Recombinant RANTES is typically expressed in Escherichia coli, followed by refolding and purification using immobilized metal affinity chromatography (IMAC) via the His tag . Key quality benchmarks include:
Bioactivity: Induces monocyte chemotaxis at 1–10 ng/mL (specific activity: 100,000–1,000,000 IU/mg) .
Stability: Lyophilized form remains stable at -18°C; reconstituted in 50 mM NaAc buffer (pH 5.0) .
RANTES interacts with receptors CCR1, CCR3, CCR4, and CCR5 to mediate diverse immune responses :
Chemotaxis: Recruits monocytes, T cells, eosinophils, and basophils to inflammation sites .
HIV Suppression: Blocks CCR5-mediated entry of R5-tropic HIV-1 strains .
Immune Modulation: Activates eosinophils, induces histamine release from basophils, and promotes NK cell proliferation .
Notably, RANTES self-aggregation and glycosaminoglycan binding modulate its dual role in HIV inhibition and enhancement .
Glomerulonephritis (GN):
Parameter | Met-RANTES | AOP-RANTES | Control |
---|---|---|---|
Glomerular Macrophages | ↓ 50% | ↓ 45% | Baseline |
iNOS Expression | ↑ 3.5-fold | ↑ 3.2-fold | Baseline |
Humoral Immune Response | Unaffected | Unaffected | Baseline |
RANTES potentiates basal glutamate release in human neocortex nerve terminals but inhibits depolarization-evoked release via pertussis toxin-sensitive pathways .
Melanoma-secreted RANTES correlates with enhanced tumor growth in nude mice, recruiting monocytes and dendritic cells to tumor sites .
HIV Therapy: CCR5 antagonism by RANTES analogs is explored for preventing viral entry .
Autoimmune Diseases: Antagonists like Met-RANTES show mixed outcomes, reducing inflammation but exacerbating tissue damage in GN models .
Obesity: Murine studies link elevated RANTES/CCR5 levels in adipose tissue to diet-induced obesity .
RANTES (CCL5) is a chemokine that serves as a chemoattractant for various leukocytes, recruiting them to sites of inflammation. It plays a critical role in immune surveillance and inflammatory responses by facilitating the migration of T cells, macrophages, eosinophils, and basophils .
Methodological approach: When investigating RANTES function, researchers typically employ chemotaxis assays with Boyden chambers to quantify cell migration. These assays involve placing RANTES in the lower chamber and measuring the migration of leukocytes from the upper chamber. Flow cytometry analysis of receptor expression before and after RANTES exposure can provide insights into activation states of responding cells.
His-tagged RANTES should be reconstituted in sterile, deionized water or appropriate buffer depending on the downstream application.
Methodological approach: For optimal stability, reconstituted RANTES should be aliquoted to avoid repeated freeze-thaw cycles, which can lead to protein degradation. For short-term storage (1-2 weeks), reconstituted protein can be kept at 4°C; for long-term storage, maintain at -20°C or -80°C. Prior to use, verify protein integrity through Western blot or functional assays to ensure the His-tag hasn't altered native protein function.
The addition of a His-tag to recombinant RANTES facilitates protein purification but may potentially affect certain protein functions.
Methodological approach: Researchers should conduct comparative analyses between His-tagged and untagged RANTES, including:
Receptor binding assays to assess if the tag affects binding affinity
Chemotaxis assays to compare functional potency
Structural analyses using circular dichroism to evaluate potential conformational changes
Dimerization studies, as His-tags may influence the oligomerization properties of RANTES
RANTES plays a complex role in the tumor microenvironment (TME) of colorectal cancer (CRC). Research indicates significantly higher levels of RANTES in tumor tissues compared to margin tissues, suggesting its importance in cancer progression .
Methodological approach: To investigate RANTES in the TME, researchers should:
Compare RANTES expression between tumor and margin tissues using ELISA or immunohistochemistry
Assess correlation with other inflammatory markers including PD-L1, IFN-γ, TNF-α, and TGF-β
Examine the spatial distribution of RANTES-expressing cells relative to tumor cells using multiplex immunofluorescence
Analyze the relationship between RANTES levels and tumor-infiltrating lymphocytes (TILs)
RANTES tumor levels correlate significantly with angiogenesis factors such as VEGF-A and VEGF-C, suggesting its role in promoting blood vessel and lymphatic vessel formation in tumors .
Multiplex protein assays to simultaneously measure RANTES, VEGF-A, and VEGF-C
Co-localization studies using immunofluorescence microscopy
Microvessel density (MVD) quantification in tissue sections with high versus low RANTES expression
In vitro endothelial tube formation assays using conditioned media from RANTES-stimulated cells
Statistical analysis using multivariate modeling to control for confounding factors
Research contradictions are common in RANTES studies due to differences in experimental conditions, cell types, and methodologies.
Methodological approach: Implement a systematic framework using nanopublications to detect contradictions:
Represent research claims in a standardized format that includes assertions and provenance data
Employ reasoning over nanopublication assertion and provenance graphs to identify contradictory claims
Analyze the sources of contradictions based on experimental characteristics
This approach performs better for categorical characteristics than numerical ones, providing a method to identify contradictions even with limited studies .
Proper controls are critical for reliable RANTES research in cancer tissues.
Always include paired tumor and margin tissue samples from the same patient
Use technical replicates to ensure measurement reliability
Include positive controls (tissues known to express RANTES) and negative controls (tissues with minimal RANTES expression)
For immunostaining, include isotype controls to account for non-specific binding
When analyzing RANTES-expressing cells, include phenotypic markers to identify cell types (e.g., CD3 for T cells, CD68 for macrophages)
RANTES shows correlations with immune checkpoint molecules like PD-L1, suggesting potential roles in immune escape mechanisms in cancer .
