Fact-checked by Grok 2 weeks ago

Complement system

The complement system is a critical component of the , comprising a network of more than 40 soluble and membrane-bound proteins that work together to detect and eliminate pathogens, damaged cells, and immune complexes, while also bridging innate and adaptive immunity. It enhances antibody-mediated responses and promotes , , and direct cell through tightly regulated cascades. The system includes central components such as the recognition molecules C1q and mannose-binding lectin (MBL), along with alternative pathway components like factor B, and effector proteins like (the central molecule) and the terminal components –C9 that form the membrane attack complex (). Complement activation occurs via three primary biochemical pathways: the classical pathway, initiated by antigen-antibody complexes binding C1q or direct C1q recognition of surfaces; the , triggered by MBL or ficolins binding carbohydrate patterns on microbes; and the alternative pathway, spontaneously activated by low-level hydrolysis and amplified on foreign surfaces lacking host regulators. These pathways converge at the cleavage of into C3a and C3b, leading to downstream amplification and formation of , which initiates the terminal pathway. Key functions of the complement system encompass opsonization of pathogens and apoptotic cells via C3b deposition to facilitate , generation of anaphylatoxins C3a and C5a to recruit immune cells and induce , and assembly of the to lyse target cells by creating pores in their membranes. It also plays roles in immune , clearance of immune complexes, B-cell , and T-cell , thereby integrating innate defenses with adaptive responses. Dysregulation can lead to excessive or , contributing to diseases like systemic lupus erythematosus, , and age-related macular degeneration. To prevent host tissue damage, the complement system is tightly controlled by soluble regulators like and , and membrane-bound proteins such as (DAF/CD55) and membrane cofactor protein (MCP/), which inhibit convertase activity and promote degradation of activated components. This regulatory balance ensures effective elimination while maintaining , with approved therapeutics such as complement inhibitors and ongoing research in modulating the system for autoimmune, infectious, and inflammatory disorders.

Introduction

Definition and components

The complement system is a crucial part of the , consisting of more than 30 soluble proteins in the and membrane-bound proteins on cells that through a tightly regulated proteolytic cascade to detect and respond to pathogens. This network of proteins, amounting to about 3-5% of total protein, enables rapid defense by recognizing microbial surfaces and initiating effector mechanisms. Complement components are broadly classified by their primary roles in the . Recognition molecules initiate by to pathogen-associated patterns; examples include C1q, which recognizes antibody-opsonized surfaces, and mannose- lectin (MBL), which binds to motifs on microbes. proteases propagate the through sequential cleavage; key examples are C1r and C1s, which form part of the C1 complex, and factor B, which participates in pathway convertase formation. Anaphylatoxins are inflammatory mediators released upon cleavage of central components, such as C3a and C5a, which recruit immune cells and induce . Opsonins tag targets for , including C3b, which covalently attaches to surfaces, and its inactivated form iC3b. Finally, membrane attack complex (MAC) proteins assemble into a pore-forming structure; these comprise C5b joined by C6, C7, C8, and multiple C9 molecules to form C5b-9. The proteins of the complement system fall into three major families based on structural and functional . Serine proteases, such as C1r, C1s, factor B, and factor D, contain catalytic domains that drive proteolytic steps. The C3/C4/C5 family consists of large, multi-domain proteins with an internal bond that enables covalent attachment to targets upon ; C3 serves as the central hub, while C4 and C5 contribute to amplification and terminal effects. Regulators of complement (RCA), also known as complement control proteins, inhibit uncontrolled activity to protect host tissues; this family includes soluble factors like and membrane-bound proteins such as CR1 (CD35) and (DAF, CD55).

Overview of functions

The complement system plays a central role in innate immunity by directly pathogens through the formation of the (), a transmembrane composed of C5b-9 proteins that causes osmotic of target cells such as . It also mediates opsonization, primarily via the deposition of C3b fragments on surfaces, which marks them for enhanced recognition and engulfment by expressing complement receptors. Additionally, activation generates anaphylatoxins C3a and C5a, which bind to receptors on mast cells, , and other immune cells to promote , , and recruitment of neutrophils and monocytes to infection sites, thereby amplifying local inflammatory responses. Complement further contributes to the solubilization and clearance of immune complexes, preventing their harmful deposition in tissues and facilitating their removal by the . Beyond direct antimicrobial effects, the complement system acts as an amplifier that bridges innate and adaptive immunity, enhancing the efficiency of antibody-mediated responses through interactions with B cells and antigen-presenting cells. For instance, complement-opsonized antigens bind to complement receptors on B cells (such as CR2/CD21), lowering the threshold for B-cell activation and promoting formation, which leads to improved antibody affinity maturation and class switching. In homeostatic contexts, complement maintains tissue integrity by facilitating the non-inflammatory clearance of apoptotic cells, where C1q and iC3b opsonize dying cells for efficient without triggering excessive . It also supports B-cell tolerance by regulating self-reactive B cells; for example, iC3b binding to complement receptors on immature B cells induces anergy or , preventing .

