MIG Mouse

MIG Mouse Recombinant (CXCL9)
Shipped with Ice Packs
In Stock

Description

Introduction to Mouse MIG (CXCL9)

Mouse MIG is a 12.2 kDa protein containing 105 amino acid residues, including four conserved cysteine residues characteristic of CXC chemokines . It is encoded by the Cxcl9 gene (Entrez Gene ID: 17329) and serves as a T-cell chemoattractant inducible by interferon-γ . Unlike most CXC chemokines, MIG selectively attracts lymphocytes while remaining inactive toward neutrophils .

Key Aliases:

  • Protein: CXCL9, monokine induced by interferon-γ

  • Gene: Cxcl9, Mig, Scyb9

  • UniProt ID: P18340 .

Role in Immune Response and Disease Pathogenesis

MIG plays a pivotal role in orchestrating Th1-mediated immunity, as demonstrated in studies of viral CNS infections (e.g., mouse hepatitis virus, MHV) .

Key Findings from MHV Infection Models

  • MIG Expression Kinetics:

    • Detectable at days 7 and 12 post-infection (p.i.), absent at early (day 2) or late (day 35) stages .

  • Impact of MIG Neutralization:

    • Mortality: >90% mortality in anti-MIG-treated mice vs. ~50% in controls by day 12 p.i. .

    • Viral Burden: 5.4 ± 0.4 log units in anti-MIG-treated mice vs. 2.0 ± 0.2 log units in controls .

    • T-Cell Infiltration:

      • CD4+ T-cells: 45% decrease at day 7 p.i. (anti-MIG vs. control) .

      • CD8+ T-cells: 60% decrease at day 7 p.i. .

    • Cytokine Shifts:

      • Reduced IFN-γ and IFN-β levels.

      • Increased IL-10 (anti-inflammatory Th2 cytokine) .

These results underscore MIG’s necessity in sustaining protective Th1 responses during viral infections .

Detection and Quantification Methods

Two primary methods are used to measure mouse MIG: Cytometric Bead Arrays (CBA) and ELISA.

Comparison of Detection Methods

ParameterBD CBA Mouse MIG Flex Set R&D Systems Quantikine ELISA
Assay TypeBead-based immunoassaySolid-phase ELISA
Sensitivity10 pg/mL (assay range: 10–2,500 pg/mL) 5–10 pg/mL (assay range: 5–1,500 pg/mL)
Sample TypesSerum, cell culture supernatants Serum, cell culture supernatants, tissue homogenates
Multiplex CapabilityCompatible with BD CBA Master Buffer Kits Single-target detection

Precision Data (ELISA) :

Sample TypeMean (pg/mL)SDCV%
Cell Culture (Intra)77, 216, 8156.2, 14.8, 688.1, 6.9, 8.3
Serum (Inter)66, 190, 7415.5, 14.1, 39.58.3, 7.4, 5.3

Research Findings and Applications

  • Therapeutic Targeting: Neutralizing MIG may impair anti-viral immunity but could modulate inflammatory diseases .

  • Cancer and Autoimmunity: While not directly studied in provided sources, MIG’s role in T-cell recruitment positions it as a candidate for immunotherapy .

  • Diagnostic Use: Both CBA and ELISA kits enable precise quantification of MIG in preclinical studies .

Product Specs

Introduction
Chemokine (C-X-C motif) ligand 9 (CXCL9), also known as Monokine induced by MIG, is a small cytokine in the CXC chemokine family. This chemokine attracts T-cells and shares close relation to two other CXC chemokines, CXCL10 and CXCL11. The genes for all three chemokines are located near each other on human chromosome 4. CXCL9, CXCL10, and CXCL11 all activate chemotactic functions through interactions with the chemokine receptor CXCR3.
Description
Recombinant Mouse MIG (CXCK9) is produced in E. coli. It is a single, non-glycosylated polypeptide chain consisting of 105 amino acids with a molecular weight of 12.2 kDa. The protein is purified using proprietary chromatographic techniques.
Physical Appearance
White, sterile, lyophilized powder.
Formulation
The protein was filtered through a 0.2µm filter then lyophilized from a 0.5 mg/mL solution in 20 mM phosphate buffer (pH 7.4) and 100 mM NaCl.
Solubility
To reconstitute the lyophilized MIG, it is recommended to dissolve it in sterile 18 MΩ-cm H2O to a concentration of at least 100 µg/mL. This solution can be further diluted in other aqueous solutions.
Stability
Lyophilized MIG is stable at room temperature for up to 3 weeks. However, it is recommended to store the lyophilized protein desiccated below -18°C for long-term storage. After reconstitution, CXCL9 should be stored at 4°C for 2-7 days. For long-term storage, add a carrier protein (0.1% HSA or BSA) and store below -18°C. Avoid repeated freeze-thaw cycles.
Purity
Purity is determined by the following methods: (a) RP-HPLC analysis (b) SDS-PAGE analysis. Purity is greater than 97.0%.
Biological Activity
Biological activity is determined by the chemoattraction of human lymphocytes at a concentration range of 0.1-1 ng/mL.
Synonyms

