C5a binds to two principal receptors:
Receptor | Function |
---|---|
C5aR1 (CD88) | G-protein-coupled receptor (GPCR) mediating chemotaxis, cytokine release, and β-arrestin recruitment |
C5L2 (GPR77) | β-arrestin-coupled receptor with immunomodulatory roles |
G-protein Coupling: C5aR1 activates G<sub>i</sub>, G<sub>q</sub>, and G<sub>16</sub> pathways, triggering calcium influx, MAPK activation, and cytokine production .
β-Arrestin Recruitment: Mediates receptor internalization and terminates signaling .
Cryo-EM Insights: C5a binds C5aR1 via a three-site interaction involving the N-terminal domain, extracellular loops (ECL2), and transmembrane helices .
C5a modulates both innate and adaptive immunity:
Chemotaxis: Potent recruitment of neutrophils, eosinophils, and monocytes to infection sites .
Inflammation: Increases vascular permeability, adhesion molecule expression (e.g., ICAM-1), and ROS production .
Tissue Regeneration: Stimulates hepatocyte proliferation and liver repair .
C5a exacerbates sepsis by:
Neutrophil Dysfunction: Suppressing phagocytosis and inducing apoptosis via caspase activation .
Cytokine Storm: Synergizing with LPS to amplify TNF-α, IL-6, and IL-8 production .
Thymocyte Apoptosis: Contributing to immune exhaustion in septic shock .
Therapeutic Target: Neutralizing C5a improves survival in primate sepsis models .
C5a enhances β-amyloid-induced neurotoxicity via:
Microglial Activation: Increasing TNF-α, IL-1β, and IL-6 release through JAK/STAT3 signaling .
C5aR1 Antagonism: PMX205 reduces neuroinflammation and protects neurons in in vitro models .
Elevated C5a correlates with poor prognosis in metastatic renal cell carcinoma (mRCC), linking to therapeutic resistance and tumor progression .
C5a induces neutrophil-derived MV shedding via C5aR1-Arf6 signaling, amplifying inflammation:
Mechanism | Outcome |
---|---|
C5aR1 Activation | Triggers Arf6-dependent MV release, reducing surface C5aR1 |
Pro-Inflammatory MVs | Activate neutrophils, increase ROS, and induce IL-6 in whole blood models |
C5a exhibits context-dependent effects:
Complement C5a (C5a) is a protein involved in the complement system, a crucial part of the body's immune response. C5a is created when a specific protein, C5, is split into two parts: C5a and C5b. C5a acts like a signal flare, attracting immune cells to the site of infection or damage. This process is called chemotaxis. Meanwhile, C5b helps form a complex that can punch holes in the membranes of invading cells.
This product contains human Complement C5a, a protein with a molecular weight of 10.4 kDa. It is produced using human plasma.
The product is a clear, sterile solution that has been passed through a filter to remove any bacteria.
The C5a protein is supplied in a solution containing 120 mM NaCl (sodium chloride) and 10mM HEPES (a buffering agent) at a pH of 7.2.
To ensure product stability, store C5a Human at 4°C and use within 2-4 weeks. For longer-term storage, freeze at or below -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for extended storage. Avoid repeatedly freezing and thawing the product.
This product has a purity greater than 95%, as determined by SDS-PAGE, a technique used to separate and analyze proteins.
The plasma used to produce this product has been rigorously tested and confirmed negative for a range of viruses, including HIV-1, HIV-2, HCV, HTLV-I & II, STS, and HBSAG, ensuring product safety.
Complement Component 5, C3 and PZP-Like Alpha-2-Macroglobulin Domain-Containing Protein 4, C5a Anaphylatoxin, Prepro-C5, CPAMD4, Anaphylatoxin C5a Analog, ECLZB, C5A, C5D, C5b, C5.
Human Plasma.
What is C5a protein and what are its primary functions in the immune system?
C5a is a 74-amino acid protein composed of four α-helices in an antiparallel configuration with bridging disulfide bonds. It is formed through proteolytic cleavage of complement component C5, typically by C5 convertase in the plasma . The molecular weight of glycosylated C5a is approximately 12-14.5 kDa, while bacterially expressed recombinant human C5a has a lower molecular weight of 8-9 kDa due to the absence of glycosylation .
