaches: Neutralizing antibodies, receptor antagonists, or aptamers

    • Expression correlation: Comprehensive analysis of GRO gamma levels across disease stages

  • Preclinical model selection:

    • Acute inflammation models: Air pouch, peritonitis, or dermatitis models

    • Chronic inflammation models: Colitis, arthritis, or lung inflammation models

    • Humanized mouse models: Reconstituted with human immune cells for better translation

  • Therapeutic modality evaluation:

    • Compare direct GRO gamma neutralization vs. receptor antagonism

    • Assess small molecule vs. biologic approaches

    • Evaluate tissue-targeted delivery strategies to minimize systemic effects

  • Biomarker development:

    • Identify downstream markers that correlate with GRO gamma pathway inhibition

    • Develop assays to measure target engagement in clinical samples

    • Establish pharmacodynamic markers for dose optimization

  • Safety assessment considerations:

    • Evaluate effects on neutrophil antimicrobial functions

    • Assess compensatory upregulation of other chemokines

    • Monitor for immunosuppression in infection challenge models

Table 3: GRO γ-targeting Therapeutic Approaches and Associated Methodological Considerations

ApproachAdvantagesLimitationsKey ReadoutsSpecial Considerations
Neutralizing antibodiesHigh specificity, long half-lifeLimited tissue penetrationFree GRO γ levels, neutrophil infiltrationPotential immunogenicity
Small molecule CXCR2 antagonistsOral bioavailability, tissue penetrationMay affect signaling by other chemokinesReceptor occupancy, signaling inhibitionReceptor specificity testing required
AptamersTunable half-life, low immunogenicityComplex manufacturingTarget binding, neutrophil migrationStability in biological fluids
siRNA/antisense approachesSustained target reductionDelivery challengesmRNA knockdown efficiencyCarrier system optimization
Gene editingComplete target eliminationOff-target effects, deliveryIndel frequency, protein eliminationEthical considerations for permanent modifications

How can researchers integrate GRO γ studies with systems biology approaches?

Integrating GRO gamma research with systems biology requires methodological sophistication:

  • Multi-omics integration strategies:

    • Combine transcriptomics, proteomics, and metabolomics data from GRO gamma-stimulated systems

    • Develop computational workflows to identify emergent patterns across data types

    • Apply network analysis to position GRO gamma within inflammatory networks

  • Mathematical modeling approaches:

    • Develop ordinary differential equation models of GRO gamma signaling kinetics

    • Create agent-based models of neutrophil migration in response to GRO gamma gradients

    • Implement machine learning algorithms to predict GRO gamma-dependent outcomes from multi-parameter datasets

  • Experimental design considerations:

    • Include comprehensive time course sampling to capture network dynamics

    • Measure multiple outputs simultaneously using multiplexed technologies

    • Systematically perturb network components to validate model predictions

  • Visualization and analysis tools:

    • Implement Cytoscape or similar platforms for network visualization

    • Utilize pathway enrichment approaches that incorporate topological information

    • Develop custom analysis pipelines that integrate public databases with experimental data

  • Collaborative framework development:

    • Establish interdisciplinary teams including immunologists, computational biologists, and mathematicians

    • Develop shared data standards and repositories for GRO gamma research

    • Create accessible tools for researchers without computational expertise

What methodological approaches are recommended for studying post-translational modifications of GRO γ?

Post-translational modifications (PTMs) of GRO gamma require specialized analytical approaches:

  • Mass spectrometry-based identification:

    • High-resolution MS/MS for comprehensive PTM mapping

    • Multiple fragmentation techniques (CID, ETD, HCD) for optimal coverage

    • Enrichment strategies for low-abundance modifications

    • Quantitative approaches (SILAC, TMT) to assess PTM stoichiometry

  • Site-directed mutagenesis validation:

    • Generate point mutations at potential modification sites

    • Compare biological activity of wild-type vs. mutant proteins

    • Create modification-mimicking mutations where applicable

  • Protease protection assays:

    • Assess modification-induced conformational changes through differential protease sensitivity

    • Map protected regions to structural elements

    • Correlate with functional outcomes

  • PTM-specific antibody development:

    • Generate antibodies against specific modified forms

    • Validate specificity using modified and unmodified recombinant proteins

    • Apply in Western blot and immunoprecipitation studies

  • Functional impact assessment:

    • Compare receptor binding kinetics of modified vs. unmodified forms

    • Assess signaling pathway activation differences

    • Evaluate neutrophil chemotaxis potency changes

    • Measure protein stability and half-life alterations

Table 4: Common Post-translational Modifications of Chemokines and Analytical Approaches

