MCP 4 Human

Monocyte Chemotactic Protein-4 Human Recombinant (CCL13)
Shipped with Ice Packs
In Stock

Description

Biological Functions and Mechanisms

MCP-4 binds to G-protein-coupled receptors (GPCRs), primarily CCR2, CCR3, and CCR5, to mediate immune cell migration and activation . Key functional roles include:

Immune Cell Recruitment

  • Eosinophils: Induces chemotaxis and respiratory burst, releasing reactive oxygen species (ROS) at concentrations of 10–200 ng/mL .

  • Basophils: Triggers histamine release and calcium flux .

  • Monocytes and T-lymphocytes: Promotes migration to inflammatory sites .

Tissue-Specific Expression

  • Dermal Fibroblasts: Express MCP-4 mRNA in response to IFN-γ, IL-4, and TNF-α, suggesting a role in allergic and autoimmune diseases .

  • Asthma and Sinusitis: Elevated in bronchoalveolar lavage fluid and nasal epithelia of asthmatic and sinusitis patients .

Signaling Pathways

MCP-4 activates signaling cascades involving:

  1. G-protein activation via CCR2/3/5.

  2. Protein kinase C (PKC) and tyrosine kinase pathways.

  3. Phosphatidylinositol-3-kinase (PI3K), critical for actin polymerization and ROS production .

Clinical Relevance and Pathological Associations

MCP-4 is implicated in chronic inflammatory and autoimmune disorders:

ConditionMCP-4 RoleEvidence
AsthmaEosinophil recruitment in airwaysElevated in sputum and BAL fluid
Atopic DermatitisT-cell infiltrationLinked to Th2-driven inflammation
Major Depressive Disorder (MDD)NeuroinflammationAltered serum levels correlate with MDD severity
Renal InflammationMonocyte recruitmentObserved in kidney disease models

Recombinant Proteins

Commercially available MCP-4 variants include:

VendorPurityActivityCatalog ID
BPS Bioscience≥98%Chemoattractant (20–40 ng/mL)90214-B
R&D SystemsN/ABioassay applications327-P4
PeproTech≥95%Immunoassay and signaling studies300-24

Detection Techniques

  • SELDI-TOF-MS: Identifies two isoforms in human serum, suggesting post-translational modifications .

  • Crystallography: Resolves MCP-4 structure at 1.70 Å, highlighting dimerization interfaces .

Signaling Receptors and Cross-Talk

MCP-4 interacts with CCR2, CCR3, and CCR5, receptors shared with other CC chemokines (e.g., MCP-1, MCP-2). Its dual binding to CCR3 (eotaxin receptor) and CCR2 (MCP-1 receptor) allows synergistic effects in eosinophil and monocyte activation .

Product Specs

Introduction
Chemokine (C-C motif) ligand 13 (CCL13 / MCP-4) is a small cytokine that belongs to the CC chemokine family. This chemokine is found on chromosome 17 in humans, within a cluster of other CC chemokines. MCP-4 binds to cell surface chemokine receptors such as CCR2, CCR3, and CCR5, which induces chemotaxis in monocytes, eosinophils, T lymphocytes, and basophils. It plays a role in allergic reactions, including asthma. The production of MCP-4 can be induced by inflammatory cytokines such as interleukin-1 and TNF-α.
Description
Recombinant Human Monocyte Chemotactic Protein-4 is produced in E. coli. It is a non-glycosylated polypeptide chain containing 75 amino acids with a molecular mass of 8.6 kDa. This MCP-4 protein is purified using proprietary chromatographic techniques.
Physical Appearance
White, lyophilized powder, sterile-filtered.
Formulation
The protein is lyophilized from a sterile solution of 20mM PB, pH 7.4, and 130mM NaCl at a concentration of 1 mg/ml.
Solubility
Reconstitute the lyophilized MCP-4 in sterile 18M-cm H2O to a concentration of at least 100 µg/ml. This solution can be further diluted with other aqueous solutions.
Stability
Lyophilized MCP-4 remains stable at room temperature for 3 weeks; however, it should be stored desiccated at -18°C or below. After reconstitution, MCP-4 should be stored at 4°C for 2-7 days. For long-term storage, add a carrier protein (0.1% HSA or BSA). Avoid freeze-thaw cycles.
Purity
Purity exceeds 96.0% as determined by: (a) Reverse-phase high-performance liquid chromatography (RP-HPLC) analysis and (b) SDS-PAGE analysis.
Biological Activity
Specific activity is measured as the ability of MCP-4 to chemoattract human monocytes at a concentration of 10-100 ng/ml, which corresponds to a specific activity of 10,000-100,000 units/mg.
Synonyms
Small inducible cytokine A13, CCL13, Monocyte chemotactic protein 4, MCP-4, Monocyte chemoattractant protein 4, CK-beta-10, NCC-1, chemokine (C-C motif) ligand 13, NCC1, CKb10, SCYL1, SCYA13, MGC17134.
Source
Escherichia Coli.
Amino Acid Sequence
QPDALNVPSTCCFTFSSKKISLQRLKSYVITTSRCPQKAV
IFRTKLGKEICADPKEKWVQNYMKHLGRKAHTLKT.