As a powerful phlogistic (inflammatory) factor, C5a functions as a chemoattractant for neutrophils and other innate immune cells, guiding them to sites of infection or injury . It exerts its biological effects by binding to two distinct seven-transmembrane receptors: C5aR1 (CD88) and C5aR2 . Upon receptor activation, C5a triggers numerous inflammatory responses including:
Recruitment of innate immune cells to infection or injury sites
Secretion of pro-inflammatory cytokines and chemokines
Enhancement of phagocytosis
Activation of respiratory burst in neutrophils
Upregulation of adhesion molecules
While C5a provides rapid protection during infectious challenges, persistent or dysregulated activation can contribute to inflammatory disorders, autoimmune diseases, and conditions like COVID-19-associated coagulopathy .
How is C5a generated in physiological conditions?
Phagocytic cells can directly generate C5a from C5 through cell-specific proteases. Studies have demonstrated that when human blood neutrophils or rat alveolar macrophages (AMs) are activated with phorbol 12-myristate 13-acetate (PMA) or lipopolysaccharide (LPS), they can cleave C5 to produce biologically active C5a . This alternative pathway has several notable characteristics:
Activated neutrophils cause extensive cleavage of C5, while activated macrophages exhibit more selective proteolytic activity
The process is specific to phagocytic cells, as peripheral blood mononuclear cells and alveolar epithelial cells fail to cleave C5 even when stimulated
Serine protease inhibitors (soybean trypsin inhibitor and secretory leukocyte protease inhibitor) block C5a generation by activated macrophages
The process requires divalent cations, as EDTA prevents C5a generation
This mechanism does not require new protein synthesis, as protein synthesis inhibitors like cycloheximide or actinomycin D do not block C5a generation
This phagocyte-mediated generation of C5a provides a rapid mechanism for amplifying inflammatory responses at sites of infection, independent of the systemic complement cascade.
What experimental methods are commonly used to detect and measure C5a activity?
Several complementary experimental approaches are used to detect, quantify, and assess the biological activity of C5a:
Western Blotting:
Western blot analysis with specific antibodies against human or rat C5a can detect C5a generated by phagocytic cells or present in biological samples . Glycosylated C5a typically aligns with the 14.3-kDa marker, while recombinant human C5a shows a main band between 6.5-14.3 kDa and a less intense band between 14.3-21.5 kDa .
Functional Assays:
Various functional assays can measure C5a biological activity:
Neutrophil chemotaxis assays assess the ability of C5a to induce directional migration of neutrophils in vitro
pERK1/2 signaling assays in C5aR1-expressing cells (e.g., CHO-hC5aR1 cells, primary human macrophages) measure receptor activation
β-arrestin recruitment assays in C5aR2-transfected HEK293 cells confirm C5aR2 activation
In vivo neutrophil mobilization assays evaluate the biological activity of synthetic or recombinant C5a
Calcium flux assays detect immediate receptor activation in responsive cells
Advanced Imaging Techniques:
High-resolution lattice light-sheet microscopy on live cells can visualize C5a-induced C5aR1 internalization and colocalization with trafficking proteins like Rab5a-tdTomato .
Binding Assays:
Biophysical techniques can assess C5a-receptor interactions:
Circular dichroism (CD) spectroscopy measures conformational changes upon binding and can be used to estimate binding affinities
Fluorescence titration studies complement CD data for affinity determination
Surface plasmon resonance provides real-time binding kinetics
These methodologies collectively enable comprehensive characterization of C5a in various experimental contexts, from basic biochemical studies to complex disease models.
How do C5aR1 and C5aR2 differ in their signaling mechanisms and functions?
C5a exerts its biological effects through two distinct seven-transmembrane receptors that differ significantly in their signaling mechanisms and functional outcomes:
C5aR1 (CD88):
Couples to heterotrimeric G proteins, primarily Gαi, leading to inhibition of adenylyl cyclase and activation of phospholipase C
Activates multiple signaling cascades including:
MAPK pathways (particularly ERK1/2 phosphorylation)
Phosphatidylinositol 3-kinase (PI3K)/Akt signaling
Calcium mobilization
NF-κB activation
Mediates most pro-inflammatory effects of C5a
Undergoes internalization upon C5a binding, with trafficking regulated by small GTPases like Rab5a
C5aR2 (formerly C5L2):
Initially considered a decoy receptor but now recognized to have signaling capabilities
Signals primarily through β-arrestin recruitment rather than G protein coupling
Does not couple to G proteins due to replacement of key residues in the DRY motif
May have both pro- and anti-inflammatory functions depending on context
Can modulate C5aR1 signaling through heteromer formation
Less well-characterized than C5aR1
Key Differences:
C5aR1 signaling leads to rapid cellular responses (chemotaxis, degranulation), while C5aR2 effects may be more regulatory
C5aR1 internalizes rapidly upon ligand binding, while C5aR2 may have different trafficking kinetics
C5aR1 is the primary target for drug development, though dual targeting approaches are emerging
C5a binding to C5aR1 involves a complex "two-site" binding model with distinct interaction domains
Understanding these differences is crucial for developing targeted therapeutics and interpreting experimental results in C5a research.
