MIP 1a Mouse

Macrophage Inflammatory Protein-1 Alpha Mouse Recombinant (CCL3)
Shipped with Ice Packs
In Stock

Description

Inflammatory and Immune Regulation

MIP-1α recruits neutrophils, monocytes, and T cells via CCR1, CCR4, and CCR5 receptors, contributing to acute inflammation . Key findings:

FunctionMechanism/OutcomeSource
Viral defenseRequired for inflammatory response to influenza and coxsackievirus; knockout mice show reduced resistance
HematopoiesisInhibits stem cell proliferation in vitro; not essential for normal hematopoiesis in vivo
Wound healingNo significant delay in re-epithelialization, angiogenesis, or collagen synthesis in MIP-1α−/− mice

Notably, MIP-1α−/− mice exhibit normal wound repair, suggesting compensatory mechanisms by other chemokines .

Hematopoietic and Antiviral Effects

MIP-1α/CCL3 exhibits dual roles:

  • Hematopoiesis: Suppresses stem cell proliferation but is dispensable for steady-state blood production .

  • Antiviral activity: Forms heterodimers with MIP-1β to inhibit HIV-1 replication .

Analytical Techniques

MethodDetection LimitApplicationsSource
CBA Flex Set2.3 pg/mL (theoretical)Serum, cell culture supernatants
Quantikine ELISA~2.7 pg/mLRelative quantification in biological fluids

Both methods use recombinant mouse MIP-1α standards and antibodies, validated for specificity and cross-reactivity .

Disease Models and Therapeutic Targets

MIP-1α/CCL3 is studied in:

  • Inflammatory diseases: Potential therapeutic target for autoimmune disorders (e.g., arthritis).

  • Viral infections: Inhibitors may reduce inflammation while preserving antiviral responses .

Experimental Tools

Recombinant MIP-1α is used to:

  • Induce chemotaxis: ED₅₀ of 0.4–2.0 ng/mL for CCR5-expressing BaF3 cells .

  • Study receptor signaling: CCR1/CCR5 binding assays and dimerization studies .

Product Specs

Introduction
Macrophage Inflammatory Proteins (MIPs), specifically MIP-1a and MIP-1b (now known as CCL3 and CCL4, respectively), are chemokines primarily produced by macrophages in response to bacterial endotoxins. These chemokines play a crucial role in inflammation by activating granulocytes, leading to acute neutrophilic inflammation. Additionally, they stimulate fibroblasts and macrophages to produce other pro-inflammatory cytokines like interleukin 1 (IL-1), IL-6, and TNF-a. Notably, the genes responsible for CCL3 and CCL4 are both situated on human chromosome 17.
Description
Recombinant Mouse Macrophage Inflammatory Protein-1 alpha, produced in E. coli, is a single, non-glycosylated polypeptide chain comprising 69 amino acids. With a molecular weight of 7820 Daltons, this protein is purified using proprietary chromatographic methods.
Physical Appearance
Sterile Filtered White lyophilized powder.
Formulation
The protein solution (1mg/ml) was lyophilized without the addition of any additives.
Solubility
To reconstitute the lyophilized Macrophage Inflammatory Protein-1a, it is recommended to dissolve it in sterile 18MΩ-cm H2O to a concentration of at least 100µg/ml. This solution can be further diluted in other aqueous solutions.
Stability
While lyophilized MIP-1a remains stable at room temperature for up to 3 weeks, it is best stored desiccated at temperatures below -18°C. After reconstitution, CCL3 should be stored at 4°C for 2-7 days. For long-term storage, freeze at -18°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
The purity is determined by two methods: RP-HPLC analysis and SDS-PAGE analysis, both indicating a purity greater than 99.0%.
Biological Activity
The biological activity is determined based on the chemoattractant potential of Balb3/C splenocytes. Using a concentration of 1-10ng/ml corresponds to a specific activity ranging from 100,000 to 1,000,000 IU/mg.
Synonyms

Small inducible cytokine A3, CCL3, Macrophage inflammatory protein 1-alpha, MIP-1-alpha, Tonsillar lymphocyte LD78 alpha protein, G0/G1 switch regulatory protein 19-1, G0S19-1 protein, SIS-beta, PAT 464.1, chemokine (C-C motif) ligand 3, MIP1A, SCYA3, G0S19-1, LD78ALPHA, TY-5, SIS-alpha, L2G25B.

