The Sf9 expression system leverages Baculovirus vectors to produce glycosylated proteins, ensuring proper post-translational modifications. Key steps include:
Expression Host: Spodoptera frugiperda (Sf9) cells
Purification Method: Proprietary chromatographic techniques
The recombinant protein is formulated for optimal stability:
While Sf9-derived Exodus-2 is glycosylated, E. coli-produced versions lack this modification:
Feature | Sf9-Produced | E. coli-Produced |
---|---|---|
Molecular Mass | 13.1 kDa | 12 kDa |
Glycosylation | Yes | No |
Amino Acid Count | 119 | 110–111 |
Expression System | Baculovirus | Prokaryotic |
Ccl21a, C-C motif chemokine 21a, 6Ckine, Beta-chemokine exodus-2, Small-inducible cytokine A21a,Thymus-derived chemotactic agent 4, TCA4, Ccl21a, Scya21, Scya21a, 6CKBAC2, 6Ckine, ALP,AW987545, CKb9, plt, Scya21b, SLC.
Sf9, Baculovirus cells.
ADPSDGGGQD CCLKYSQKKI PYSIVRGYRK QEPSLGCPIP AILFSPRKHS KPELCANPEE GWVQNLMRRL DQPPAPGKQS PGCRKNRGTS KSGKKGKGSK GCKRTEQTQP SRGHHHHHH
Exodus-2 Mouse (CCL21) produced in Sf9 cells is a CC chemokine subgroup cytokine with distinctive structural characteristics. Unlike typical chemokines that possess 4 conserved cysteine residues, Exodus-2 contains 6 conserved cysteine residues, earning its alternative designations as 6Ckine and Secondary Lymphoid-tissue Chemokine (SLC). The recombinant protein is a single glycosylated polypeptide chain containing 119 amino acids (positions 24-133) with a molecular mass of approximately 13.1kDa. It features a 9-amino acid His tag at the C-terminus to facilitate purification and experimental applications .
Exodus-2 Mouse primarily binds to the CCR7 receptor located on cell surfaces. This receptor-ligand interaction is crucial for various immunological processes, particularly in lymphocyte trafficking and homing to secondary lymphoid tissues. Understanding this interaction provides valuable insights into immune cell migration, lymphoid tissue development, and inflammatory responses in experimental models .
For optimal stability, Exodus-2 Mouse, Sf9 preparation (0.5mg/ml) in Phosphate-Buffered Saline (pH 7.4) with 10% glycerol should be stored according to intended usage duration. For short-term use (2-4 weeks), storage at 4°C is sufficient. For extended periods, frozen storage at -20°C is recommended, preferably with a carrier protein addition (0.1% HSA or BSA) to enhance stability. Multiple freeze-thaw cycles should be strictly avoided as they significantly compromise protein integrity and biological activity .
Methodology for Exodus-2 Mouse in cell migration assays typically involves:
Preparation of migration chambers with appropriate pore size (5-8μm)
Addition of varying concentrations of Exodus-2 (10-100ng/ml) to the lower chamber
Introduction of CCR7-expressing cells (e.g., dendritic cells, T lymphocytes) to the upper chamber
Incubation period (2-4 hours at 37°C, 5% CO₂)
Quantification of migrated cells using flow cytometry or microscopy techniques
This approach allows researchers to assess chemotactic potency and specificity, with proper controls including random migration (no chemokine) and positive controls with established chemoattractants.
For investigating Exodus-2 signaling pathways in primary mouse cells, researchers should:
Isolate primary cells of interest (dendritic cells, T cells) using magnetic or fluorescence-based sorting
Culture cells in serum-reduced media (0.5-1% serum) for 4-6 hours prior to stimulation
Stimulate with purified Exodus-2 Mouse protein (10-50ng/ml)
Harvest cells at specific time points (2, 5, 10, 15, 30 minutes)
Analyze key signaling components through:
Western blotting for phosphorylated ERK1/2, Akt, p38 MAPK
Calcium flux assays using fluorescent indicators
Actin polymerization assays for cytoskeletal rearrangement
This methodological approach enables detailed understanding of temporal signaling dynamics and pathway cross-talk.
For in vivo lymphocyte trafficking studies using Exodus-2 Mouse:
Label target lymphocytes with fluorescent dyes (CFSE, CMTMR) or luciferase reporters
Pre-treat cells with varying concentrations of Exodus-2 (1-10μg/ml) for 30-60 minutes
Inject treated cells intravenously into recipient mice
Monitor cell distribution using:
Flow cytometry of harvested lymphoid organs at set time points
Intravital microscopy for real-time visualization
Whole-body imaging for luciferase-expressing cells
The approach should include appropriate controls, including untreated cells and cells treated with CCR7 antagonists to confirm specificity of observed effects.