Design co-culture experiments with tumor cells and immune cells in the presence/absence of RANTES
Measure changes in PD-L1 expression after RANTES stimulation using flow cytometry or Western blotting
Use RANTES receptor antagonists to confirm specificity of effects
Perform ChIP assays to examine whether RANTES signaling directly affects PD-L1 promoter activity
Validate findings in mouse models using RANTES knockout or receptor blockade approaches
Experimental settings significantly impact RANTES study outcomes, and careful consideration of these variables is essential for proper interpretation.
Catalog key experimental variables including:
Cell/tissue types (primary cells vs. cell lines, tissue of origin)
Species differences (human vs. mouse models)
Concentration ranges of RANTES used
Duration of exposure
Presence of other cytokines or serum components
Create standardized reporting templates to facilitate cross-study comparisons
Consider meta-regression analyses to identify variables that moderate effect sizes
Proper statistical analysis is crucial for understanding RANTES relationships with other inflammatory markers.
Begin with correlation analyses (Pearson or Spearman, depending on data distribution)
Progress to multivariate analyses to control for confounding factors
Consider hierarchical clustering to identify patterns of co-expression
Use principal component analysis to reduce dimensionality when examining multiple cytokines
Implement mediation analysis to explore causal relationships
Cytokine/Factor | Correlation with RANTES in Tumor | Correlation with RANTES in Margin | Potential Functional Implication |
---|---|---|---|
PD-L1 | Significant positive | Significant positive | Immune escape mechanisms |
IFN-γ | Not specified | Significant positive | Inflammatory response regulation |
TNF-α | Significant positive | Significant positive | Pro-inflammatory signaling |
TGF-β | Significant positive | Significant positive | Immunosuppression, EMT |
VEGF-A | Significant positive | Significant positive | Angiogenesis |
VEGF-C | Significant positive | Significant positive | Lymphangiogenesis |
This data suggests RANTES may play a multifaceted role in modulating both immune responses and vascular formation in colorectal cancer .
RANTES studies often yield seemingly contradictory results due to context-dependent functions.
Systematically compare experimental conditions between contradictory studies
Consider cell-specific effects (different cell types may respond differently to RANTES)
Examine concentration-dependent effects (RANTES may have biphasic effects)
Analyze temporal factors (acute vs. chronic exposure)
Create a formal contradiction analysis framework that includes:
Translating RANTES findings from in vitro to in vivo settings requires careful consideration of physiological complexity.
Implement a tiered experimental approach:
Begin with purified recombinant proteins in simple systems
Progress to co-culture systems with multiple cell types
Advance to 3D organoid models that better recapitulate tissue architecture
Validate in appropriate animal models with humanized components when possible
Compare RANTES concentrations used in vitro with those measured in relevant biological fluids
Consider the impact of the extracellular matrix, which is often absent in vitro but may influence RANTES gradient formation and receptor binding in vivo
Understanding RANTES in immune escape presents several methodological challenges.
Develop more physiologically relevant models that incorporate multiple cell types
Use single-cell approaches to capture heterogeneity in responses
Implement dynamic imaging to track cellular interactions in real-time
Consider the impact of other chemokines that may compete for the same receptors
Develop conditional knockout models to study cell-specific effects of RANTES in immune escape
RANTES expression and function vary considerably across cancer types and even within individual tumors.
Perform comprehensive tissue microarray analyses across cancer types
Integrate genomic, transcriptomic, and proteomic data to identify factors regulating RANTES expression
Use single-cell RNA sequencing to map heterogeneity within tumors
Correlate RANTES expression patterns with clinical outcomes to assess prognostic significance
Develop cancer-type specific scoring systems for RANTES expression and activity
Several emerging technologies promise to enhance RANTES research.
CRISPR-Cas9 gene editing to create precise modifications in RANTES or its receptors
Optogenetic approaches to achieve temporal control of RANTES signaling
Advanced imaging techniques such as intravital microscopy to visualize RANTES-mediated cell recruitment in vivo
Computational modeling of chemokine gradients and cellular responses
Machine learning algorithms to identify patterns in large datasets that may reveal novel aspects of RANTES biology
RANTES, also known as CCL5 (C-C motif chemokine ligand 5), is a member of the chemokine family. Chemokines are small cytokines or signaling proteins secreted by cells, and they play a crucial role in immune responses by directing the migration of immune cells to sites of inflammation or injury . RANTES is classified as a beta-chemokine due to its structure and function .
RANTES is an 8 kDa protein that is chemotactic for T cells, eosinophils, and basophils . It plays a significant role in recruiting leukocytes to inflammatory sites, thereby contributing to the body’s immune response . The recombinant form of RANTES, tagged with a His (histidine) tag, is produced using recombinant DNA technology, which allows for the expression of the protein in host cells such as E. coli or HEK293 cells .
RANTES plays a pivotal role in the inflammatory immune response. It attracts and activates leukocytes, including T cells, eosinophils, and basophils, to sites of infection or injury . This chemokine is involved in various physiological and pathological processes, including immune surveillance, wound healing, and the development of inflammatory diseases .
RANTES exerts its effects by binding to specific receptors on the surface of target cells. The primary receptors for RANTES are CCR1, CCR3, and CCR5, which are G protein-coupled receptors . Upon binding to these receptors, RANTES activates intracellular signaling pathways that lead to the migration and activation of immune cells .
The expression and activity of RANTES are tightly regulated at multiple levels. Transcriptional regulation is influenced by various cytokines and transcription factors that respond to inflammatory signals . Post-translational modifications, such as glycosylation, also play a role in modulating the stability and activity of RANTES .