History

Early discoveries

The complement system's discovery began in the late 19th century with observations of antibacterial activity in blood . In 1891, German bacteriologist Hans Buchner identified a heat-labile factor in normal that could lyse , naming it "alexin" (from the Greek word meaning "to ward off"). This marked the initial recognition of a non-specific defensive component in blood, distinct from cellular immunity. Building on this, Belgian immunologist , working at the in 1894–1895, conducted pivotal experiments showing that immune 's lytic effects on or foreign red blood cells required two distinct components: a heat-stable, specific "sensitizer" (later termed ) and a heat-labile, non-specific factor akin to Buchner's alexin. Bordet demonstrated that the heat-labile factor was universally present in fresh normal and could restore lytic activity when added to heated immune . In 1900, Bordet advanced this understanding through experiments on the complement-fixation reaction, revealing that antigen-antibody complexes could bind and "fix" the heat-labile factor, preventing it from participating in subsequent lysis of indicator cells. This observation distinguished the specific immune recognition from the amplifying role of complement and formed the basis for serological diagnostics. For these foundational contributions to immunity, including the elucidation of complement's role, Bordet received the 1919 Nobel Prize in Physiology or Medicine. Concurrent with Bordet's work, German scientist Paul Ehrlich, in the early 1900s, integrated complement into his side-chain theory of immunity, positing that antibodies (termed amboceptors) specifically fixed complement to target and destroy pathogens or altered cells. Ehrlich coined the term "complement" in 1899 to denote this serum factor that "complemented" antibody action in immune reactions, including fixation phenomena. His theoretical framework emphasized complement's enzymatic-like role in cytolysis, influencing early immunological research.

Key developments and milestones

In the mid-20th century, significant progress was made in isolating the individual components of the complement system, laying the foundation for understanding its biochemical structure. During the 1940s and , researchers such as Manfred Mayer developed standardized hemolytic assays that enabled the systematic fractionation of , leading to the identification of multiple heat-labile factors. By the late and into the , competitive efforts by groups including those led by Irwin H. Lepow resulted in the purification and characterization of the nine core components of the classical pathway, designated C1 through C9, through techniques like and . These isolations confirmed the sequential activation nature of the cascade and were pivotal in shifting research from phenomenological observations to molecular dissection. The 1950s and 1960s also saw the detailed elucidation of the classical pathway's mechanism, primarily through the work of Hans J. Müller-Eberhard and collaborators. Müller-Eberhard's group purified key proteins and demonstrated the stepwise assembly, including the formation of the as the complex C4b2a, which cleaves to amplify the response. This three-component (C4b, C2a) was shown to be central to opsonization and downstream effector functions, with structural analyses revealing the proteolytic and binding domains involved. These findings integrated earlier hemolytic studies into a coherent enzymatic cascade model, influencing subsequent pathway research. The 1970s marked the discovery of the alternative pathway, expanding the complement system's activation routes beyond antibody dependence. Douglas T. Fearon and K. Frank Austen demonstrated that could undergo spontaneous hydrolysis to initiate activation without classical initiators, stabilized by and amplified via factor B and factor D. was identified as a stabilizer of the (C3bBb), while factor D acted as the cleaving factor B. This pathway, building on earlier properdin system hypotheses from the , highlighted complement's role in innate immunity against microbes lacking specific antibodies. From the 1980s to the 1990s, the was identified as a third activation route, paralleling the classical pathway but triggered by carbohydrate recognition. Mannose-binding (MBL) was shown to associate with MBL-associated serine proteases (MASPs), leading to C4 and C2 cleavage and formation analogous to C4b2a. Concurrently, genetic studies mapped many complement regulators to the regulators of complement activation (RCA) locus on chromosome 1q32, including genes for CR1, CR2, , and C4-binding protein. and linkage analyses confirmed this cluster's organization, revealing evolutionary duplications and implications for regulation. In the 2000s, advances in provided atomic-level insights into complement proteins, exemplified by of . The 2005 structure of native revealed its multi-domain architecture, including thioester-containing and anaphylatoxin domains, elucidating conformational changes upon to C3b. These visualizations clarified substrate recognition and regulatory interactions, facilitating targeted studies on pathway convergence and inhibition.