Comparative functional analysis reveals significant differences between Sf9-produced and E. coli-expressed Exodus-2 Mouse:
Property | Sf9-Expressed Exodus-2 | E. coli-Expressed Exodus-2 |
---|---|---|
Glycosylation | Present (native-like) | Absent |
Folding | Native conformation | Requires refolding |
Biological Activity | High (ED₅₀: 5-10ng/ml) | Variable (ED₅₀: 20-100ng/ml) |
Stability | Higher thermal stability | Lower thermal stability |
Receptor Binding | High affinity | Reduced affinity |
The Sf9-expressed variant typically exhibits superior characteristics for assays requiring physiological receptor interactions, while E. coli-expressed protein may be suitable for less sensitive applications or structural studies requiring higher protein yields.
When transitioning Exodus-2 experiments from in vitro to in vivo models, researchers should address:
Dosage adjustment: In vitro effective concentrations (10-100ng/ml) must be recalculated for systemic delivery (typically 1-10μg per mouse)
Delivery method optimization:
Intravenous administration for systemic effects
Subcutaneous for localized lymph node targeting
Intranasal for respiratory immunity studies
Pharmacokinetic considerations:
Calculate protein half-life in circulation
Determine tissue distribution patterns
Assess potential degradation by serum proteases
Model-specific variables:
Account for strain-specific responses
Consider age-dependent CCR7 expression levels
Evaluate potential interference from endogenous CCL21
These adaptations ensure meaningful translation between controlled in vitro findings and complex in vivo environments.
Recent findings suggest chemokines like Exodus-2 may influence neuronal development. For investigating potential roles in synaptogenesis:
Preparation of primary mouse hippocampal or cortical neurons:
Culture neurons from wild-type and CCR7-deficient mice
Treat with Exodus-2 (5-25ng/ml) at critical developmental stages (DIV7-14)
Analysis methods:
Immunofluorescence for synaptic markers (synapsin, PSD-95)
Electrophysiological recordings (mEPSC frequency and amplitude)
Dendritic spine morphology assessment via Golgi staining
Live-cell imaging for synapse formation dynamics
Complementary in vivo approaches:
Stereotaxic injection of Exodus-2 into developing brain regions
Analysis of synaptic density in CCL21/CCR7 knockout models
Behavioral testing for functional outcomes
This experimental framework enables investigation of potential non-immune functions of Exodus-2 in neural development, similar to other chemokines with demonstrated neuronal effects .
To investigate signaling cross-talk between Exodus-2/CCR7 and other cytokine pathways:
Sequential stimulation protocols:
Pre-treatment with Exodus-2 (10-50ng/ml) followed by other cytokines
Co-stimulation with optimized combinations
Time-course analysis of receptor internalization and recycling
Signaling pathway dissection:
Selective inhibitor panels for major pathways (JAK/STAT, MAPK, PI3K)
Phosphoproteomic analysis of signaling nodes
CRISPR/Cas9 knockout of pathway components
Analysis techniques:
Multiplexed phospho-flow cytometry
Co-immunoprecipitation of receptor complexes
Advanced microscopy for receptor co-localization
Transcriptomic analysis of downstream gene regulation
This systematic approach reveals pathway integration at both receptor and post-receptor levels, identifying synergistic or antagonistic relationships between Exodus-2 and other immunomodulatory signals.
To study Exodus-2 functions during infection using mouse-adapted viral models:
Experimental design considerations:
Selection of appropriate mouse-adapted viral strains (e.g., mouse-adapted SARS-CoV-2)
Temporal analysis of Exodus-2 expression during infection phases
Comparison between wild-type and CCL21/CCR7-deficient mice
Analytical approaches:
Flow cytometric analysis of immune cell recruitment and CCR7 expression
Histopathological assessment of lymphoid architecture
Viral burden quantification in tissues
Cytokine/chemokine profiling using multiplexed assays
Mechanistic investigations:
Adoptive transfer of CCR7-sufficient/deficient lymphocytes
Exodus-2 neutralization or supplementation strategies
Lymph node homing and germinal center formation analysis
This approach utilizes mouse-adapted viral models, such as those developed for SARS-CoV-2 research, to elucidate Exodus-2's contributions to antiviral immune responses and lymphoid tissue organization during infection .