Q&A

What is MCP-4 and what is its role in the human immune system?

MCP-4 (Monocyte Chemoattractant Protein-4), also known as CCL13, is a member of the CC chemokine family that regulates recruitment of immune cells into inflammatory foci. It consists of 76 amino acid residues and has a molecular weight of approximately 9 kDa. As a critical mediator of immune cell trafficking, MCP-4 functions as a potent chemoattractant for multiple cell types including monocytes, eosinophils, basophils, and T lymphocytes .

MCP-4 exerts its biological activities by binding to specific G protein-coupled receptors, primarily CCR2B, CCR3, and CCR5, initiating intracellular signaling cascades that promote directed cell migration . This selective receptor engagement explains MCP-4's ability to simultaneously recruit different immune cell populations to sites of inflammation. The protein has been implicated in the pathogenesis of various inflammatory conditions including allergic respiratory diseases, renal inflammation, and atopic dermatitis .

How does MCP-4 differ structurally and functionally from other chemokines in the MCP family?

MCP-4 belongs to a distinct structurally-related subclass of CC chemokines, sharing sequence homology with other MCP family members. While functionally similar to other MCPs in recruiting monocytes, MCP-4 has a broader spectrum of cellular targets and receptor binding profiles. Unlike some other chemokines that signal primarily through a single receptor, MCP-4 engages multiple receptors (CCR2, CCR3, and CCR5), giving it versatility in immune cell recruitment .

Functionally, MCP-4 demonstrates potent eosinophil chemoattractant properties comparable to eotaxin. Research has shown that MCP-4 and eotaxin cause similar increases in eosinophil intracytoplasmic Ca²⁺ and exhibit complete cross-desensitization. Furthermore, MCP-4 can abolish eosinophil Ca²⁺ responses to MCP-3 and partially desensitize responses to macrophage inflammatory protein-1alpha, indicating significant receptor overlap and functional interactions between these chemokines .

What cellular mechanisms regulate MCP-4 expression?

MCP-4 expression is tightly regulated by inflammatory cytokines and immunomodulatory agents. Several key regulatory mechanisms have been identified:

  • Cytokine induction: MCP-4 is strongly induced by inflammatory cytokines, particularly TNF-alpha and IL-1 in epithelial and endothelial cells .

  • Synergistic regulation: Both Th1 and Th2 cytokines can enhance MCP-4 expression. IFN-gamma (Th1 cytokine) and IL-4 (Th2 cytokine) synergize with TNF-alpha and IL-1 to amplify MCP-4 mRNA accumulation, explaining its presence in both allergic and non-allergic inflammatory conditions .

  • Glucocorticoid inhibition: Anti-inflammatory glucocorticoids like budesonide can inhibit MCP-4 mRNA expression, as demonstrated in BEAS-2B epithelial cells, providing a mechanism for steroid efficacy in inflammatory conditions .

  • Cell-specific expression patterns: MCP-4 mRNA is prominently expressed in human lung and heart tissues, and can be detected in bronchoalveolar lavage cells from both asthmatic and non-asthmatic subjects .

These regulatory mechanisms make MCP-4 expression responsive to the local inflammatory environment, allowing context-specific immune cell recruitment.

What are the most reliable methods for measuring MCP-4 levels in human biological samples?

Several validated methodologies exist for measuring MCP-4 in human biological samples, each with specific advantages depending on research objectives:

  • Enzyme-Linked Immunosorbent Assay (ELISA): The Human MCP-4 solid-phase sandwich ELISA represents the gold standard for quantifying MCP-4 in human serum, plasma, or cell culture medium. This method uses target-specific antibodies pre-coated in microplate wells, followed by addition of detector antibodies and substrate solution to generate a measurable signal proportional to MCP-4 concentration. Modern ELISA kits undergo rigorous validation for sensitivity, specificity, precision, and lot-to-lot consistency .

  • Electrochemiluminescence Assays: U-PLEX Human MCP-4 Assays utilize electrochemiluminescence technology on platforms like SECTOR plates or QuickPlex Ultra plates. These assays offer enhanced sensitivity and wider dynamic range compared to traditional ELISA .

  • In situ hybridization: For tissue localization of MCP-4 expression, in situ hybridization identifies cells actively producing MCP-4 mRNA in tissue sections, revealing spatial distribution patterns of expression .

  • Immunohistochemistry: Complementary to in situ hybridization, immunohistochemical analysis with specific anti-MCP-4 antibodies can visualize protein distribution in tissue sections, allowing correlation with inflammatory infiltrates .

When selecting a measurement approach, researchers should consider factors including sample type, expected concentration range, availability of specialized equipment, and whether protein or mRNA quantification is more relevant to their research question.

How can I optimize an ELISA protocol to detect low concentrations of MCP-4 in complex biological matrices?