Small inducible cytokine B9, CXCL9, MIG, chemokine (C-X-C motif) ligand 9, CMK, Humig, SCYB9, crg-10, M119.

Source
Escherichia Coli.
Amino Acid Sequence
The sequence of the first five N-terminal amino acids was determined and was found to be, Thr-Leu-Val-Ile-Arg.

Q&A

What is MIG (CXCL9) in mouse models and what biological functions does it serve?

MIG (Monokine Induced by Gamma interferon), also designated as CXCL9, is a chemokine protein originally identified through its induction by IFN-γ in macrophages and other cells. In mouse models, MIG functions primarily as a T-cell chemoattractant involved in inflammatory responses and immune cell recruitment to sites of infection or inflammation. The protein plays critical roles in multiple immunological processes including antimicrobial defense, tumor suppression, and regulation of leukocyte trafficking .

Unlike other chemokines, MIG expression is tightly regulated by IFN-γ signaling pathways, making it a valuable biomarker for Th1-type immune responses in mouse models. The protein exerts its biological effects through binding to the CXCR3 receptor, which it shares with other related chemokines. When designing experiments to study MIG function, researchers should consider its involvement in multiple disease models including viral infections, autoimmune diseases, transplant rejection, and cancer immunosurveillance.

What are the standard detection methods for quantifying MIG in mouse samples?

Two primary methodologies dominate MIG detection in mouse samples: ELISA-based assays and bead-based immunoassays such as Cytometric Bead Array (CBA).

The ELISA approach, exemplified by the Quantikine Mouse MIG Immunoassay, utilizes E. coli-expressed recombinant mouse MIG and specific antibodies in a solid-phase ELISA format with a typical assay duration of approximately 4.5 hours. This method is validated for detecting MIG in multiple sample types including cell culture supernatants, tissue homogenates, and serum . The assay has been confirmed to accurately quantitate both recombinant and naturally occurring mouse MIG, with natural mouse MIG samples producing linear curves parallel to standard curves .

Alternatively, the BD™ CBA Mouse MIG Flex Set employs a bead-based immunoassay approach capable of multiplexing with other analytes. This system uses flow cytometry for detection and measurement, offering a detection range of 10-2,500 pg/mL . The CBA method requires additional components such as the BD CBA Mouse/Rat Soluble Protein Master Buffer Kit and appropriate analysis software such as FCAP Array™ Software .

Both methodologies demonstrate high specificity, with commercial assays showing minimal cross-reactivity with other mouse cytokines or chemokines .

How should mouse samples be prepared for optimal MIG detection?

Proper sample preparation is crucial for accurate MIG detection. For serum collection, allow blood to clot for 30 minutes at room temperature before centrifugation at approximately 1,000-2,000 × g for 10 minutes. The resulting serum should be immediately transferred to clean polypropylene tubes and stored appropriately if not analyzed immediately.

For cell culture supernatants, carefully remove particulates by centrifugation and analyze immediately or aliquot and store at ≤ -20°C to prevent protein degradation. Avoid repeated freeze-thaw cycles which can compromise MIG stability and immunoreactivity.

When preparing tissue homogenates, tissues should be rapidly harvested and processed in appropriate buffer systems containing protease inhibitors. Homogenization should be performed at cold temperatures to minimize proteolytic degradation, followed by centrifugation to remove cellular debris before analysis.

All samples benefit from standardization of collection and storage protocols to ensure consistency between experiments. Critical pre-analytical variables that can affect MIG detection include sample collection timing, anticoagulant choice (for plasma), processing delays, and storage conditions .