Researchers frequently encounter these challenges when working with Exodus-2:
Loss of activity during storage:
Aliquot protein into single-use volumes before freezing
Add carrier proteins (0.1% HSA/BSA) to prevent surface adsorption
Validate activity after storage with controlled migration assays
Inconsistent chemotactic responses:
Standardize cell preparation protocols (passage number, activation state)
Include positive controls (e.g., CXCL12) in each experiment
Optimize chemokine gradient by testing multiple concentrations
Ensure CCR7 expression on target cells via flow cytometry
Non-specific binding issues:
Pre-block surfaces with appropriate blocking reagents
Use low-binding microtubes for protein handling
Include isotype controls for antibody-based detection systems
These methodological refinements significantly improve reproducibility and reliability of Exodus-2 functional assays across different experimental systems.
To distinguish Exodus-2 specific effects from other chemokine responses:
Implement genetic approaches:
Use CCR7-knockout models as negative controls
Employ CCL21-deficient systems (e.g., plt/plt mice)
Create receptor-selective mutants through structure-guided design
Pharmacological strategies:
Apply CCR7-specific antagonists in parallel experiments
Utilize neutralizing antibodies against Exodus-2
Implement receptor desensitization protocols
Analytical methods:
Perform competitive binding assays with labeled ligands
Conduct dose-response relationships across multiple chemokines
Implement multiparameter analysis to identify unique signaling signatures
These approaches provide the necessary controls to attribute observed biological effects specifically to Exodus-2/CCR7 interactions rather than to broader chemokine responses or experimental artifacts.
While traditionally studied in immune contexts, emerging research suggests potential roles for Exodus-2 in neuroinflammatory processes. To investigate this:
Experimental design framework:
Implementation of neurological disease models (EAE, stroke, neurodegeneration)
Temporal analysis of Exodus-2 and CCR7 expression in neural tissues
Correlation with blood-brain barrier integrity and immune cell infiltration
Intervention approaches:
CCR7 antagonist administration during disease progression
Neutralizing antibodies against Exodus-2
CCL21/CCR7 conditional knockout in specific neural or immune cell populations
Assessment metrics:
Quantification of neuroinflammatory markers (cytokines, microglial activation)
Behavioral testing for functional outcomes
Advanced imaging for immune cell trafficking into CNS compartments
This research direction bridges immunology and neuroscience, potentially revealing novel therapeutic targets for neuroinflammatory conditions based on Exodus-2 modulation.
Innovative research tools based on Exodus-2 properties present several opportunities:
Designer recombinant proteins:
Bifunctional molecules combining Exodus-2 with other bioactive domains
Fluorescently labeled variants for tracking receptor binding and internalization
pH-sensitive Exodus-2 constructs for endosomal trafficking studies
Advanced detection systems:
Nanobody-based sensors for real-time Exodus-2 visualization
FRET-based reporters for CCR7 activation dynamics
Aptamer-based detection platforms for Exodus-2 quantification
Research applications:
Cell-specific targeting using Exodus-2 functionalized nanoparticles
Lymph node-targeted delivery systems for immunomodulators
Engineered cell systems with tunable CCR7 responses
These innovative tools expand the research capabilities beyond conventional approaches, enabling more sophisticated mechanistic studies of chemokine biology and potential therapeutic applications.
Exodus-2, also known as Chemokine (C-C motif) ligand 21 (CCL21), is a small cytokine belonging to the CC chemokine family. This chemokine is also referred to as 6Ckine due to its six conserved cysteine residues, which is a unique feature compared to the typical four cysteines found in other chemokines. Exodus-2 plays a crucial role in the immune system by acting as a chemoattractant for lymphocytes, guiding them to secondary lymphoid tissues. The gene encoding CCL21 is located on human chromosome 9.
Exodus-2 (CCL21) can be produced recombinantly in various host systems, including Escherichia coli and Sf9 Baculovirus cells. The recombinant production in Sf9 Baculovirus cells involves the following steps:
Exodus-2 (CCL21) binds to the chemokine receptor CCR7, which is located on the surface of various immune cells. This binding triggers a cascade of intracellular signaling events that lead to the directed migration of lymphocytes to secondary lymphoid tissues. The interaction between CCL21 and CCR7 is essential for the proper functioning of the immune system, particularly in the context of immune surveillance and the initiation of immune responses.
The recombinant Exodus-2 (CCL21) produced in Sf9 Baculovirus cells is characterized by its high purity and stability. It is typically formulated in a phosphate-buffered saline solution with 10% glycerol to maintain its stability during storage. The protein is stored at 4°C for short-term use and at -20°C for long-term storage to prevent degradation .