Optimizing ELISA protocols for detecting low concentrations of MCP-4 in complex matrices requires systematic attention to multiple technical factors:

  • Sample preparation optimization:

    • For serum/plasma: Consider overnight incubation at 4°C to improve antigen-antibody binding kinetics

    • For tissue homogenates: Optimize extraction buffers with appropriate detergents and protease inhibitors to maximize MCP-4 recovery while minimizing matrix interference

    • Implement pre-clearing steps (centrifugation, filtration) to remove particulates that could interfere with antibody binding

  • Assay sensitivity enhancement:

    • Extend primary antibody incubation time (overnight at 4°C)

    • Optimize substrate development time through careful time-course experiments

    • Consider signal amplification systems (e.g., avidin-biotin complexes) when extremely low detection limits are required

    • Implement sample concentration techniques for very dilute samples

  • Background reduction strategies:

    • Increase blocking reagent concentration (3-5% BSA or specialized blocking solutions)

    • Add carrier proteins to diluents to minimize non-specific binding

    • Incorporate additional washing steps with mild detergents to reduce background signal

    • Test different plate types to minimize non-specific binding

  • Standard curve optimization:

    • Expand the lower range of the standard curve with additional dilution points

    • Prepare fresh recombinant MCP-4 standards of verified bioactivity

    • Use the same matrix for standards as for samples (matrix-matched calibration)

Always validate optimized protocols by assessing recovery of known spike-in concentrations of recombinant MCP-4 in actual sample matrices to confirm assay accuracy across the detection range.

What approaches can be used to detect MCP-4 receptor expression and functionality in primary human cells?

Detection of MCP-4 receptor expression and functionality in primary human cells requires complementary techniques addressing different aspects of receptor biology:

  • Receptor expression analysis:

    • Flow cytometry with fluorophore-conjugated antibodies against CCR2, CCR3, and CCR5 provides quantitative single-cell analysis of receptor surface expression

    • Quantitative RT-PCR measures receptor mRNA expression levels when antibodies are unavailable or to complement protein detection

    • Immunohistochemistry/immunofluorescence localizes receptors in tissue contexts, allowing correlation with anatomical structures and cellular populations

  • Functional receptor assays:

    • Calcium flux measurements: As demonstrated in research with eosinophils, measuring intracytoplasmic Ca²⁺ mobilization following MCP-4 stimulation provides direct evidence of functional receptor signaling

    • Chemotaxis assays: Transwell migration assays quantify directional cell movement toward MCP-4 gradients, demonstrating functional chemotactic responses

    • Receptor internalization: Measuring receptor disappearance from cell surface after MCP-4 exposure indicates receptor engagement and consequent endocytosis

  • Receptor specificity determination:

    • Cross-desensitization experiments: Sequential stimulation with MCP-4 and other chemokines (e.g., eotaxin, MCP-3) followed by calcium flux measurements can identify shared receptor usage

    • Competitive binding assays: Using radiolabeled ligands like ¹²⁵I-eotaxin or ¹²⁵I-MCP-1 with unlabeled MCP-4 as competitor identifies receptor binding specificity

    • Receptor antagonist studies: Selective receptor antagonists can block specific receptors to determine which are required for MCP-4 signaling

  • Cell type-specific receptor function:

    • Isolation of primary cell populations (monocytes, eosinophils, T cells) followed by receptor analysis provides cell type-specific insights

    • Single-cell approaches combining receptor detection with functional readouts can reveal heterogeneity within seemingly homogeneous populations

These complementary approaches provide a comprehensive view of MCP-4 receptor biology in primary human cells, critical for understanding differential responses across immune cell populations.

What is the evidence implicating MCP-4 in allergic respiratory diseases?

Multiple lines of evidence implicate MCP-4 in allergic respiratory diseases, particularly asthma:

  • Elevated expression in affected tissues:

    • MCP-4 mRNA has been detected in bronchoalveolar lavage cells from asthmatic subjects

    • Expression is greater in sputum, epithelium, submucosal inflammatory cells, and bronchoalveolar lavage fluid of asthmatics compared to healthy individuals

  • Cellular sources in airways:

    • Human bronchial epithelial cell lines express MCP-4 mRNA after cytokine stimulation

    • Epithelial cells represent a critical source of MCP-4 at the air-tissue interface, where initial allergen contact occurs

  • Functional effects on key cells in allergic inflammation:

    • MCP-4 induces potent chemotaxis of eosinophils, central effector cells in allergic airway inflammation

    • It stimulates histamine release from IL-3-primed peripheral blood basophils, contributing to immediate hypersensitivity reactions

    • It attracts T lymphocytes that can perpetuate allergic responses through cytokine production

  • Receptor engagement relevant to allergic pathways:

    • MCP-4 signals through CCR3, the eotaxin receptor selectively expressed on eosinophils

    • Complete cross-desensitization experiments between MCP-4 and eotaxin indicate shared signaling pathways central to allergic cell recruitment

  • Responsiveness to therapeutic interventions:

    • Glucocorticoids, first-line anti-inflammatory treatments for asthma, inhibit MCP-4 mRNA expression in bronchial epithelial cells (demonstrated with budesonide in BEAS-2B cells)

These findings collectively position MCP-4 as a significant mediator in allergic respiratory pathophysiology, capable of orchestrating recruitment and activation of multiple inflammatory cell types central to allergic responses.