Source
Escherichia Coli.
Amino Acid Sequence
The sequence of the first five N-terminal amino acids was determined and was found to be, Ala-Pro-Tyr-Gly-Ala.

Q&A

What is MIP-1α and what are its primary functions in mice?

MIP-1α (also known as CCL3) is a member of the CC or beta chemokine subfamily that was originally isolated from LPS-stimulated murine macrophage cell line cultures. It functions primarily as a chemoattractant to various cell types including monocytes, T cells, B cells, and eosinophils . The mature form of murine MIP-1α contains 69 amino acids, exists as dimers in solution, and tends to undergo reversible aggregation .

In mice, MIP-1α participates in host responses to bacterial, viral, parasitic, and fungal pathogens by regulating trafficking and activation of specific inflammatory cells. Unlike MIP-1β (which selectively attracts CD4+ lymphocytes), MIP-1α selectively attracts CD8+ lymphocytes, making this distinction important for experimental design . Additionally, MIP-1α exhibits inhibitory effects on hematopoietic stem cell proliferation, though research with knockout mice has demonstrated it is not essential for normal hematopoiesis .

What detection methods are available for measuring mouse MIP-1α in different sample types?

Mouse MIP-1α can be detected and quantified using several methodological approaches, with ELISA being the most widely utilized:

  • Sandwich ELISA: Commercially available kits use matched antibody pairs specific to mouse MIP-1α. These assays typically employ a target-specific capture antibody pre-coated onto microplate wells, to which samples bind. A detector antibody then completes the sandwich formation, and a substrate solution reacts with the enzyme-antibody-target complex to produce a measurable signal proportional to the MIP-1α concentration .

  • Sample compatibility: Validated ELISA methods can detect MIP-1α in various sample types including:

    • Cell culture supernatants

    • Serum

    • Plasma

    • Tissue homogenates

  • Performance characteristics: When selecting a detection method, consider the following metrics from validated assays:

ParameterTypical Performance
Intra-Assay Precision (CV%)2.7-4.4%
Inter-Assay Precision (CV%)6.3-6.7%
Recovery in Cell Culture Supernates105% (95-114% range)
Recovery in Serum97% (85-108% range)

These values represent benchmark performance standards for MIP-1α detection methodologies .

How do MIP-1α knockout mice differ from wild-type mice in immune response studies?

MIP-1α knockout (MIP-1α null) mice exhibit several distinctive characteristics that make them valuable models for studying chemokine functions:

  • Normal hematopoiesis: Despite MIP-1α's in vitro inhibitory effects on hematopoietic stem cells, knockout mice show no overt abnormalities in peripheral blood or bone marrow cell populations, indicating that MIP-1α is not essential for normal hematopoiesis under physiological conditions .

  • Viral response alterations: MIP-1α null mice demonstrate:

    • Reduced inflammatory responses to influenza virus infection

    • Resistance to coxsackievirus-induced myocarditis

  • Inflammatory response modulation: These knockout models have established that MIP-1α is specifically required for normal inflammatory responses to certain viral pathogens, providing a research tool for studying inflammation regulation .

When designing experiments with these knockout models, researchers should account for these baseline differences when interpreting results, particularly in infectious disease and inflammatory response studies.

What are the methodological considerations for optimizing MIP-1α detection in multiparameter mouse studies?

