C5a Mouse

Complement Component C5a Mouse Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to C5a Mouse

C5a is a 74-residue glycoprotein fragment derived from the complement factor C5 during immune activation. In mouse models, recombinant C5a (rC5a) is widely used to study its role in inflammation, immune regulation, and disease pathogenesis. Structurally, mouse C5a (mC5a) shares 60% sequence identity with human C5a (hC5a) and 82% with rat C5a . Its primary functions include chemotaxis, endothelial activation, and modulation of leukocyte responses .

Production Methods

Mouse C5a is synthesized via two primary approaches:

Recombinant Expression

  • Source: E. coli with His-tagged constructs .

  • Sequence: MRGSHHHHHHGSDYDIPTTENLYFQGGSNLHLLRQKIEEQAAKYKHSVPKKCCYDGARVNFYETCEERVARVTIGPLCIAFNECCTIANKIRKESPHKPVQLGR .

  • Molecular Weight: ~12 kDa .

  • Purity: ≥95% by SDS-PAGE .

Chemical Synthesis

  • Advantages: Enables site-specific modifications (e.g., non-natural amino acids) and avoids microbial contaminants .

  • Validation: Confirmed via β-arrestin recruitment assays and neutrophil chemotaxis tests .

Research Applications and Findings

C5a Mouse has been pivotal in studying inflammatory diseases and receptor biology:

StudyModelKey Findings
Acute Liver Failure (ALF) D-GalN/LPS-challenged miceC5aR blockade reduces ALT levels, cytokines (TNF-α, IL-6), and mortality.
Meningococcal Sepsis N. meningitidis infectionC5aR1 knockout mice exhibit improved survival and reduced bacteremia.
Receptor Signaling CHO-hC5aR1 cellsSynthetic mC5a activates ERK1/2 signaling comparably to recombinant C5a.

Therapeutic Implications

  1. C5aR1 Inhibition:

    • PMX53 and PMX205 are small-molecule antagonists that block C5aR1 in mice, reducing inflammation in sepsis .

    • Note: PMX53 lacks activity in human neutrophils, highlighting species-specific receptor interactions .

  2. Synthetic C5a Tools:

    • Lanthanide-chelated synthetic mC5a enables high-throughput screening for C5aR1 ligands .

Product Specs

Introduction
Mouse Complement 5a (C5a) is a glycoprotein belonging to the anaphylatoxin family. These proteins are structurally and functionally similar. C5a, a 77-amino acid peptide, is generated when C5 convertase enzymatically cleaves the C5 α chain during the classical and alternative complement pathways (C4b2a3b, C3bBb3b). Characterized by four α helices and three intrachain disulfide bonds forming a triple loop structure, mouse C5a interacts with a G-protein coupled receptor (GPCR) known as C5aR/CD88. Functioning as a potent chemoattractant and anaphylatoxin, C5a acts on all leukocyte types and various other cells, including endothelial, smooth muscle, kidney, liver, and neural cells. In bronchial epithelial cells, mouse C5a stimulates the release of IL-8. Additionally, it triggers an oxidative burst in macrophages and neutrophils, leading to histamine release from basophils and mast cells. C5a's anaphylatoxin activity indirectly affects hepatocytes by interacting with nonparenchymal cells through prostanoid secretion.
Description
Recombinant Mouse C5a, produced in E. coli, is a single, non-glycosylated polypeptide chain comprising 77 amino acids. With a molecular weight of 9 kDa, Mouse C5a is purified using proprietary chromatographic techniques.
Physical Appearance
Sterile Filtered White lyophilized (freeze-dried) powder.
Formulation
Mouse C5a is lyophilized from a concentrated solution (1 mg/mL) in 20 mM PB, pH 7.5, and 350 mM NaCl.
Solubility

To reconstitute the lyophilized Mouse C5a, it is recommended to dissolve it in sterile 18 MΩ-cm H2O at a concentration of at least 100 µg/mL. The reconstituted solution can be further diluted in other aqueous solutions.