How does MCP-4 contribute to renal inflammatory conditions?

MCP-4 plays a significant role in renal inflammatory conditions through several mechanisms:

  • Expression patterns in kidney inflammation:

    • In situ hybridization and immunohistochemical analyses of biopsy material from patients with acute renal allograft rejection and vasculitic glomerulonephritis have demonstrated MCP-4 expression at both mRNA and protein levels

    • MCP-4 is primarily expressed at peritubular, periglomerular, and perivascular sites, irrespective of the specific inflammatory condition

  • Cellular sources within kidneys:

    • Proximal tubular epithelial cells grown in culture from cortical fragments of human kidney show low levels of constitutive MCP-4 expression, detectable by western blotting

    • This expression is upregulated in response to pro-inflammatory cytokines TNF-alpha and IFN-gamma, suggesting local amplification during inflammation

  • Association with inflammatory infiltrates:

    • MCP-4 expression is associated with infiltrating CD3-positive lymphocytes and CD68-positive monocyte/macrophages

    • CCR2-, CCR3-, and CCR5-expressing leukocyte populations are identified at sites of MCP-4 expression, with double-staining techniques revealing that these chemokine receptor-expressing cells are primarily CD68-positive (monocytes/macrophages)

  • Proposed pathogenic mechanism:

    • MCP-4 appears to function as a critical mediator in the recruitment and retention of monocytes/macrophages in renal inflammation

    • The strategic localization at peritubular, periglomerular, and perivascular sites positions MCP-4 to direct inflammatory cells toward key functional structures within the kidney

These findings suggest that MCP-4 contributes to the development and progression of renal inflammatory conditions by orchestrating the trafficking of inflammatory cells to specific kidney compartments, potentially representing a therapeutic target for kidney diseases characterized by mononuclear cell infiltration.

What is the role of MCP-4 in the differential recruitment of immune cells in Th1 versus Th2 inflammatory conditions?

MCP-4 demonstrates a remarkable capacity to participate in both Th1 and Th2 inflammatory conditions through complex regulation of differential immune cell recruitment:

  • Dual expression in distinct inflammatory phenotypes:

    • MCP-4 is expressed in the epithelial mucosa of patients with both Th2-type allergic and Th1-type nonallergic sinusitis

    • This dual expression pattern distinguishes MCP-4 from chemokines restricted to either Th1 or Th2 inflammatory environments

  • Cytokine-dependent regulation across inflammatory phenotypes:

    • Both IFN-gamma (the prototypical Th1 cytokine) and IL-4 (the signature Th2 cytokine) synergize with TNF-alpha and IL-1 in inducing MCP-4 mRNA accumulation

    • This synergistic regulation by both Th1 and Th2 cytokines provides a molecular explanation for MCP-4's presence in divergent inflammatory conditions

  • Cell-selective recruitment patterns:

    • Through engagement of multiple chemokine receptors (CCR2, CCR3, CCR5), MCP-4 can simultaneously attract different immune cell populations expressing various receptor combinations

    • CCR3 engagement preferentially recruits eosinophils and basophils (Th2-associated cells)

    • CCR2 and CCR5 engagement attracts monocytes and specific T cell subsets that can participate in either Th1 or Th2 responses

  • Functional consequences in tissue inflammation:

    • In Th2-predominant allergic conditions, MCP-4 contributes to eosinophil and basophil recruitment, histamine release, and consequent allergic inflammation

    • In Th1-predominant inflammatory conditions, MCP-4 may preferentially recruit monocytes/macrophages and Th1 cells, promoting cell-mediated immunity

This versatility in both induction pathways and cellular targeting allows MCP-4 to function as a bridge between innate and adaptive immunity in diverse inflammatory contexts, offering a molecular explanation for the observed accumulation of monocytes, eosinophils, and basophils in both Th1- and Th2-type immune responses .

How should I design experiments to investigate MCP-4's role in human inflammatory diseases?

Designing robust experiments to investigate MCP-4's role in human inflammatory diseases requires a comprehensive approach spanning multiple levels of analysis:

  • Clinical sample acquisition and characterization:

    • Collect matched samples from patients with inflammatory disease and appropriate controls (consider age, sex, medication status)

    • Obtain samples from relevant anatomical sites (e.g., bronchoalveolar lavage for respiratory diseases, kidney biopsies for renal inflammation)

    • Thoroughly document clinical parameters, disease severity, and treatment history

    • Consider longitudinal sampling to capture disease progression and treatment response

  • Expression analysis strategy:

    • Implement parallel quantification of MCP-4 at protein level (ELISA, immunohistochemistry) and mRNA level (qRT-PCR, in situ hybridization)

    • Correlate MCP-4 levels with disease-relevant parameters and other inflammatory mediators

    • Determine cellular sources through co-localization studies combining MCP-4 detection with cell-specific markers