When incorporating MIP-1α measurements into complex experimental designs, several methodological considerations can enhance data quality and interpretation:

  • Sample collection and processing standardization:

    • For serum collection, standardize clotting time to minimize variability

    • For cell culture supernatants, standardize cell density and stimulation time

    • Process all samples identically to avoid introducing technical variation

  • Assay selection based on experimental design:

    • For studies requiring absolute quantification, use ELISA methods with recombinant standards

    • When examining naturally occurring MIP-1α, select assays validated with naturally derived MIP-1α

    • For multiplex analysis, verify lack of cross-reactivity with other chemokines, particularly MIP-1β

  • Calibration and quality control:

    • Include multiple concentrations spanning physiologically relevant ranges

    • The following data table provides reference ranges for quality control samples:

Sample TypeTypical Concentration Range
Unstimulated cell culture5-20 pg/mL
LPS-stimulated macrophages50-250 pg/mL
Normal mouse serum10-40 pg/mL
Inflammation models50-500 pg/mL
  • Data normalization approaches:

    • For tissue homogenates, normalize to total protein content

    • For cell culture supernatants, normalize to cell number or metabolic activity

How do different stimuli affect MIP-1α production in murine models, and what are the implications for experimental design?

The production of MIP-1α by murine cells is highly stimulus-dependent, which has significant implications for experimental design:

  • Stimulus-specific production profiles:

    • Endogenous stimuli: Interleukin-1β and Interferon-γ induce significant MIP-1α secretion from monocytes

    • Pathogen-associated molecular patterns: Lipopolysaccharide (LPS) and lipoteichoic acid from gram-positive bacteria are potent inducers

    • Viral infection: Influenza virus and coxsackievirus infections trigger distinct MIP-1α production patterns

  • Cell type-specific responses:

    • Macrophages produce high levels of MIP-1α upon stimulation

    • T lymphocytes require different activation signals

    • Dendritic cells show distinct production kinetics

  • Temporal considerations:

    • Acute stimulation typically produces peak MIP-1α levels within 4-24 hours

    • Chronic stimulation may lead to desensitization or sustained production

    • Sampling timepoints should be carefully selected based on the specific stimulus

  • Methodological recommendations:

    • Include multiple timepoints in initial experiments to establish optimal sampling windows

    • Use dose-response curves for stimuli to identify threshold and saturation levels

    • Include positive controls with well-characterized stimuli (e.g., LPS at 100 ng/mL)

    • Control for potential confounding factors such as serum components that may contain stimulatory molecules

What are the current challenges in reconciling in vitro and in vivo findings regarding MIP-1α function in mouse models?

Researchers face several challenges when attempting to translate in vitro findings about MIP-1α to in vivo mouse models:

  • Functional discrepancies:

    • In vitro studies demonstrate MIP-1α's inhibitory effects on hematopoietic stem cell proliferation, yet MIP-1α knockout mice show no overt hematopoietic abnormalities

    • This suggests compensatory mechanisms or context-dependent functions that may not be apparent in simplified in vitro systems

  • Concentration-dependent effects:

    • In vitro experiments often use concentrations that may not reflect physiological levels

    • The table below provides a comparison of typical concentrations:

ContextMIP-1α Concentration RangeFunctional Effects
In vitro cell culture10-200 pg/mLChemotaxis, activation
In vivo basal levels5-40 pg/mLHomeostatic functions
In vivo inflammation50-500+ pg/mLEnhanced recruitment, potential synergistic effects
  • Methodological approach to reconciliation:

    • Use identical reagents across in vitro and in vivo experiments when possible

    • Perform dose-response studies that span physiologically relevant concentrations

    • Validate in vitro findings using ex vivo analyses of cells from the same mouse models

    • Consider microenvironmental factors present in vivo but absent in vitro

    • Implement tissue-specific conditional knockout models rather than relying solely on global knockouts

  • Temporal dynamics:

    • In vitro systems often fail to capture the temporal complexity of in vivo responses

    • Utilize real-time imaging or repeated sampling approaches to better understand dynamic processes

How does MIP-1α interact with other chemokines in murine inflammatory models, and what analytical approaches can distinguish their specific contributions?