What is the evidence for the "two-site" binding model of C5a-C5aR1 interaction and what are its implications for drug development?
The "two-site" binding model describes the complex interaction between C5a and its primary receptor C5aR1. This model has been supported by various biophysical studies and has significant implications for drug design:
Binding Model Components:
The two-site binding interaction involves:
Site 1: The interaction between the core of C5a and the extracellular loops/transmembrane domains of C5aR1
Site 2: The interaction between the C-terminal region of C5a and the N-terminal domain (NT) of C5aR1
Experimental Evidence:
Biophysical studies using synthetic peptides corresponding to receptor domains provide strong evidence for this model:
Circular dichroism (CD) and fluorescence titration studies with NT-peptides (SR3 and SR5) show binding affinities to C5a in the range of 105-193 nM
Interestingly, the SR4 peptide containing tyrosines but lacking multiple aspartic acids failed to demonstrate quantifiable binding to C5a
These findings suggest that aspartic acid residues in the N-terminal domain may be more crucial for C5a binding than previously thought
Structural Changes During Binding:
The binding interaction induces significant conformational changes:
CD studies show alterations in the secondary structure upon binding
These conformational changes likely contribute to receptor activation and signal transduction
Molecular dynamics simulations provide further insights into these dynamic interactions
Implications for Drug Development:
Understanding this binding model creates opportunities for targeted therapeutic design:
Small molecule antagonists can be designed to disrupt specific binding interfaces
Peptide fragments from the receptor can serve as templates for antagonist development
Modification of the C-terminal region of C5a can create antagonists that bind but don't activate
Structure-based design can target specific binding pockets within the receptor
This two-site binding model provides a framework for rational drug design and helps explain the complex pharmacology of C5aR1 ligands, including why some compounds act as agonists while others function as antagonists.
How does Rab5a regulate C5a receptor trafficking and signaling in immune cells?
Rab5a, a small GTPase of the Rab family, plays a crucial role in regulating C5a receptor trafficking and signaling, particularly in macrophages. This regulatory mechanism has significant implications for inflammation and immune responses:
Trafficking Mechanism:
Upon C5a binding to C5aR1, a sophisticated trafficking process occurs:
C5aR1 undergoes internalization from the cell surface
Rab5a is recruited to the activated receptor complex
High-resolution lattice light-sheet microscopy has demonstrated that C5aR1-GFP colocalizes with Rab5a-tdTomato in live cells following C5a stimulation
This colocalization does not occur with dominant negative mutant Rab5a-S34N-tdTomato, indicating that functional Rab5a is required
Signaling Pathway:
Rab5a-mediated trafficking influences downstream signaling events through a specific cascade:
C5a activation of C5aR1 recruits β-arrestin2 via Rab5a trafficking
This leads to activation of phosphatidylinositol 3-kinase (PI3K)/Akt signaling
The activated signaling pathway ultimately results in:
Functional Significance:
This regulatory mechanism has important functional consequences:
Controls the magnitude and duration of C5a-induced inflammatory responses
Provides a potential target for modulating C5a-mediated inflammation
Links complement activation with intracellular vesicular trafficking pathways
May contribute to dysregulated inflammation in pathological conditions
This relationship between Rab5a and C5aR1 represents an important intersection between the complement system and cellular trafficking machinery, offering potential new targets for therapeutic intervention in inflammatory diseases.
What is the evidence linking C5a to disease pathogenesis and what therapeutic approaches target the C5a pathway?