    • Analyze receptor expression (CCR2, CCR3, CCR5) on relevant immune cell populations

  • Functional studies with patient-derived material:

    • Isolate primary cells from patients and controls (e.g., airway epithelial cells, kidney cells, immune cells)

    • Assess their MCP-4 production capacity under basal and stimulated conditions

    • Evaluate responses to disease-relevant stimuli and therapeutic agents

    • Compare chemotactic responses of immune cells toward MCP-4 between patients and controls

  • Mechanistic investigations using in vitro models:

    • Develop disease-relevant cell culture systems (e.g., air-liquid interface cultures for respiratory studies)

    • Apply cytokines found elevated in the specific disease to induce MCP-4

    • Use receptor antagonists or neutralizing antibodies to block MCP-4 function

    • Consider siRNA knockdown or CRISPR-based approaches to modulate MCP-4 expression

  • Translational relevance:

    • Correlate experimental findings with treatment responses in patients

    • Assess MCP-4 as a potential biomarker for disease activity or treatment response

    • Evaluate effects of current therapies on MCP-4 expression and function

This multi-faceted approach addresses MCP-4 biology from complementary angles, strengthening causal inferences about its role in human inflammatory diseases.

What cell culture models are most appropriate for studying MCP-4 production and function?

Selecting appropriate cell culture models for studying MCP-4 production and function requires consideration of disease relevance, physiological accuracy, and technical feasibility:

  • Epithelial cell models:

    • Bronchial epithelial cell lines (e.g., BEAS-2B) have demonstrated cytokine-inducible MCP-4 expression and responsiveness to glucocorticoid inhibition

    • Primary human bronchial epithelial cells cultured at air-liquid interface provide a physiologically relevant model that maintains epithelial polarization and differentiation

    • Proximal tubular epithelial cells cultured from cortical fragments of human kidney exhibit both constitutive and inducible MCP-4 expression, making them valuable for studying renal inflammation

  • Endothelial cell models:

    • Human umbilical vein endothelial cells (HUVECs) activated with TNF-alpha and IL-1 express MCP-4

    • Tissue-specific endothelial cells (e.g., lung microvascular endothelial cells) may better represent organ-specific vascular responses

    • Co-culture systems with endothelial cells and immune cells allow assessment of transendothelial migration in response to MCP-4

  • Immune cell models:

    • Peripheral blood mononuclear cells enable studies of MCP-4 effects on primary human monocytes and lymphocytes

    • Purified eosinophils and basophils allow detailed investigation of MCP-4-induced chemotaxis and activation

    • IL-3-primed basophils provide a model for studying MCP-4-induced histamine release

    • Bone marrow-derived cells can model developmental effects on immune cell responsiveness

  • Receptor expression systems:

    • Cell lines transfected with individual chemokine receptors (CCR2b, CCR3, CCR5) enable dissection of receptor-specific responses to MCP-4

    • Mouse lymphoma cells transfected with human chemokine receptors have successfully demonstrated MCP-4-activated cell migration via either CCR2b or CCR3

  • Three-dimensional and co-culture models:

    • Spheroid cultures better approximate in vivo tissue architecture and cell-cell interactions

    • Multi-cell type co-cultures (e.g., epithelial cells with immune cells) model complex interactions in inflamed tissues

    • Microfluidic "organ-on-chip" platforms enable real-time visualization of cell recruitment along MCP-4 gradients

The optimal model system should be selected based on specific research questions, balancing physiological relevance with technical feasibility and reproducibility.

What are the key considerations for designing experiments to test MCP-4 antagonists or inhibitors?

Designing experiments to evaluate MCP-4 antagonists or inhibitors requires careful attention to multiple factors that influence efficacy assessment and translational relevance:

  • Target validation and specificity considerations:

    • Confirm that your antagonist selectively inhibits MCP-4 without affecting related chemokines

    • Determine whether the antagonist acts by directly binding MCP-4 or by blocking its receptors (CCR2, CCR3, CCR5)

    • For receptor antagonists, characterize specificity against individual receptors using transfected cell lines expressing single receptors

    • Include positive controls with established inhibitors where available

  • In vitro efficacy evaluation framework:

    • Binding assays: Measure displacement of radiolabeled MCP-4 from receptors or competitive inhibition of ¹²⁵I-eotaxin or ¹²⁵I-MCP-1 binding

    • Functional assays: Assess inhibition of:

      • Calcium flux in receptor-expressing cells or primary eosinophils

      • Chemotaxis using Transwell or real-time migration tracking systems

      • Histamine release from IL-3-primed basophils

    • Cell type-specific effects: Test inhibitor efficacy across multiple relevant cell types (monocytes, eosinophils, basophils, T cells)

  • Dose-response relationship characterization:

    • Establish full inhibition curves with wide concentration ranges (at least 5-6 concentrations spanning 3-4 log units)

    • Determine IC₅₀ values for different functional endpoints, which may vary based on receptor density or cellular context

    • Assess potential biphasic effects or paradoxical responses at extreme concentrations