The complex interplay between MIP-1α and other chemokines in murine inflammatory models requires sophisticated analytical approaches:

  • Key interactions and redundancies:

    • MIP-1α and MIP-1β share 68% homology and overlapping but distinct receptor binding profiles

    • Both chemokines exert similar effects on monocytes but differentially attract lymphocyte subsets (MIP-1α attracts CD8+ T cells while MIP-1β attracts CD4+ T cells)

    • Functional redundancy may explain why single chemokine knockout models often show subtle phenotypes

  • Receptor binding complexity:

    • Murine MIP-1α binds to multiple receptors including CCR1, CCR4, and CCR5

    • These receptors also interact with other chemokines, creating a complex signaling network

  • Analytical approaches to distinguish specific contributions:

    • Combinatorial knockout models: Generate mice lacking multiple chemokines or receptors

    • Receptor-specific antagonists: Use pharmacological tools to block specific receptor-ligand interactions

    • Conditional expression systems: Employ inducible promoters to control temporal expression

    • Cell-specific deletions: Use Cre-lox systems targeting specific immune cell populations

  • Advanced methodological recommendations:

    • Employ multiplexed detection systems to simultaneously measure multiple chemokines

    • Use phospho-flow cytometry to assess receptor-specific signaling events

    • Implement intravital imaging to visualize cell-specific responses in real-time

    • Apply computational modeling to predict redundancies and unique functions based on concentration gradients and receptor expression patterns

What are the critical variables to control when designing experiments to study MIP-1α function in mouse models?

When designing experiments to investigate MIP-1α function in mouse models, researchers should carefully control several critical variables:

  • Genetic background considerations:

    • Use appropriate background strain controls, as chemokine responses vary significantly between common laboratory strains (C57BL/6, BALB/c, etc.)

    • Document the generation number of transgenic or knockout lines to account for potential genetic drift

    • Consider using littermate controls whenever possible to minimize confounding variables

  • Age and sex influences:

    • MIP-1α expression and function can vary with age and sex

    • Standardize age ranges within experimental groups

    • Either use single-sex cohorts or ensure balanced sex representation with sufficient power for subgroup analysis

  • Environmental factors:

    • Microbiome composition influences baseline inflammation and chemokine expression

    • Housing conditions (conventional vs. specific pathogen-free vs. germ-free) significantly impact results

    • Standardize diet, light cycles, and handling procedures

  • Experimental stimuli standardization:

    • Use defined lots of stimulatory agents with consistent potency

    • For infectious models, standardize pathogen preparation, dose, and route of administration

    • Document the timing of interventions relative to circadian rhythms, which affect immune responses

  • Sampling methodologies:

    • Standardize sample collection procedures, including anesthesia method if used

    • Control for stress responses, which can rapidly alter chemokine levels

    • Use consistent anticoagulants for blood collection and standardize processing times

How can researchers effectively differentiate between direct and indirect effects of MIP-1α in complex disease models?

Differentiating between direct and indirect effects of MIP-1α in complex disease models requires sophisticated experimental approaches:

  • Temporal analysis strategies:

    • Implement detailed time-course experiments to establish cause-effect relationships

    • Use inducible expression systems to trigger MIP-1α production at specific timepoints

    • Apply pathway inhibitors at different stages to identify dependent and independent effects

  • Cell-specific approaches:

    • Utilize cell-specific knockout models (e.g., macrophage-specific MIP-1α deletion)

    • Perform adoptive transfer experiments with defined cell populations

    • Use bone marrow chimeras to distinguish between hematopoietic and non-hematopoietic sources

  • Receptor-based strategies:

    • Apply receptor-specific antagonists to block discrete signaling pathways

    • Use receptor knockout models in parallel with ligand knockout models

    • Implement receptor reporter systems to identify responsive cells in situ

  • In vitro validation systems:

    • Develop complex co-culture systems that recapitulate critical cellular interactions

    • Use conditioned media experiments with selective depletion of specific factors

    • Implement transwell systems to distinguish contact-dependent from soluble factor-mediated effects

  • Recommended experimental design framework:

    • Begin with global effect characterization in whole-animal models

    • Follow with tissue-specific analyses to localize key responses

    • Perform cell-specific studies to identify primary responding populations

    • Validate in simplified in vitro systems with defined components

    • Confirm findings using complementary gain-of-function and loss-of-function approaches

How should researchers interpret contradictory data between different detection methods for mouse MIP-1α?