Modification TypeDetection MethodFunctional ImpactAnalytical Challenges
CitrullinationMass spectrometry, anti-citrulline antibodiesAltered receptor binding and signalingLow abundance, neutral mass shift
NitrationAnti-nitrotyrosine antibodies, mass spectrometryReduced biological activitySite-specific effects, sample oxidation
Proteolytic processingN-terminal sequencing, mass spectrometryEnhanced or reduced activity depending on siteMultiple cleavage products, dynamic process
GlycosylationLectin binding, mass spectrometryAltered stability and receptor interactionHeterogeneous structures, difficult to analyze
DimerizationNon-reducing SDS-PAGE, crosslinkingModified receptor activation patternsPreserving native interaction during analysis

How can researchers effectively investigate the role of GRO γ in the tumor microenvironment?

Studying GRO gamma in the complex tumor microenvironment (TME) requires specialized approaches:

  • Spatial mapping methodologies:

    • Multiplex immunofluorescence to co-localize GRO gamma with cell type markers

    • Spatial transcriptomics to map expression gradients within tumors

    • 3D reconstruction techniques to visualize chemokine networks in the TME

  • Cell type-specific analysis:

    • Single-cell RNA-seq to identify cellular sources and responders

    • Flow cytometry with intracellular cytokine staining for protein-level confirmation

    • Conditional knockout models to assess cell type-specific contributions

  • Dynamic assessment approaches:

    • Intravital microscopy to visualize neutrophil recruitment in real-time

    • Implantable sensors for continuous monitoring of chemokine levels

    • Serial sampling approaches to track changes during tumor progression

  • Intervention strategies:

    • Genetic manipulation: Cell type-specific deletion using Cre-lox systems

    • Pharmacological: Compare systemic vs. intratumoral delivery of inhibitors

    • Timing considerations: Intervention at different stages of tumor development

  • Translational correlation:

    • Patient sample analysis to correlate findings with clinical outcomes

    • Development of ex vivo tumor explant systems to test modulators

    • Derivation of predictive biomarkers based on GRO gamma pathway activity

The constitutive expression of GRO proteins in certain tumor cell lines suggests an important role in cancer biology that extends beyond inflammatory responses , requiring careful dissection of both tumor-promoting and tumor-suppressing functions in different contexts.

Product Science Overview

Introduction

GRO-Gamma, also known as Chemokine (C-X-C motif) ligand 3 (CXCL3), is a member of the CXC chemokine family. This family of proteins is known for its role in chemotaxis, where they guide the migration of immune cells to sites of inflammation or injury. The recombinant form of GRO-Gamma, tagged with a polyhistidine (His) tag, is produced for research and therapeutic purposes.

Structure and Expression

The recombinant human GRO-Gamma (CXCL3) protein is typically expressed in Escherichia coli (E. coli) systems. The protein consists of 91 amino acids and has a predicted molecular mass of approximately 10.1 kDa . The His tag, usually located at the N-terminus, facilitates purification and detection of the protein.

Biological Function

CXCL3 plays a crucial role in various physiological and pathological processes:

  • Chemotaxis: It attracts neutrophils and basophils to sites of infection or inflammation .
  • Inflammation: CXCL3 is involved in the inflammatory response and can exert its effects on endothelial cells in an autocrine manner .
  • Cancer: It is implicated in the progression and metastasis of certain cancers, including prostate cancer .
  • Adipogenesis: CXCL3 acts as an adipokine, promoting adipogenesis through autocrine and paracrine pathways .
Industrial Production

The production of recombinant GRO-Gamma involves several key steps:

  1. Gene Cloning: The gene encoding CXCL3 is cloned into an expression vector that includes a sequence for the His tag.
  2. Transformation: The vector is introduced into E. coli cells, which are then cultured under conditions that promote protein expression.
  3. Purification: The His-tagged protein is purified using affinity chromatography, which exploits the affinity between the His tag and nickel ions.
  4. Validation: The purity and activity of the recombinant protein are validated using techniques such as SDS-PAGE and functional assays .
Applications

Recombinant GRO-Gamma is used in various research and clinical applications:

  • Cancer Research: Studying the role of CXCL3 in cancer progression and metastasis.
  • Inflammation Studies: Investigating the mechanisms of inflammation and the role of chemokines.
  • Drug Development: Screening potential therapeutic agents that target CXCL3 or its receptor, CXCR2.

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.