C5a has been implicated in numerous inflammatory and autoimmune diseases. Strong evidence exists for its role in conditions like periodontitis and COVID-19:
Periodontitis:
Recent genetic evidence supports C5a's role in periodontitis pathogenesis:
A drug target instrumental variable (IV) approach using 26 independent 'cis' single nucleotide polymorphisms associated with plasma C5 levels revealed that genetically proxied inhibition of C5 reduced the risk of periodontitis (Odds ratio 0.89 per 1 standard deviation reduction in C5; 95% confidence Interval 0.80–0.98, p-value=0.022)
Secondary analysis suggested involvement of IL-17 within the potential causal pathway
These findings prioritize C5 inhibitors as potential adjunctive therapeutic strategies for periodontitis
COVID-19:
Emerging data implicate C5a in COVID-19-associated coagulopathy:
C5a/C5aR signaling participates in the cascade of events involved in COVID-19-associated hypercoagulable states
C5a initially provides rapid protection during infectious challenge but can transition to a pathological role
Targeting C5a/C5aRs signaling represents a potential novel therapeutic approach for COVID-19 patients with coagulation abnormalities
Therapeutic Approaches:
Several strategies target the C5a pathway in disease:
C5 Inhibition:
C5a-Specific Neutralization:
Targets C5a specifically without affecting C5b formation
Examples include IFX-1 (vilobelimab, anti-C5a monoclonal antibody)
More selective than C5 inhibition
C5aR1 Antagonists:
Block C5a binding to C5aR1
Examples include avacopan (small molecule antagonist)
Allow normal function of other complement components
Dual Receptor Antagonists:
Target both C5aR1 and C5aR2
Potentially more comprehensive blockade of C5a effects
Upstream Complement Inhibitors:
Target earlier steps in complement activation
May reduce C5a generation indirectly
These therapeutic approaches are in various stages of clinical development for conditions including ANCA-associated vasculitis, COVID-19, psoriasis, and periodontitis.
How do phagocytic cells specifically generate C5a and what mechanisms regulate this process?
The generation of C5a by phagocytic cells represents an alternative pathway for complement activation that operates independently of the traditional complement cascade. This process involves cell-specific proteases and regulatory mechanisms:
Cell Specificity:
Only specific phagocytic cells demonstrate the ability to generate C5a from C5:
Human blood neutrophils and rat alveolar macrophages efficiently generate C5a when activated
Peripheral blood mononuclear cells and rat alveolar epithelial cells fail to cleave C5 even when stimulated
This specificity suggests specialized enzymatic machinery in neutrophils and macrophages
Activation Requirements:
The generation of C5a by phagocytes requires cellular activation:
Phorbol 12-myristate 13-acetate (PMA), a protein kinase C activator, effectively stimulates C5a generation by both neutrophils and macrophages
Lipopolysaccharide (LPS) can also activate rat alveolar macrophages to generate C5a
The process is independent of new protein synthesis, as treatment with cycloheximide or actinomycin D does not inhibit C5a generation
Enzymatic Mechanisms:
The proteases involved in C5a generation by phagocytes have been partially characterized:
Serine proteases play a crucial role, as demonstrated by inhibition with soybean trypsin inhibitor (SBTI) and secretory leukocyte protease inhibitor (SLPI)
The process requires divalent cations, as EDTA prevents C5a generation
Activated neutrophils cause extensive cleavage of C5, whereas activated macrophages demonstrate much more selective proteolytic activity
Validation of Generated C5a:
The C5a generated by activated phagocytes has been confirmed to be biologically active:
Western blot analysis shows the generated C5a aligns with C5a immunoprecipitated from activated human serum
The product is chemotactically active for neutrophils
This cell-specific generation of C5a provides a rapid mechanism for amplifying inflammatory responses at sites of infection, creating a direct link between phagocyte activation and complement-mediated inflammation.
What approaches can be used to synthesize functional C5a for research applications?
Synthetic C5a provides several advantages over recombinant or purified C5a for research applications. Key approaches and considerations include:
Chemical Synthesis Methods:
Chemical synthesis offers a precise approach to producing C5a:
Solid-phase peptide synthesis can efficiently produce both human (hC5a) and mouse C5a (mC5a) without requiring ligation chemistry
Post-synthesis folding procedures are critical for establishing the correct disulfide bonding and tertiary structure
The synthesis can incorporate the full 74-amino acid sequence with appropriate modifications
Advantages of Synthetic C5a:
Synthetic C5a offers several benefits over other sources:
Ability to introduce non-natural amino acids and site-specific modifications
Lower probability of contamination with microbial molecules or other endogenous proteins
Consistent batch-to-batch quality and purity
Flexibility to create species-specific variants (human, mouse, etc.)