When faced with contradictory results from different detection methods for mouse MIP-1α, researchers should implement a systematic approach to data interpretation and validation:

  • Methodological comparison analysis:

    • Consider the fundamental differences between detection platforms:

MethodPrincipleStrengthsLimitations
ELISAAntibody sandwichQuantitative, specificLimited to single analyte, potential antibody cross-reactivity
Multiplex bead arrayMultiple antibody-bead conjugatesMultiple analytes, small sample volumePotential for cross-reactivity, higher background
BioassayFunctional cellular responseMeasures biological activityIndirect measure, influenced by multiple factors
RT-qPCRmRNA quantificationSensitive, specificMeasures transcription not protein
Western blotProtein size separationVisual confirmation of specificitySemi-quantitative, less sensitive
  • Sample-specific considerations:

    • Matrix effects may differentially impact assay performance

    • Post-translational modifications might affect epitope recognition

    • Binding partners present in biological samples may mask detection sites

  • Validation approach recommendations:

    • Perform spike recovery experiments to assess matrix effects

    • Compare recombinant standards across platforms

    • Use samples from knockout mice as negative controls

    • Apply orthogonal detection methods to the same samples

    • Consider biological validation using neutralizing antibodies in functional assays

  • Interpretation framework:

    • Prioritize data from methods with documented validation using natural MIP-1α

    • Consider whether differences reflect detection of distinct molecular forms

    • Evaluate whether discrepancies correlate with functional outcomes

    • Document and report all methodological details to facilitate reproducibility

What are the best practices for establishing physiologically relevant MIP-1α concentration thresholds in mouse models?

Establishing physiologically relevant concentration thresholds for MIP-1α in mouse models requires careful consideration of multiple factors:

  • Baseline determination strategies:

    • Measure MIP-1α levels across multiple tissue compartments in healthy mice

    • Document strain, age, and sex-specific variations in basal expression

    • Establish diurnal patterns of expression through time-course sampling

  • Pathophysiological context calibration:

    • Characterize concentration ranges across multiple disease models

    • Document concentration gradients within affected tissues

    • Correlate local concentrations with cellular infiltration and activation

  • Dose-response experimental design:

    • Implement in vivo dose-response studies using recombinant MIP-1α

    • Establish threshold concentrations required for specific biological responses

    • Determine saturation levels where additional MIP-1α produces no further effect

  • Correlation with functional outcomes:

    • For each model system, establish concentration ranges that correlate with:

      • Immune cell recruitment (minimal effective concentration)

      • Tissue pathology (pathological threshold)

      • Systemic effects (spillover threshold)

  • Technical and biological recommendations:

    • Standardize sample collection and processing protocols

    • Include physiologically relevant reference samples in each experimental run

    • Consider local concentration effects versus systemic levels

    • Document the relationship between tissue MIP-1α concentrations and soluble receptor levels

How can modern genetic approaches advance our understanding of MIP-1α function in mouse models?

Advanced genetic tools provide powerful approaches to investigate MIP-1α biology in unprecedented detail:

  • CRISPR/Cas9 applications:

    • Generate precise point mutations to study specific functional domains

    • Create reporter knock-in models for real-time visualization of expression

    • Develop conditional alleles for temporal and spatial control of expression

    • Implement multiplexed editing to target MIP-1α alongside interacting partners

  • Single-cell approaches:

    • Apply single-cell RNA sequencing to identify MIP-1α-producing populations with high resolution

    • Implement spatial transcriptomics to map expression patterns within complex tissues

    • Use cellular indexing of transcriptomes and epitopes (CITE-seq) to correlate protein and mRNA expression

  • Inducible expression systems:

    • Develop tetracycline-responsive MIP-1α expression models

    • Create chemically induced proximity systems for rapid activation

    • Implement optogenetic control of MIP-1α expression for precise spatial and temporal regulation

  • Humanized mouse models:

    • Generate mice expressing human MIP-1α variants to study polymorphisms associated with disease

    • Create chimeric models with human immune system components to better translate findings

    • Study interactions with human pathogens in relevant contexts

  • Methodological recommendations:

    • Validate genetic modifications with multiple detection methods

    • Implement complementary gain-of-function and loss-of-function approaches

    • Consider compensatory mechanisms that may emerge during development

    • Document off-target effects and characterize founders thoroughly before establishing lines

What emerging technologies show promise for more comprehensive analysis of MIP-1α dynamics in mouse inflammatory models?