How can researchers address potential interference factors when measuring MIG in complex biological samples?

When measuring MIG in complex biological samples, multiple interference factors can compromise assay accuracy. Implement these methodological approaches to minimize interference:

  • Heterophilic antibody interference: Pre-absorb samples with irrelevant antibodies of the same species as the capture/detection antibodies used in the assay. For the BD CBA Mouse MIG Flex Set, samples containing high levels of immunoglobulins should be diluted appropriately before testing .

  • Matrix effects: Prepare standards in the same matrix as test samples when possible. For tissue homogenates or complex biological fluids, consider matrix-matched calibration or parallel dilution analysis to identify and correct for non-specific matrix effects.

  • Cross-reactivity: While commercial assays report minimal cross-reactivity with other cytokines, independent validation is recommended, especially when analyzing samples from experimental models with complex inflammatory profiles. The RayBiotech Mouse MIG ELISA specifically reports no cross-reactivity with multiple tested mouse cytokines including CD30, L CD30, CD40, CRG-2, CTACK, CXCL16, and Eotaxin .

  • Proteolytic degradation: Add protease inhibitors during sample collection and processing. Consider analyzing samples immediately after collection or properly aliquot and store at temperatures that minimize proteolytic activity.

  • Interfering substances: For serum samples with high lipid content, consider additional centrifugation steps. For hemolyzed samples, correction algorithms may be necessary or samples may need to be excluded.

The use of appropriate negative and positive controls, including spike-recovery experiments, can help identify and quantify potential interference effects in specific sample types.

What are the critical parameters for designing MIG induction experiments in mouse models?

When designing experiments to study MIG induction in mouse models, several critical parameters require careful consideration:

  • Stimulus selection and dosing: IFN-γ is the classical inducer of MIG expression, but optimal concentrations and timing vary by cell type and experimental context. Titration experiments should establish dose-response relationships in your specific system.

  • Strain-dependent variations: Different mouse strains may exhibit varying baseline levels and induction kinetics of MIG. C57BL/6 is commonly used, but strain-specific differences should be accounted for in experimental design and interpretation.

  • Cell type considerations: While macrophages are classical MIG producers, other cell types including endothelial cells, fibroblasts, and some epithelial cells also express MIG upon stimulation. Cell-type specific responses should be characterized in complex in vivo models.

  • Temporal dynamics: MIG expression typically peaks 12-24 hours after IFN-γ stimulation, but kinetics vary across tissues and experimental conditions. Time-course studies are essential for capturing maximal induction and resolution phases.

  • Co-stimulation effects: While IFN-γ is the primary inducer, other cytokines (particularly TNF-α) can synergistically enhance MIG production. Experimental design should account for these potential interactions in inflammatory settings.

  • In vivo challenges: For pathogen challenge models, carefully select infectious dose, route, and timing of sample collection. For inflammatory models, standardize the stimulus, route, and assessment timepoints.

Appropriate positive controls (e.g., established IFN-γ inducible genes like IRF1) and negative controls (e.g., stimulation in the presence of JAK/STAT pathway inhibitors) should be incorporated to validate experimental systems .

How should researchers interpret variations in MIG levels across different mouse tissues and experimental conditions?

Interpreting variations in MIG levels requires understanding the biological and technical factors influencing measurements:

When comparing MIG levels between experimental groups, consider normalizing to appropriate housekeeping genes (for mRNA) or total protein content (for protein measurements) to account for potential differences in sample cellularity or quality.

What methodological differences exist between ELISA and Cytometric Bead Array for mouse MIG quantification?

The two dominant technologies for mouse MIG quantification—ELISA and Cytometric Bead Array (CBA)—differ in several important methodological aspects:

ParameterELISA (e.g., Quantikine)Cytometric Bead Array (e.g., BD CBA)
PrincipleSolid-phase sandwich ELISA using plate-bound antibodiesBead-based immunoassay with flow cytometric detection
MultiplexingSingle analyte per wellCan be multiplexed with other BD CBA Flex Sets
Detection RangeTypically 10-1000 pg/mL10-2,500 pg/mL (for BD CBA Mouse MIG Flex Set)
Assay TimeApproximately 4.5 hoursVariable depending on multiplexing
EquipmentPlate reader (absorbance)Flow cytometer and analysis software
Sample VolumeTypically 50-100 μLConfigurable based on multiplexing needs
AdvantagesEstablished methodology, minimal equipment requirementsMultiplexing capability, sample conservation

The ELISA methodology employs recombinant mouse MIG and specific antibodies in a traditional sandwich ELISA format, while the CBA approach uses fluorescent beads with distinct positions (e.g., position D9 for the BD CBA Mouse MIG Flex Set) to allow multiplexed analysis .