  • Experimental conditions optimization:

    • Test inhibitors in both prevention (pre-incubation before MCP-4 addition) and reversal (addition after MCP-4 stimulation) protocols

    • Evaluate duration of inhibitory effects through time-course experiments

    • Include relevant disease-associated conditions (e.g., hypoxia, acidic pH) that may affect inhibitor efficacy

    • Test in the presence of biological matrices (serum, inflammatory exudates) to account for protein binding effects

  • Translational considerations:

    • Use primary human cells whenever possible, ideally from both healthy donors and patients with relevant inflammatory conditions

    • Include controls with clinically used anti-inflammatory agents like glucocorticoids that inhibit MCP-4 expression

    • Design experiments that model therapeutic intervention at different disease stages

    • Consider combination approaches targeting multiple chemokines or inflammatory pathways

These methodological considerations provide a framework for rigorous evaluation of potential MCP-4-targeting therapeutics while addressing the complexities of chemokine biology in inflammatory diseases.

How can single-cell technologies advance our understanding of MCP-4 biology in human tissues?

Single-cell technologies offer unprecedented insights into MCP-4 biology by revealing cellular heterogeneity, spatial relationships, and dynamic responses that are masked in bulk tissue analyses:

  • Single-cell RNA sequencing (scRNA-seq) applications:

    • Precise identification of MCP-4-producing cell populations within complex tissues, revealing unexpected cellular sources

    • Characterization of heterogeneous receptor expression patterns (CCR2, CCR3, CCR5) across immune cell subsets

    • Trajectory analysis to track temporal evolution of MCP-4 responses during inflammation progression

    • Correlation of MCP-4 expression with global transcriptional programs to identify co-regulated pathways

    • Comparative analysis between healthy and inflamed tissues to identify disease-specific shifts in MCP-4 network activity

  • Single-cell proteomics and secretomics:

    • Mass cytometry (CyTOF) with metal-labeled antibodies against MCP-4 and its receptors provides protein-level quantification in thousands of individual cells

    • Single-cell secretion assays using microwell technologies can quantify MCP-4 production by individual cells upon stimulation

    • Multiparameter characterization linking MCP-4 production to cellular phenotypes and activation states

  • Spatial transcriptomics and in situ technologies:

    • Multiplexed RNA fluorescence in situ hybridization (FISH) localizes MCP-4 expression within anatomical tissue contexts

    • Spatial transcriptomics platforms map MCP-4 expression gradients relative to structural features and inflammatory foci

    • Correlative analyses identifying spatial relationships between MCP-4-producing cells and receptor-expressing cells in intact tissues

    • Sequential immunofluorescence or multiplexed ion beam imaging (MIBI) to simultaneously visualize multiple proteins including MCP-4 and its receptors

  • Live-cell functional imaging:

    • Intravital microscopy in humanized mouse models to visualize MCP-4-mediated cell recruitment in real-time

    • Calcium imaging at single-cell resolution to capture heterogeneous receptor signaling responses to MCP-4

    • CRISPR-Cas9 genome editing combined with fluorescent reporters to track MCP-4 pathway activation

  • Integrative computational approaches:

    • Cell-cell communication analysis algorithms to predict MCP-4-mediated interactions between tissue-resident cells

    • Machine learning approaches to identify cellular states associated with high MCP-4 responsiveness

    • Network analysis linking MCP-4 to broader inflammatory circuits within tissues

These technologies collectively overcome the limitations of population-averaged measurements, revealing the cellular architecture and dynamics of MCP-4 biology with unprecedented resolution in human tissues.

What are the challenges and solutions in developing therapeutic approaches targeting MCP-4 in inflammatory diseases?

Developing therapeutic approaches targeting MCP-4 in inflammatory diseases presents significant challenges that require innovative solutions:

  • Target specificity challenges:

    • Challenge: MCP-4 shares receptors (CCR2, CCR3, CCR5) with multiple other chemokines, risking off-target effects when blocking receptors

    • Solutions:

      • Develop highly selective MCP-4 neutralizing antibodies targeting unique epitopes

      • Design receptor antagonists with binding kinetics or allosteric properties that preferentially disrupt MCP-4 interactions

      • Implement tissue-targeted delivery approaches to concentrate inhibitors at disease sites

  • Redundancy in chemokine networks:

    • Challenge: Functional redundancy among chemokines may limit efficacy of single-target approaches

    • Solutions:

      • Identify disease contexts where MCP-4 plays non-redundant roles

      • Develop dual/multi-targeting approaches addressing complementary chemokine pathways

      • Target downstream convergent signaling pathways common to multiple chemokine receptors

      • Use systems biology approaches to identify critical nodes in chemokine networks

  • Context-dependent functions:

    • Challenge: MCP-4 participates in both Th1 and Th2 inflammatory responses with potentially different roles

    • Solutions:

      • Implement precision medicine approaches with biomarker-guided patient selection

      • Develop context-sensitive therapeutic strategies tailored to specific disease endotypes