Functionalization Options:
Synthetic C5a can be functionalized for specific research applications:
Introduction of lanthanide chelating cages to facilitate screening for ligand binding to C5aR1
Incorporation of fluorescent labels for imaging studies
Addition of biotinylation sites for pull-down experiments
Creation of photocrosslinkable derivatives for binding site identification
Validation Methods:
Synthetic C5a must be rigorously validated to ensure proper structure and function:
Comparison of pERK1/2 signaling in CHO-hC5aR1 cells and primary human macrophages (for hC5a) and in RAW264.7 cells (for mC5a)
Confirmation of C5aR2 activation by measuring β-arrestin recruitment in C5aR2-transfected HEK293 cells
Neutrophil chemotaxis assays in vitro
Structural analysis using circular dichroism spectroscopy
Feature | Synthetic C5a | Recombinant C5a | Purified Endogenous C5a |
---|---|---|---|
Purity | Very high | High with potential contaminants | Variable |
Batch consistency | High | Moderate | Low |
Modification potential | Extensive | Limited | None |
Glycosylation | Absent unless specifically added | Depends on expression system | Present |
Molecular weight (hC5a) | ~8-9 kDa (non-glycosylated) | Variable | ~12-14.5 kDa |
Biological activity | Equivalent to recombinant | Standard reference | Standard reference |
Application flexibility | High | Moderate | Low |
Synthetic C5a represents a valuable alternative to recombinant or purified C5a, particularly for specialized applications requiring modifications or high purity.
What experimental models are most appropriate for studying C5a function in vitro and in vivo?
Selecting appropriate experimental models is crucial for studying C5a biology. Various in vitro and in vivo models offer complementary insights:
In Vitro Cellular Models:
Receptor-Transfected Cell Lines:
CHO-hC5aR1 cells: Chinese hamster ovary cells stably expressing human C5aR1; useful for signaling studies and drug screening
HEK293-C5aR2 cells: Human embryonic kidney cells expressing C5aR2; appropriate for β-arrestin recruitment studies
Primary Cells:
Human neutrophils: Excellent model for chemotaxis, respiratory burst, and calcium flux studies
Human monocyte-derived macrophages (HMDMs): Appropriate for studying C5a-induced chemotaxis and chemokine secretion
Rat alveolar macrophages: Used to study C5a generation and receptor signaling
Macrophage Cell Lines:
RAW264.7 cells: Murine macrophage cell line; useful for studying mouse C5a effects
THP-1 cells: Human monocytic cell line that can be differentiated into macrophage-like cells
Functional Assays:
Chemotaxis Assays:
These assays measure the directional migration of cells toward C5a gradients:
Transwell migration assays using neutrophils or macrophages
Real-time migration tracking with video microscopy
3D collagen matrix migration assays for more physiological conditions
Signaling Assays:
ERK1/2 phosphorylation in C5aR1-expressing cells provides a quantitative measure of receptor activation
Calcium flux assays detect immediate receptor activation
β-arrestin recruitment assays specifically assess C5aR2 activation
Advanced Imaging:
High-resolution lattice light-sheet microscopy can visualize C5a-induced receptor internalization and trafficking in live cells
Fluorescence resonance energy transfer (FRET) to monitor receptor-effector interactions
Confocal microscopy to track receptor localization
In Vivo Models:
Several animal models are valuable for studying C5a function:
C5a-induced neutrophil mobilization in mice provides a direct assessment of in vivo activity
Inflammatory disease models (arthritis, lung inflammation, sepsis)
Genetic models with C5aR1 or C5aR2 deficiency to elucidate receptor-specific effects
Humanized mouse models expressing human C5aRs for translational research
Disease-Specific Models:
Models relevant to specific C5a-associated diseases include:
Experimental periodontitis models, supported by genetic evidence linking C5a to periodontitis risk
Sepsis models where complement activation contributes to pathology
The choice of model should align with the specific research question, considering factors such as species differences in C5a receptors, physiological relevance, and technical feasibility.
What are the key considerations when designing experiments to study C5a-C5aR interactions?