For accurate comparisons between studies, researchers should maintain consistency in methodology, as absolute values may differ between platforms. When changing assay platforms, appropriate validation and correlation studies should be performed.

What are the recommended approaches for validating experimental results when studying MIG in mouse models?

Rigorous validation of experimental results is essential for MIG research. Implement these approaches:

  • Technical validation strategies:

    • Run samples in duplicate or triplicate to assess technical reproducibility

    • Include standard curves with each assay run to account for inter-assay variation

    • Use both positive controls (e.g., IFN-γ stimulated samples) and negative controls

    • Perform spike-recovery experiments to validate assay performance in your specific sample matrices

    • Consider orthogonal detection methods (e.g., validating ELISA results with Western blot or PCR)

  • Biological validation approaches:

    • Use genetic models (e.g., IFN-γ receptor knockout mice) to confirm specificity of induction

    • Employ neutralizing antibodies against IFN-γ to demonstrate cytokine-specific regulation

    • Compare results across different mouse strains to ensure robustness of findings

    • Correlate protein measurements with mRNA expression data

  • Statistical considerations:

    • Determine appropriate sample sizes through power analysis

    • Apply appropriate statistical tests based on data distribution

    • Use correction for multiple comparisons when appropriate

    • Report both statistical significance and effect sizes

  • Replication strategies:

    • Repeat key experiments independently to confirm reproducibility

    • Consider biological replicates across different experimental cohorts

    • Validate critical findings using different experimental approaches

Maintaining detailed records of assay conditions, reagent lots, and sample handling procedures is essential for troubleshooting unexpected results and ensuring reproducibility .

How can researchers optimize the standard curve preparation for accurate MIG quantification?

Proper standard curve preparation is critical for accurate MIG quantification:

  • Standard reconstitution: For lyophilized standards, such as those in the BD CBA Mouse MIG Flex Set, follow precise reconstitution protocols. The standard should be reconstituted in the specified volume (e.g., 4.0 mL of Assay Diluent for the BD CBA kit) to achieve the top standard concentration (2,500 pg/mL) .

  • Serial dilution technique: Prepare a complete standard curve using serial dilutions from the top standard. For the BD CBA Mouse MIG Flex Set, this typically covers 10-2,500 pg/mL . Use appropriate pipetting techniques:

    • Use fresh pipette tips for each dilution step

    • Mix thoroughly but gently between dilutions (avoid introducing bubbles)

    • Maintain consistent timing between dilution preparation and assay initiation

  • Diluent considerations: Prepare standards in the same diluent used for samples whenever possible. For serum or plasma samples, consider preparing standards in a matrix-matched diluent to account for matrix effects.

  • Storage and stability: Use freshly prepared standards for each assay run. As specified for the BD CBA kit, discard unused reconstituted standard rather than storing for future use .

  • Curve fitting approaches: Apply appropriate curve-fitting algorithms based on the expected response characteristics. Four-parameter logistic regression typically provides optimal fit for most immunoassays.

  • Validation parameters:

    • Calculate the coefficient of determination (R²) to assess curve fit quality (target >0.99)

    • Evaluate back-calculated concentrations of standards (acceptance criteria typically ±15-20% of nominal)

    • Determine the lower limit of quantification based on precision profiles

For the BD CBA Mouse MIG Flex Set, the standard curve can be analyzed using FCAP Array Software, which provides appropriate curve-fitting algorithms specifically designed for bead-based immunoassays .

What are common sources of variability in mouse MIG measurements and how can they be addressed?

Several factors contribute to variability in mouse MIG measurements:

  • Pre-analytical variables:

    • Sample collection timing: Standardize collection relative to experimental interventions

    • Processing delays: Minimize time between collection and processing

    • Storage conditions: Maintain consistent freezing/thawing procedures

    • Hemolysis: Avoid hemolyzed samples or account for interference

    Solution: Develop and strictly adhere to standard operating procedures for sample handling.