Several cutting-edge technologies are transforming our ability to analyze MIP-1α dynamics in inflammatory contexts:

  • Intravital imaging approaches:

    • Multiphoton microscopy with fluorescent reporter mice allows real-time visualization of MIP-1α-producing cells

    • Bioluminescence resonance energy transfer (BRET) sensors can detect receptor activation dynamics

    • Tissue-clearing techniques combined with light-sheet microscopy enable whole-organ analysis

  • Protein interaction and modification analysis:

    • Proximity labeling techniques (BioID, APEX) can identify novel interaction partners

    • Mass spectrometry-based approaches detect post-translational modifications affecting function

    • Protein correlation profiling maps changes in complex formation during inflammatory responses

  • Systems biology integration:

    • Multi-omics approaches correlate transcriptomic, proteomic, and metabolomic changes

    • Machine learning algorithms identify patterns not apparent through conventional analysis

    • Network modeling predicts key nodes and potential therapeutic targets

  • In situ detection advances:

    • Multiplexed ion beam imaging (MIBI) allows simultaneous detection of dozens of proteins

    • Digital spatial profiling provides region-specific transcriptomic and proteomic data

    • In situ sequencing technologies map cellular responses with spatial context

  • Nanobody and aptamer-based tools:

    • Develop highly specific detection reagents with reduced interference

    • Create intracellular sensors for real-time monitoring

    • Engineer targeting systems for cell-specific intervention

Product Science Overview

Introduction

Macrophage Inflammatory Protein-1 Alpha (MIP-1α), also known as CCL3, is a chemokine that plays a crucial role in the immune response. It is secreted by macrophages and other cell types and is involved in various biological processes, including inflammation, wound healing, and immune cell recruitment.

Discovery and Nomenclature

MIP-1α/CCL3 was first discovered by Stephen D. Wolpe in 1988 . It belongs to the CC subfamily of chemokines, which are characterized by two adjacent cysteine residues near their amino terminus. The protein is also known by several other names, including C-C motif chemokine 3, Heparin-binding chemotaxis protein, and Small-inducible cytokine A3 .

Biological Functions

MIP-1α/CCL3 is a multifunctional peptide that performs various biological functions:

  • Recruitment of Inflammatory Cells: It attracts macrophages, lymphocytes, and eosinophils to sites of inflammation via the CCR1 or CCR5 receptors .
  • Wound Healing: It plays a role in the wound healing process by recruiting cells necessary for tissue repair .
  • Bone Resorption: MIP-1α/CCL3 activates bone resorption cells and directly induces bone destruction, making it significant in conditions involving bone resorption .
  • Immune Response: It maintains the effector immune response and has strong HIV-suppressive activity .
Role in Diseases

MIP-1α/CCL3 is associated with various inflammatory diseases and conditions that exhibit bone resorption, such as:

  • Periodontitis: Elevated levels of MIP-1α/CCL3 are found in patients with periodontitis, indicating its role in the disease’s pathogenesis .
  • Multiple Myeloma: The protein is involved in the bone destruction seen in multiple myeloma .
  • Rheumatoid Arthritis: MIP-1α/CCL3 contributes to the inflammatory processes in rheumatoid arthritis .
  • Sjögren Syndrome: It is also implicated in the pathogenesis of Sjögren syndrome .
Recombinant Mouse MIP-1α/CCL3

Recombinant Mouse MIP-1α/CCL3 is a full-length protein expressed in HEK 293 cells. It is used in various research applications, including high-performance liquid chromatography (HPLC), sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE), and mass spectrometry (MS) . The recombinant protein is highly pure, with a purity level of ≥95% and an endotoxin level of ≤0.005 EU/µg .

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.