      • Design temporal intervention strategies aligned with disease phase-specific roles of MCP-4

  • Balancing efficacy versus immunosuppression:

    • Challenge: Complete MCP-4 inhibition might impair beneficial immune responses

    • Solutions:

      • Establish partial inhibition strategies that modulate rather than ablate MCP-4 function

      • Develop therapeutic windows that normalize rather than eliminate MCP-4 signaling

      • Design pulsed or intermittent dosing regimens that permit recovery periods

      • Create conditional inhibitors that function primarily under inflammatory conditions

  • Translational gaps between models and human disease:

    • Challenge: Animal models may not accurately represent human MCP-4 biology

    • Solutions:

      • Utilize human tissue explant cultures to verify therapeutic effects

      • Develop humanized mouse models expressing human MCP-4 and receptors

      • Implement early-phase adaptive trial designs with robust biomarker assessment

      • Leverage patient-derived organoids for personalized efficacy prediction

  • Delivery and pharmacokinetic considerations:

    • Challenge: Achieving sustained inhibition at inflammatory sites while minimizing systemic exposure

    • Solutions:

      • Develop engineered antibody formats with tissue-targeting domains

      • Implement local delivery approaches for compartmentalized diseases

      • Design prodrug approaches activated by inflammation-associated enzymes

      • Create controlled-release formulations for chronic inflammatory conditions

These integrated approaches address the complex challenges inherent in targeting chemokine networks while maximizing therapeutic potential for MCP-4-directed interventions in inflammatory diseases.

How can systems biology approaches help elucidate MCP-4's role in complex inflammatory networks?

Systems biology approaches offer powerful frameworks for understanding MCP-4's position and functions within complex inflammatory networks:

  • Multi-omics integration strategies:

    • Combine transcriptomics, proteomics, and metabolomics data from inflammatory disease tissues to position MCP-4 within broader response networks

    • Correlate MCP-4 levels with global gene expression patterns to identify co-regulated modules

    • Analyze post-translational modifications of MCP-4 through phosphoproteomics and glycoproteomics to uncover regulatory mechanisms

    • Implement parallel reaction monitoring mass spectrometry to quantify MCP-4 alongside hundreds of other inflammatory mediators simultaneously

  • Network modeling and analysis:

    • Construct dynamical models of chemokine signaling networks incorporating MCP-4 and its receptors

    • Apply Boolean network modeling to predict the effects of MCP-4 perturbation on inflammatory circuits

    • Use Bayesian network inference to discover causal relationships between MCP-4 and other inflammatory mediators

    • Implement sensitivity analysis to identify critical nodes that modulate MCP-4 production or responsiveness

  • Computational analysis of receptor-ligand interactions:

    • Apply molecular dynamics simulations to model MCP-4 interactions with CCR2, CCR3, and CCR5 at atomic resolution

    • Predict binding energetics and conformational changes upon receptor engagement

    • Model competition and synergy between MCP-4 and other chemokines for shared receptors

    • Simulate spatial gradients of MCP-4 within tissue microenvironments using partial differential equation models

  • Multi-scale integration approaches:

    • Develop cell-to-tissue models linking MCP-4-induced single-cell behaviors to tissue-level inflammatory patterns

    • Integrate temporal data across multiple timescales from rapid receptor signaling to chronic disease progression

    • Connect molecular events to cellular decisions and ultimately clinical outcomes through hierarchical modeling

    • Apply agent-based modeling to simulate emergent patterns of leukocyte recruitment driven by MCP-4 gradients

  • Therapeutic response prediction:

    • Develop machine learning algorithms to predict patient responsiveness to MCP-4-targeting therapies

    • Identify network-based biomarkers that indicate central versus peripheral roles of MCP-4 in individual patients

    • Model compensatory mechanisms that might emerge following MCP-4 inhibition

    • Simulate combination therapy approaches targeting multiple nodes in MCP-4-containing networks

These systems approaches move beyond reductionist views of single molecules to understand how MCP-4 functions within interconnected networks, enabling more effective therapeutic strategies that account for system-level properties of inflammatory responses.

What are the most promising future research directions for understanding MCP-4's role in human disease?

The investigation of MCP-4's roles in human disease has yielded significant insights, but several promising research directions remain to be fully explored:

  • Precision medicine applications:

    • Development of MCP-4 as a biomarker for patient stratification in inflammatory diseases

    • Identification of genetic variants affecting MCP-4 production or receptor interactions

    • Correlation of MCP-4 levels with treatment responses to existing therapies

    • Creation of MCP-4-based molecular endotyping approaches for personalized therapeutic selection

  • Tissue microenvironment and chronicity mechanisms:

    • Investigation of how MCP-4 contributes to chronic versus acute inflammation

    • Exploration of the role of MCP-4 in tissue remodeling and fibrosis beyond initial leukocyte recruitment

    • Characterization of MCP-4's interactions with tissue-resident immune cells and non-immune structural cells

    • Analysis of MCP-4's contributions to establishment of inflammatory memory within tissues

  • Broader disease applications:

    • Extension of MCP-4 research beyond currently studied allergic and renal conditions to other inflammatory diseases

    • Investigation of potential roles in metabolic inflammation, neurodegenerative diseases, and cancer

    • Evaluation of MCP-4 as a mediator of comorbidities between different inflammatory conditions

    • Assessment of MCP-4's contributions to aging-associated chronic inflammation

  • Developmental and regulatory biology:

    • Characterization of epigenetic mechanisms controlling MCP-4 expression

    • Investigation of developmental programming affecting MCP-4 responsiveness

    • Elucidation of non-coding RNA regulation of MCP-4 production and signaling

    • Exploration of circadian and environmental influences on MCP-4 biology

  • Novel therapeutic modalities:

    • Development of bispecific antibodies simultaneously targeting MCP-4 and complementary inflammatory mediators

    • Creation of selective receptor modulators that bias signaling away from pathogenic outcomes

    • Implementation of mRNA or antisense therapeutics for targeted inhibition of MCP-4 production

    • Exploration of cell-based therapies engineered to respond to or modulate MCP-4 signaling

These research directions promise to deepen our understanding of MCP-4 biology while accelerating translation of these insights into novel diagnostic and therapeutic applications for inflammatory diseases.

How can inter-disciplinary collaborations advance MCP-4 research and therapeutic development?

Inter-disciplinary collaborations offer transformative potential to accelerate MCP-4 research and therapeutic development by integrating diverse expertise and methodologies:

  • Immunology-structural biology partnerships:

    • Combining immunological insights with structural approaches to visualize MCP-4-receptor interactions through cryo-electron microscopy

    • Applying NMR spectroscopy to characterize dynamic receptor conformational changes upon MCP-4 binding

    • Using structure-based drug design to develop selective MCP-4 antagonists with favorable pharmacological properties

    • Resolving the structural basis for MCP-4's unique ability to engage multiple chemokine receptors

  • Clinical-basic science integration:

    • Establishing biorepositories with matched clinical data and biological samples from inflammatory disease patients

    • Creating closed-loop systems where clinical observations drive laboratory investigations and vice versa

    • Conducting "reverse translational" studies using patient-derived materials to validate laboratory findings

    • Integrating real-world clinical outcomes data with molecular insights about MCP-4 biology

  • Engineering-immunology collaborations:

    • Developing microfluidic systems to model complex MCP-4 gradients and visualize cellular responses

    • Creating biosensors for real-time detection of MCP-4 in biological fluids or tissues

    • Designing controlled-release systems for precise spatiotemporal delivery of MCP-4 modulators

    • Applying tissue engineering approaches to build three-dimensional models of MCP-4-mediated inflammation

  • Computational-experimental synergies:

    • Implementing machine learning to identify patterns in large-scale MCP-4 datasets that escape human detection

    • Using artificial intelligence to predict optimal combination therapies involving MCP-4 modulation

    • Developing in silico clinical trials to predict outcomes of MCP-4-targeting therapeutic strategies

    • Creating digital twins of inflammatory microenvironments to simulate MCP-4 intervention effects

  • Public health-molecular biology connections:

    • Correlating population-level inflammatory disease patterns with MCP-4 genetic variants

    • Investigating environmental factors that modulate MCP-4 expression or function

    • Exploring cost-effectiveness of precision medicine approaches based on MCP-4 biomarkers

    • Developing implementation science strategies for MCP-4-based diagnostics or therapeutics

Product Science Overview

Induction and Expression

CCL13 is induced by inflammatory proteins such as interleukin-1 (IL-1) and tumor necrosis factor-alpha (TNF-alpha) . The gene encoding CCL13 is located on human chromosome 17, within a large cluster of other CC chemokines . This clustering suggests a coordinated regulation and function of these chemokines in immune responses.

Receptors and Signaling

CCL13 functions by binding to three different G protein-coupled receptors: CCR2, CCR3, and CCR5 . These receptors are expressed on the surface of target cells and mediate the chemotactic response, leading to the directed movement of immune cells towards the site of inflammation or infection . This signaling is particularly important in the context of allergic responses and other inflammatory conditions.

Biological Activity

The biological activity of CCL13 includes the induction of chemotaxis in monocytes, eosinophils, T lymphocytes, and basophils . This chemotactic activity is essential for the recruitment of these immune cells to sites of inflammation, where they can exert their effector functions. The ability of CCL13 to attract a diverse range of immune cells highlights its importance in coordinating the immune response.

Recombinant Production

Human recombinant CCL13 is produced using an expression system in Escherichia coli (E. coli) . The recombinant protein is purified to a high degree of purity, typically greater than 98%, and is free from endotoxins . This high level of purity is essential for its use in research applications, where it is used to study the chemotactic properties and signaling mechanisms of CCL13.

Applications in Research

Recombinant CCL13 is widely used in various research applications, including Western blotting, enzyme-linked immunosorbent assay (ELISA), and functional assays . These applications help researchers to understand the role of CCL13 in immune responses and to explore its potential as a therapeutic target in inflammatory and allergic diseases.

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.