  • Analytical variables:

    • Reagent variability: Track lot numbers and perform lot-to-lot validation

    • Temperature fluctuations: Maintain consistent ambient conditions during assay

    • Pipetting errors: Use calibrated pipettes and consistent technique

    • Incubation timing: Use timers to ensure precise incubation periods

    • Washing efficiency: Standardize washing procedures

    Solution: Include internal quality control samples in each assay run to monitor analytical performance.

  • Biological variables:

    • Circadian fluctuations: Collect samples at consistent times of day

    • Acute stress responses: Minimize handling stress before sample collection

    • Recent infections: Monitor health status of experimental animals

    • Age and sex differences: Use age and sex-matched controls

    Solution: Design experiments with appropriate stratification and control groups.

  • Post-analytical variables:

    • Data normalization approaches: Use consistent normalization strategies

    • Outlier handling: Apply predefined criteria for outlier identification

    • Statistical methods: Select appropriate statistical tests

    Solution: Predefine data analysis workflows before study initiation.

Implementing a comprehensive quality control program, including monitoring assay performance metrics over time, can help identify and mitigate sources of variability .

How should researchers approach troubleshooting when MIG detection assays produce unexpected results?

When encountering unexpected results in MIG detection assays, employ this systematic troubleshooting framework:

  • Verify reagent integrity:

    • Check expiration dates and storage conditions of all reagents

    • For the BD CBA Mouse MIG Flex Set, verify that PE Detection Reagent and Capture Beads have been protected from light exposure

    • Consider preparing fresh working reagents if questionable

  • Review assay execution:

    • Confirm correct reconstitution of standards (e.g., in 4.0 mL Assay Diluent for BD CBA kit)

    • Verify incubation times and temperatures adhered to protocol

    • Check washing procedures for consistency

    • For CBA assays, verify flow cytometer settings and calibration

  • Examine sample quality:

    • Assess samples for signs of degradation or contamination

    • Consider running spike-recovery experiments to test for inhibitory factors

    • Analyze samples at multiple dilutions to check for linearity

  • Address high background issues:

    • Increase washing steps or washing stringency

    • Check for cross-contamination between wells

    • Evaluate reagent cross-reactivity potential

    • For CBA assays, verify proper gating strategies

  • Investigate low or no signal problems:

    • Confirm sample handling didn't inactivate the target protein

    • Test positive control samples (e.g., IFN-γ stimulated macrophage supernatants)

    • Verify detection antibody functionality

    • For CBA assays, ensure proper identification of bead populations (e.g., D9 position for BD CBA Mouse MIG)

  • Address poor standard curve:

    • Ensure proper reconstitution and dilution of standards

    • Verify accuracy of standard concentrations

    • Check pipetting technique and equipment calibration

When transitioning between different assay platforms or when establishing new protocols, consider running parallel analyses to validate results across methodologies .

What strategies can minimize inter-assay variability when measuring MIG across multiple experiments?

To minimize inter-assay variability in longitudinal MIG studies:

  • Standardize reagents and materials:

    • Use reagents from the same lot when possible, especially critical components like capture and detection antibodies

    • Purchase larger kit sizes (e.g., 5-plate kits) rather than multiple small kits

    • Create and freeze aliquots of critical reagents to maintain consistency

    • For the BD CBA Mouse MIG Flex Set, maintain consistent sourcing of the corresponding Master Buffer Kit (Cat. No. 558266 or 558267)

  • Implement quality control measures:

    • Include internal quality control samples of known concentration in every assay run

    • Track assay performance metrics (sensitivity, precision, accuracy) over time

    • Establish acceptance criteria for standard curves (e.g., R² >0.99)

    • Create control charts to monitor assay drift

  • Optimize assay execution:

    • Use automated liquid handling systems when possible

    • Standardize incubation conditions using calibrated equipment

    • Train multiple operators using the same standard operating procedures

    • Perform assays at consistent times of day

  • Implement data normalization strategies:

    • Use reference standards across multiple plates/assays

    • Consider plate-specific normalization factors when analyzing multi-plate datasets

    • For flow cytometry-based assays like CBA, include fluorescence calibration beads

  • Plan experimental design to minimize batch effects:

    • Randomize samples across plates rather than grouping by experimental condition

    • Process critical comparisons within the same assay run when possible

    • When complete randomization isn't feasible, use balanced incomplete block designs

  • Data analysis approaches:

    • Consider statistical methods that account for batch effects (e.g., mixed effects models)

    • Apply appropriate normalization techniques during data processing

    • Maintain consistent analysis pipelines across experiments

By implementing these strategies, researchers can significantly reduce variability in longitudinal studies of mouse MIG, increasing statistical power and confidence in observed biological effects .