Stability
Lyophilized Mouse C5a remains stable at room temperature for 3 weeks; however, it should be stored desiccated below -18°C for long-term storage. After reconstitution, Mouse C5a should be stored at 4°C for 2-7 days. For future use, store below -18°C. Avoid repeated freeze-thaw cycles.
Purity
Purity greater than 95.0% as determined by:
(a) Reverse-phase high-performance liquid chromatography (RP-HPLC) analysis.
(b) Sodium dodecyl-sulfate polyacrylamide gel electrophoresis (SDS-PAGE) analysis.
Biological Activity
The ED50 of Recombinant Mouse C5a, determined by its ability to induce N-acetyl-β-D-glucosaminidase release from differentiated U937 human histiocytic lymphoma cells, was found to be 5-20 ng/mL.
Synonyms
Complement C5, Hemolytic complement, C5, Hc, He, C5a.
Source
Escherichia Coli.
Amino Acid Sequence
The sequence of the first five N-terminal amino acids was determined and was found to be Asn-Leu-His-Leu-Leu.

Q&A

What is mouse C5a and how does it function in the complement system?

Mouse C5a is a bioactive cleavage product released from plasma component C5 during complement activation. It functions as a potent pro-inflammatory agent involved in mediating various cellular immune responses . The mouse C5a protein consists of amino acids Asn679-Arg755, with structural studies revealing it forms a four-helix bundle motif . C5a exerts its biological effects primarily through binding to the C5a receptor (C5aR), triggering downstream signaling cascades that lead to inflammatory responses. The cleavage of C5 to generate C5a serves as a reliable indicator of complement activation in both in vivo and in vitro systems .

The significance of C5a in immune regulation is evidenced by studies demonstrating that C5a overexpression can accelerate pathological processes such as atherosclerosis in susceptible mouse models, promoting macrophage recruitment, foam cell formation, and inflammatory activation . Functionally, mouse C5a induces various cellular responses, including the release of N-acetyl-beta-D-glucosaminidase from differentiated human histiocytic lymphoma cells, with an ED50 of 5-20 ng/mL .

How do human and mouse C5a proteins differ structurally and functionally?

Significant structural and functional differences exist between human and mouse C5a proteins that researchers must consider when designing experiments or translating findings:

FeatureHuman C5aMouse C5a
StructureThree-helix bundle in C5a-desArg formFour-helix bundle in both C5a and C5a-desArg forms
C5a-desArg activitySignificantly reduced compared to C5aSimilar activity level as full C5a
N-terminal regionExtended N-terminal helix in C5a-desArgForms fourth helical motif in four-helix bundle

Crystal structure analysis reveals that while human C5a-A8 (a variant) forms a three-helix bundle with an extended N-terminal helix (similar to human C5a-desArg), both mouse C5a and C5a-desArg fold into a four-helix bundle motif . This architectural difference significantly impacts receptor activation patterns across species.

A particularly important functional distinction is that murine C5a-desArg, unlike its human counterpart, maintains the same level of activation on its cognate receptor as intact murine C5a . This key difference must be considered when designing mouse model experiments intended to have translational relevance for human conditions.

Which mouse strains are appropriate for C5a-related research?

The selection of mouse strains is critical for C5a research, as genetic C5 deficiency exists naturally in several inbred strains:

Mouse StrainC5 StatusSusceptibility to C5a-Mediated Pathologies
C57BL/6C5 sufficientSusceptible to PbA-induced cerebral malaria
129sv/JC5 sufficientSusceptible to PbA-induced cerebral malaria
129P3/JC5 sufficientSusceptible to PbA-induced cerebral malaria
A/JC5 deficientResistant to PbA-induced cerebral malaria
DBA/2JC5 deficientResistant to PbA-induced cerebral malaria
AKR/JC5 deficientResistant to PbA-induced cerebral malaria
B10.D2/nSnJC5 sufficientSusceptible to PbA-induced cerebral malaria
B10.D2/oSnJC5 deficientResistant to PbA-induced cerebral malaria

C5-deficient strains (A/J, DBA/2J, AKR/J) possess a known frame-shift mutation in the C5 gene and do not express functional C5, making them naturally resistant to C5a-mediated pathologies such as cerebral malaria . The existence of congenic strains like B10.D2/nSnJ (C5-sufficient) and B10.D2/oSnJ (C5-deficient) on the same genetic background (C57BL/10) provides valuable experimental tools to isolate the effects of C5/C5a .

Researchers should carefully consider strain selection based on their experimental questions, as the C5 status fundamentally affects disease susceptibility and inflammatory responses.