Studying C5a-C5aR interactions requires careful experimental design. Key considerations include:
Receptor Expression Systems:
Consider endogenous vs. overexpression systems
Use of species-matched C5a and receptors is critical due to species specificity
Quantify receptor expression levels as they impact response magnitude
Consider potential receptor heterodimer formation in cells expressing both C5aR1 and C5aR2
Ligand Selection:
Choose appropriate C5a source (recombinant, synthetic, or purified)
Account for differences between glycosylated and non-glycosylated C5a forms
Consider using receptor-selective agonists to distinguish C5aR1 from C5aR2 responses
Include appropriate positive controls and reference standards
Binding Assays:
When studying the physical interaction between C5a and its receptors:
Circular dichroism (CD) spectroscopy can detect conformational changes upon binding and estimate binding affinities (Kd)
Fluorescence spectroscopy complements CD data for binding affinity determination
Account for potential artifacts from fluorescent labeling
Subtract background signals carefully (e.g., CD signals from free peptides in buffer)
Signaling Assays:
For measuring receptor activation:
Select timepoints relevant to the signaling pathway being studied
Include both early (calcium flux, ERK phosphorylation) and late (gene expression) readouts
Use pathway-specific inhibitors to confirm signaling mechanisms
Consider receptor desensitization effects with repeated or prolonged stimulation
Trafficking Studies:
When examining receptor internalization and trafficking:
High-resolution microscopy can visualize colocalization with trafficking proteins like Rab5a
Use dominant-negative mutants (e.g., Rab5a-S34N) as controls
Consider both constitutive and ligand-induced trafficking
Track receptors over appropriate time courses to capture dynamics
Data Analysis:
Use appropriate curve-fitting for dose-response relationships
Apply nonlinear regression to estimate binding parameters from titration data
Consider statistical power when designing experiments
Account for inter-assay variability with proper normalization
Controls and Validation:
Include specificity controls (receptor antagonists, blocking antibodies)
Use receptor knockout or knockdown systems to confirm specificity
Compare results across multiple assay formats
These considerations help ensure robust, reproducible results when studying the complex interactions between C5a and its receptors.
What therapeutic strategies targeting C5a are being investigated, and what methodological approaches support their development?
Research into C5a-targeted therapeutics has expanded significantly, with several methodological approaches supporting their development:
Target Selection Rationale:
Genetic and biological evidence guides target selection:
Genetic studies link C5 levels to disease risk in conditions like periodontitis (OR 0.89 per 1 SD reduction in C5)
Mechanistic studies identify C5a's role in pathological processes such as COVID-19-associated coagulopathy
Pathway analysis suggests involvement of downstream mediators like IL-17 that may provide additional therapeutic targets
Therapeutic Modalities:
C5 Inhibition:
C5a-Specific Neutralization:
Targets C5a selectively without affecting membrane attack complex formation
Includes monoclonal antibodies and aptamers against C5a
Potentially offers improved safety profile compared to complete C5 inhibition
Requires high specificity to avoid cross-reactivity with related anaphylatoxins
C5aR1 Antagonists:
Methodological Approaches:
Structural Biology:
Understanding of C5a-C5aR binding mechanisms informs drug design:
The "two-site" binding model provides a framework for rational antagonist design
Synthetic peptides corresponding to receptor domains (e.g., SR3, SR5) serve as templates
Biophysical techniques like CD spectroscopy and fluorescence titration characterize binding interactions
Medicinal Chemistry:
Structure-activity relationship studies optimize leads
Introduction of non-natural amino acids or modifications enhances stability
Development of orally available small molecule antagonists
Preclinical Models:
In vitro activity assessed in relevant cell systems (neutrophils, macrophages)
In vivo efficacy evaluated in disease-specific animal models
Synthetic human and mouse C5a provide valuable tools for efficacy studies
Biomarker Development:
Assays to measure target engagement (C5a levels, receptor occupancy)
Downstream markers like IL-17 may serve as pharmacodynamic indicators
Genetic markers could potentially identify patients most likely to benefit from therapy
Translational Considerations:
Development of companion diagnostics to identify appropriate patients
Assessment of safety, particularly regarding infection risk
Evaluation of potential for complement-independent effects
Consideration of local vs. systemic administration depending on disease
These methodological approaches collectively support the development of C5a-targeted therapeutics across multiple disease areas, with promising applications in conditions like periodontitis and COVID-19-associated coagulopathy .
Human C5a is a 74 amino acid peptide with a molecular weight of approximately 11 kDa . The structure of C5a consists of four helices connected by peptide loops, stabilized by three disulfide bonds. The N-terminus contains a short helix, but the primary agonist activity is located at the C-terminus .
C5a is known for its role as an anaphylatoxin and a chemotactic agent . It has several key functions:
The increased production of C5a is associated with various inflammatory diseases. Abnormal complement activation, including the generation of C5a, can lead to conditions such as paroxysmal nocturnal hemoglobinuria and atypical hemolytic uremic syndrome . Therapeutic interventions targeting C5a and its receptor (C5aR) are being explored to treat these and other inflammatory diseases .
Recombinant human C5a is produced using various expression systems, including bacterial, yeast, and mammalian cells. These recombinant proteins are used in research and therapeutic applications to study the functions of C5a and to develop potential treatments for diseases involving complement activation .