How can MIG measurements be integrated with other immunological parameters for comprehensive immune profiling in mouse models?

Integrating MIG measurements with other immunological parameters enables comprehensive immune profiling:

  • Multiplexed cytokine/chemokine analysis:

    • Utilize the multiplexing capability of the BD CBA system to simultaneously measure MIG alongside other cytokines and chemokines

    • Include related CXCR3 ligands (CXCL10/IP-10, CXCL11/I-TAC) to assess coordinated chemokine responses

    • Measure both Th1 (IFN-γ, IL-12) and Th2 (IL-4, IL-13) cytokines to contextualize MIG induction within polarized immune responses

  • Cellular immune phenotyping integration:

    • Correlate MIG levels with flow cytometric quantification of immune cell subsets

    • Assess CXCR3 expression on T cells, NK cells, and other relevant populations

    • Combine with functional assays (e.g., intracellular cytokine staining) to link MIG production with cellular activation states

  • Spatial analysis approaches:

    • Complement soluble MIG measurements with immunohistochemistry or in situ hybridization

    • Consider multiplexed immunofluorescence to co-localize MIG production with specific cell types

    • Integrate with spatial transcriptomics for comprehensive tissue-level analysis

  • Systems immunology frameworks:

    • Apply computational approaches (e.g., principal component analysis, clustering algorithms) to identify patterns across multiple immune parameters

    • Develop predictive models of immune responses incorporating MIG alongside other variables

    • Utilize the Mouse Genome Informatics (MGI) Resource for integrating genetic and genomic data

  • Temporal profiling strategies:

    • Design longitudinal sampling to capture dynamic relationships between MIG and other immune parameters

    • Apply time-series analysis methods to identify leading/lagging relationships

    • Consider repeated measures statistical approaches for analysis

This integrated approach provides mechanistic insights beyond what can be achieved through isolated MIG measurements, enabling researchers to place chemokine responses within broader immunological contexts .

What are the key considerations when using MIG as a biomarker in mouse models of human disease?

When employing MIG as a biomarker in mouse models of human disease, consider these critical factors:

  • Cross-species translation:

    • While mouse and human MIG share functional homology, expression patterns and regulation may differ

    • Validate mouse findings in human samples when possible

    • Consider species-specific differences in receptor binding affinity and downstream signaling

  • Model-specific validation:

    • Establish baseline MIG kinetics in each specific disease model

    • Determine optimal sampling timepoints based on disease progression

    • Validate MIG as a biomarker against established disease metrics

  • Genetic background effects:

    • Consider strain-dependent differences in MIG expression and regulation

    • For genetically modified models, use appropriate controls on matched genetic backgrounds

    • Utilize the MGI Resource to access information on strain-specific genetic factors that may influence MIG expression

  • Intervention assessment applications:

    • Evaluate MIG changes in response to therapeutic interventions

    • Determine whether MIG changes precede, coincide with, or follow clinical improvement

    • Assess whether MIG correlation with disease severity is consistent across intervention types

  • Analytical considerations:

    • Select assay platforms based on required sensitivity and dynamic range for the specific model

    • Consider whether serum, tissue, or cellular MIG measurements are most relevant

    • Validate assay performance in the specific matrix relevant to the disease model

  • Mechanistic versus correlative biomarker:

    • Distinguish between MIG as a mechanistic contributor to pathology versus a correlative biomarker

    • Use neutralizing antibodies or genetic approaches to establish causality

    • Consider measuring both MIG protein and receptor expression/activation

By addressing these considerations, researchers can effectively leverage MIG as a biomarker in translational research, enhancing the predictive value of mouse models for human disease .

How do genetic modifications and strain differences affect MIG expression and function in mouse models?