How does C5a contribute to atherosclerosis development in mouse models?

C5a accelerates atherosclerosis development in ApoE-/- mice through multiple mechanisms:

  • Enhanced macrophage infiltration: C5a promotes macrophage chemotaxis to atherosclerotic regions in a C5a receptor-dependent manner, as demonstrated in trans-well assays .

  • Foam cell formation promotion: C5a overexpression increases lipid deposition and foam cell formation in developing lesions .

  • Inflammatory cytokine upregulation: C5a leads to elevated serum levels of pro-inflammatory cytokines, including interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) .

  • Collagen content reduction: Atherosclerotic plaques in C5a-overexpressing mice show decreased collagen content, potentially affecting plaque stability .

The proatherogenic effects of C5a are mediated specifically through the C5a receptor, as demonstrated by experiments where C5a receptor antagonists significantly blocked lesion development . Researchers investigating atherosclerosis progression can use adenoviral vectors expressing mouse C5a (Ad-C5a) to model increased C5a activity in ApoE-/- mice on high-fat diets, resulting in more extensive lesions compared to control adenovirus treatments .

What approaches are effective for C5a or C5aR blockade in mouse models?

Several strategies have proven effective for blocking C5a or its receptor in mouse models:

Blocking ApproachImplementation MethodReported Efficacy
Anti-C5aR antibodiesAdministration early in infection (2h before and 30h after)Significant protection from cerebral malaria in B10.D2/nSnJ mice
Anti-C5a antibodiesSimilar early administration protocolSignificant protection from cerebral malaria
Genetic C5 deficiencyUse of naturally C5-deficient strains or congenic transfer of C5-defective alleleComplete resistance to cerebral malaria

In cerebral malaria models, treatment with anti-C5aR antibodies conferred significant protection to C5-sufficient B10.D2/nSnJ mice compared to control serum-treated animals (p=0.0022) . Similar results were obtained using anti-C5a serum (p=0.0019) .

For genetic approaches, transferring the C5-defective allele from A/J (CM resistant) onto a C57BL/6 (CM-susceptible) background in congenic strains increased resistance to cerebral malaria; conversely, transferring the C5-sufficient allele from C57BL/6 onto an A/J background recapitulated cerebral malaria susceptibility .

These findings demonstrate that both pharmacological and genetic approaches to C5a/C5aR blockade can be highly effective, with the appropriate strategy depending on the specific research question and experimental model.

How can researchers distinguish between effects mediated by C5a versus other complement components?

Distinguishing between C5a-mediated effects and those caused by other complement components requires carefully designed experimental approaches:

  • Use of recombinant C5a protein: Apply purified recombinant mouse C5a (Asn679-Arg755) to directly assess C5a-specific effects independent of other complement activation products .

  • Specific C5a receptor antagonists: Employ C5aR antagonists to block only the C5a signaling pathway while leaving other complement functions intact .

  • Comparative studies with C5 deficiency and C5a blockade: Compare outcomes between C5-deficient mice (lacking all C5 functions) and C5-sufficient mice treated with anti-C5a or anti-C5aR antibodies (blocking only the C5a pathway) .

  • C5a versus C5b-9 differentiation: To distinguish C5a effects from those of the membrane attack complex (C5b-9), use specific anti-C5a antibodies while monitoring C5b-9 formation through immunohistochemistry or soluble terminal complement complex measurements .

In studies of cerebral malaria, researchers effectively distinguished the role of C5a from C5b-9 by demonstrating that antibody blockade of either C5a or C5aR protected susceptible mice from cerebral malaria, providing direct evidence that C5a is a mediator rather than merely a consequence of infection .

What are the optimal handling and storage conditions for recombinant mouse C5a?

Proper handling and storage of recombinant mouse C5a is crucial for maintaining biological activity:

FormReconstitutionStorage Recommendations
With carrier protein (BSA)Reconstitute at 10 μg/mL in sterile PBS containing at least 0.1% human or bovine serum albuminStore in a manual defrost freezer; avoid repeated freeze-thaw cycles
Carrier-freeReconstitute at 100 μg/mL in sterile PBSStore in a manual defrost freezer; avoid repeated freeze-thaw cycles

Commercially available recombinant mouse C5a is typically provided as a lyophilized product from a 0.2 μm filtered solution in PBS, with or without bovine serum albumin (BSA) as a carrier protein . The carrier protein enhances stability, increases shelf-life, and allows storage at more dilute concentrations .