Genetic factors significantly impact MIG expression and function in mouse research:

  • Strain-dependent differences:

    • Baseline MIG expression varies across common laboratory strains

    • C57BL/6 mice typically show robust MIG induction following IFN-γ stimulation

    • BALB/c mice may exhibit different kinetics or magnitude of response

    • Outbred stocks show greater variability, potentially limiting statistical power

  • Impact of targeted genetic modifications:

    • IFN-γ pathway knockouts (IFN-γ⁻/⁻, IFN-γR⁻/⁻, STAT1⁻/⁻) severely impair MIG induction

    • JAK inhibition similarly reduces MIG expression

    • Alterations in transcription factors (e.g., IRF1, NF-κB components) may affect MIG regulation

    • Consider using the MGI Resource to identify relevant genetic factors and strain information

  • Receptor consideration:

    • CXCR3 knockout models allow assessment of MIG-dependent processes

    • Altered expression of alternative CXCR3 ligands may compensate for MIG deficiency

    • Post-translational modifications of CXCR3 can affect signaling responses

  • Spontaneous mutations and modifiers:

    • Background mutations in inbred strains may affect immune responses

    • Genetic drift in mouse colonies can introduce variability

    • Modifier genes may influence MIG expression in complex genetic backgrounds

  • Technical considerations for genetically modified models:

    • Verify phenotypes when backcrossing mutations to different backgrounds

    • Consider potential developmental compensation in germline knockouts

    • Use conditional and inducible systems to distinguish developmental from functional roles

  • Experimental design for genetic studies:

    • Use littermate controls whenever possible

    • Consider sex-specific effects on MIG expression

    • Account for age-related changes in immune responses

Understanding these genetic influences is essential for proper experimental design and interpretation, particularly in translational research where mouse findings must be extrapolated to human contexts .

What emerging technologies are advancing the study of MIG in mouse models?

Several cutting-edge technologies are transforming mouse MIG research:

  • Single-cell technologies:

    • Single-cell RNA sequencing enables identification of specific cell populations producing MIG

    • Single-cell proteomics allows correlation of MIG production with other cellular parameters

    • Mass cytometry (CyTOF) permits high-dimensional analysis of MIG-producing cells

    • These approaches provide unprecedented resolution of cellular heterogeneity in MIG responses

  • Advanced imaging approaches:

    • Multiplex immunofluorescence imaging allows visualization of MIG alongside multiple markers

    • Intravital microscopy enables real-time visualization of MIG-dependent cell migration

    • Tissue clearing techniques combined with light-sheet microscopy allow 3D visualization of MIG gradients

    • These methods provide spatial context for MIG production and function

  • CRISPR-based genetic manipulation:

    • CRISPR/Cas9 enables rapid generation of novel mouse models with modified MIG or CXCR3

    • CRISPR screening approaches can identify novel regulators of MIG expression

    • Base editing and prime editing allow precise modification of regulatory elements

    • These tools facilitate mechanistic studies of MIG regulation and function

  • In vitro organoid systems:

    • Mouse-derived organoids provide physiologically relevant systems for studying MIG

    • Co-culture systems with immune cells enable investigation of MIG-dependent interactions

    • Microfluidic approaches allow controlled manipulation of chemokine gradients

    • These systems bridge the gap between simplified in vitro and complex in vivo models

  • Computational and systems biology approaches:

    • Machine learning algorithms can identify patterns in complex MIG-related datasets

    • Network analysis tools enable integration of MIG within broader immune signaling networks

    • Multi-omics data integration provides comprehensive views of MIG regulation

    • These computational approaches extract maximal information from experimental data

Researchers can leverage the Mouse Genome Informatics (MGI) Resource to identify appropriate genetic models and tools for implementing these advanced approaches .

How can researchers optimize experimental design for studying MIG in complex disease models?

Optimizing experimental design for MIG studies in complex disease models requires careful planning:

  • Disease-specific considerations:

    • Characterize the natural kinetics of MIG expression throughout disease progression

    • Identify critical windows where MIG may play mechanistic roles

    • Consider potential confounding factors specific to the disease model (e.g., metabolic changes, tissue damage)

  • Sampling strategy optimization:

    • Implement longitudinal sampling when possible to capture dynamic changes

    • Consider multiple tissue compartments (circulation, affected tissues, draining lymph nodes)

    • Balance comprehensive sampling with potential effects of repeated manipulation

  • Control selection:

    • Include multiple control groups (vehicle, isotype antibody controls, genetic controls)

    • Consider sham procedures to account for procedure-related inflammation

    • Use time-matched controls for longitudinal studies

  • Intervention timing:

    • Design intervention studies based on established MIG kinetics

    • Consider both prophylactic and therapeutic intervention windows

    • Include washout periods when assessing reversibility of effects

  • Power and sample size considerations:

    • Conduct pilot studies to estimate variability in the specific model

    • Perform formal power calculations based on expected effect sizes

    • Plan for potential dropouts or exclusions in long-term studies

  • Integrated assessment approaches:

    • Combine functional readouts with MIG measurements

    • Correlate MIG levels with disease severity metrics

    • Include cellular and molecular analyses to establish mechanistic links

  • Translational considerations:

    • Include clinically relevant endpoints

    • Design sampling to parallel clinical biomarker collection

    • Consider how findings might inform human studies

By implementing these design principles, researchers can generate more robust and translatable data on MIG's role in complex disease processes .

What are the key unresolved questions in mouse MIG research that warrant further investigation?

Despite significant advances, several critical questions remain in mouse MIG research:

  • Tissue-specific regulation:

    • How do tissue microenvironments differentially regulate MIG expression?

    • What are the epigenetic mechanisms controlling tissue-specific MIG responses?

    • How do resident versus infiltrating cells contribute to local MIG production?

  • Post-translational modifications and variants:

    • What is the functional significance of MIG proteolytic processing?

    • How do different MIG isoforms affect receptor binding and signaling?

    • What enzymes regulate MIG activity in different tissue contexts?

  • Non-CXCR3 mediated functions:

    • Does MIG signal through alternative receptors in specific contexts?

    • What are the CXCR3-independent functions of MIG?

    • How do MIG-glycosaminoglycan interactions regulate function?

  • Temporal dynamics and cellular targeting:

    • How does the timing of MIG expression influence immune cell trafficking patterns?

    • What determines target cell responsiveness to MIG gradients?

    • How do cells integrate signals from MIG and other chemokines?

  • Therapeutic targeting considerations:

    • Under what conditions would MIG augmentation versus inhibition be therapeutically beneficial?

    • How can MIG be selectively modulated in specific tissues without systemic effects?

    • What are the potential consequences of long-term MIG modulation?

  • Integration with other biological systems:

    • How does MIG interact with the neuroendocrine system in stress responses?

    • What is the role of MIG in metabolic regulation and adipose tissue inflammation?

    • How does the microbiome influence MIG expression and function?

  • Comparative biology:

    • How conserved are MIG functions across mouse strains and other model organisms?

    • What aspects of human MIG biology are not accurately modeled in mice?

    • How can mouse models be optimized to better predict human MIG responses?

Addressing these questions will require interdisciplinary approaches and integration of emerging technologies. The Mouse Genome Informatics (MGI) Resource and other mouse-focused databases will continue to play vital roles in supporting these research directions .

Product Science Overview

Discovery and Gene Expression

CXCL9 was initially identified as a lymphokine-activated gene in mouse macrophages . The human version of CXCL9 was later cloned using mouse CXCL9 cDNA as a probe . The gene is induced in macrophages and primary glial cells in response to interferon-gamma (IFN-γ) stimulation .

Function and Biological Activity

CXCL9/MIG plays a crucial role in the immune response by acting as a chemoattractant for activated T cells . It is predominantly expressed by monocytes, macrophages, hepatocytes, and endothelial cells . The chemokine is involved in the recruitment of these immune cells to sites of inflammation or infection, thereby contributing to the body’s defense mechanisms .

Recombinant Protein Production

Recombinant Mouse CXCL9/MIG protein is typically produced in E. coli . The recombinant protein is purified to high levels of purity, often exceeding 95% as determined by SDS-PAGE and HPLC . It is biologically active and can be used in various research applications, including chemotaxis assays .

Applications in Research

The recombinant form of CXCL9/MIG is used extensively in immunological research to study its role in immune cell recruitment and activation . It is also utilized in assays to understand the mechanisms of inflammation and the immune response to infections and diseases .

Storage and Stability

Recombinant Mouse CXCL9/MIG protein is typically lyophilized and should be stored at -20°C to -70°C for long-term stability . Upon reconstitution, it is recommended to store the protein at 2-8°C for short-term use and at -20°C to -70°C for long-term storage . Repeated freeze-thaw cycles should be avoided to maintain protein integrity .

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.