After reconstitution, it is advisable to aliquot the protein into single-use portions to prevent activity loss from multiple freeze-thaw cycles . For cell or tissue culture applications or as ELISA standards, the version with carrier protein is generally recommended, while carrier-free protein is preferred for applications where BSA might interfere .

What methods are most effective for detecting C5a in mouse samples?

Several methods can be employed for detecting C5a in mouse samples, each with specific advantages:

  • Enzyme-linked immunosorbent assay (ELISA)

    • Provides quantitative measurement of C5a levels

    • Can detect early changes in C5a concentration during disease progression

    • Example: B10.D2/nSnJ mice displayed significantly higher levels of circulating C5a as early as day 1 after Plasmodium berghei ANKA infection

  • Western blotting

    • Allows visualization of C5a protein with specificity

    • Can distinguish between intact C5 and C5a cleavage product

    • Useful for confirming the presence of recombinant C5a proteins

  • Functional bioassays

    • Measures biological activity rather than just protein presence

    • Example: N-acetyl-beta-D-glucosaminidase release from differentiated U937 human histiocytic lymphoma cells in response to mouse C5a (ED50 = 5-20 ng/mL)

  • Immunohistochemistry

    • Detects C5a deposition in tissues

    • Particularly useful for studying localized C5a effects in disease models

    • Has been used to demonstrate enhanced macrophage infiltration in atherosclerotic regions with C5a overexpression

The selection of detection method should align with research objectives: use ELISA for precise quantification, functional assays for activity confirmation, and immunohistochemistry for tissue localization studies.

How should researchers design C5a-focused mouse experiments to ensure translational relevance?

To ensure translational relevance in C5a mouse experiments, researchers should consider several important design elements:

How should researchers interpret conflicting C5a data across different mouse strains?

When encountering conflicting C5a data across mouse strains, researchers should consider several factors:

  • Genetic background beyond C5 status: Different mouse strains may have additional genetic variations affecting inflammatory responses, complement regulation, or disease susceptibility independently of C5. Studies with recombinant congenic mice demonstrate that genetic background contributes significantly to outcomes .

  • C5a receptor expression and function: Variation in C5aR expression levels or signaling efficiency between strains could affect responses to equivalent C5a concentrations.

  • Disease model specificity: The relevance of C5a may vary by disease model. For example, while C5 deficiency is protective in cerebral malaria , the same deficiency might have different effects in other inflammatory or infectious models.

  • Timing of C5a production and measurement: Early C5a production may be particularly significant in some disease models. In cerebral malaria, C5-sufficient mice display elevated C5a levels as early as day 1 post-infection .

  • Compensatory mechanisms: Long-term C5 deficiency in certain strains might lead to compensatory upregulation of other inflammatory pathways.

For rigorous interpretation, researchers should:

  • Use closely related strains differing only in C5 status (e.g., B10.D2/nSnJ vs. B10.D2/oSnJ)

  • Validate genetic findings with pharmacological interventions

  • Consider both genetic and environmental variables

  • Assess time-course data rather than single timepoints

What quantitative approaches are most appropriate for analyzing C5a effects in mouse models?

Several quantitative approaches are valuable for analyzing C5a effects in mouse models:

  • Survival analysis:

    • Kaplan-Meier survival curves with log-rank tests to compare survival between C5-sufficient and C5-deficient strains or between treatment groups

    • Example: C5-deficient B10.D2/oSnJ mice showed significantly improved survival compared to C5-sufficient B10.D2/nSnJ mice in cerebral malaria (p<0.0001, χ²=12.274)

  • Time-course analysis of C5a levels:

    • Serial measurements of C5a concentrations throughout disease progression

    • Statistical comparison of C5a levels between susceptible and resistant strains at multiple timepoints

    • Example: B10.D2/nSnJ mice displayed significantly higher C5a levels as early as day 1 after Plasmodium berghei ANKA infection

  • Quantitative assessment of histopathological changes:

    • Measurement of lesion size, inflammatory cell infiltration, and tissue damage

    • Example: Quantification of atherosclerotic lesion extent in Ad-C5a versus control adenovirus-treated ApoE-/- mice

  • Multivariate analysis:

    • Principal component analysis or multiple regression to assess relationships between C5a levels, inflammatory markers, and disease outcomes

    • Particularly valuable when analyzing complex relationships between complement activation and downstream effectors

  • Dose-response relationships:

    • EC50/ED50 determination for C5a-induced cellular responses

    • Example: ED50 of 5-20 ng/mL for recombinant mouse C5a in inducing N-acetyl-beta-D-glucosaminidase release from differentiated U937 cells

What emerging approaches might enhance C5a research in mouse models?

Several emerging approaches show promise for advancing C5a research in mouse models:

  • CRISPR/Cas9 gene editing: Creating precise modifications to C5, C5aR, or regulatory elements rather than relying on naturally occurring C5-deficient strains. This allows for more subtle manipulations than complete C5 deficiency.

  • Cell-specific C5a/C5aR targeting: Developing mouse models with cell-type specific deletion or overexpression of C5aR to distinguish the contribution of different cell populations to C5a-mediated pathology.

  • Humanized mouse models: Engineering mice to express human C5a and/or C5aR to better model human-specific aspects of C5a biology and improve translational relevance .

  • Systems biology approaches: Integrating C5a signaling data with broader -omics datasets to understand complement activation in the context of global immune responses.

  • Real-time in vivo imaging: Developing techniques to visualize C5a activity in living animals, potentially using reporter systems linked to C5a-responsive elements.

These approaches could help address current limitations in understanding the complex roles of C5a in various disease contexts and potentially identify novel therapeutic targets in the C5a pathway.

How might researchers better translate findings from C5a mouse models to human disease?

Improving translational relevance of C5a mouse studies requires several strategic approaches:

  • Acknowledge structural and functional species differences: Account for the distinct structural properties of human and mouse C5a, particularly the differential activity of C5a-desArg between species .

  • Validate findings across multiple mouse strains: Confirm results in different genetic backgrounds to ensure observations are not strain-specific artifacts.

  • Correlate with human samples: Whenever possible, validate mouse model findings with parallel analyses of human samples from relevant patient populations.

  • Consider pathway conservation: Focus on conserved signaling pathways downstream of C5aR activation rather than absolute C5a levels or specific structural interactions.

  • Develop humanized models: Use mice expressing human C5a and/or C5aR to better recapitulate human-specific aspects of complement biology.

  • Pharmacological validation: Test C5a/C5aR-targeting therapeutics that are being developed for human use in mouse models to establish predictive validity.

By implementing these approaches, researchers can enhance the translational potential of C5a mouse studies and improve their relevance to human disease mechanisms and therapeutic development.

Product Science Overview

Structure and Generation

Complement Component C5a is a 77 amino acid peptide generated from the cleavage of the C5 alpha-chain by the enzyme C5a convertase . This cleavage occurs in both the classical and alternative complement pathways, resulting in the formation of C5a and C5b. The recombinant version of mouse C5a is typically produced in E. coli and purified to high levels of purity (>97%) for research purposes .

Biological Functions

C5a is known as an anaphylatoxin, a type of molecule that can induce rapid degranulation of mast cells and basophils, leading to the release of histamine and other inflammatory mediators . It is a potent chemoattractant, recruiting immune cells such as neutrophils, eosinophils, and macrophages to sites of infection or injury . Additionally, C5a enhances the phagocytic activity of these cells, aiding in the clearance of pathogens .

Mechanism of Action

C5a exerts its effects by binding to its specific receptor, C5aR (CD88), which is expressed on the surface of various immune cells . This binding triggers a cascade of intracellular signaling events that result in the activation of these cells and the release of inflammatory cytokines and chemokines . The interaction between C5a and C5aR is crucial for the amplification of the inflammatory response and the recruitment of immune cells to the site of infection .

Applications in Research

Recombinant mouse C5a is widely used in research to study the complement system and its role in various diseases. It is used in in vitro assays to investigate the effects of C5a on immune cell activation, chemotaxis, and cytokine production . Additionally, it is employed in animal models to study the role of C5a in inflammatory diseases, such as sepsis, arthritis, and autoimmune disorders .

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.