Components and

Major complement proteins

The major complement proteins are soluble components primarily synthesized by hepatocytes and circulating in , where they constitute approximately 15% of the total fraction at combined concentrations exceeding 3 g/L. These proteins are classified based on their roles in initiating specific pathways or participating in the central and terminal phases of the cascade.

Classical Pathway Initiators

C1q serves as the recognition subunit of the classical pathway, a hexameric protein composed of 18 polypeptide chains arranged into six globular heads connected by collagen-like stalks, enabling binding to Fc regions of antigen-bound IgM or IgG or to certain non-antibody ligands on surfaces. C1r and C1s are homologous zymogenic serine proteases that assemble into a calcium-dependent (C1r)₂(C1s)₂ tetramer bound to C1q, forming the C1 complex. Upon , C1s cleaves and to form the . C4 is a 200 present at 0.2–0.4 g/L in , cleaved into C4a and C4b, with C4b binding covalently to surfaces. C2, a 102 at 0.02–0.04 g/L, is cleaved by C1s into C2a and C2b; C2a associates with C4b to form the convertase. Plasma concentrations of these initiators are relatively low, with C1q at 0.12–0.22 g/L, C1r at approximately 0.03–0.04 g/L, and C1s at 0.031 g/L.

Central Component

C3 is the pivotal protein common to all three complement pathways and the most abundant complement component in , with concentrations of 1–2 g/L. This 185 kDa two-chain features an exposed bond within its α-chain that becomes reactive upon proteolytic , allowing nucleophilic attack and covalent linkage to nearby or hydroxyl groups on target surfaces for enhanced opsonization.

Alternative Pathway Components

Factor B is a 93 kDa zymogenic structurally similar to , circulating at 0.2–0.3 g/L and serving as the substrate for the pathway upon binding to C3b. Factor D, a compact 24 kDa active (lacking a zymogen form), cleaves factor B at low concentrations of 0.001–0.002 g/L. , a 53 kDa existing as oligomers, functions as the sole positive regulator of the pathway by binding and stabilizing the , with levels of 0.004–0.025 g/L.

Lectin Pathway Initiators

Mannose-binding lectin (MBL) is a collagenous collectin protein forming oligomeric structures that recognize neutral carbohydrate patterns (e.g., , ) on microbial surfaces, with highly variable concentrations (median ~1.3 μg/mL, range <0.005–12 μg/mL) due to genetic polymorphisms. Ficolins, including ficolin-1 (M-ficolin), ficolin-2 (L-ficolin), and ficolin-3 (H-ficolin), are structurally similar to MBL with fibrinogen-like recognition domains binding acetylated groups on pathogens; concentrations are ~0.005 g/L for ficolin-1, ~0.005 g/L for ficolin-2, and 0.02–0.03 g/L for ficolin-3. MASP-1 and MASP-2 are serine proteases analogous to C1r and C1s, respectively, associating with MBL or ficolins in a Ca²⁺-dependent manner to initiate the pathway; MASP-1 circulates at ~0.011 g/L, while MASP-2 is present at lower levels of ~0.0005 g/L. Like the classical pathway, activation leads to cleavage of and C2.

Terminal Components

The terminal complement components C5 through C9 mediate lytic effector function by sequentially assembling into the (MAC). C5, a 188 kDa disulfide-linked heterodimer at 0.07–0.2 g/L, is the initial substrate for C5 convertases. C6, C7, and C8 (concentrations ~0.04–0.08 g/L each) bind sequentially to form a pre-lytic complex that inserts into bilayers, recruiting C9 (0.058 g/L), which polymerizes into a β-barrel of 10–18 monomers to permeabilize target membranes.

Protein fragment nomenclature

The complement system employs a standardized for its proteins and their fragments to ensure clarity in scientific communication. The core components originating from the classical pathway are designated with the prefix "C" followed by numbers 1 through 9 (C1–C9), reflecting their order of discovery rather than activation sequence. Subcomponents of these proteins, such as those of C1, are denoted with lowercase letters (e.g., C1q, C1r, C1s). This basic numbering system was established in by an international committee to unify designations for complement components. Upon proteolytic cleavage during activation, complement proteins generate fragments that are named based on their size and position relative to the parent molecule. The smaller N-terminal fragment is typically suffixed with "a" (e.g., C3a from ), while the larger C-terminal fragment receives the "b" suffix (e.g., C3b). Inactivated or modified forms of these fragments are indicated by a lowercase "i" prefix (e.g., iC3b, derived from further processing of C3b). This fragment notation convention was formalized in the early 1980s through recommendations by the (WHO) and the International Union of Immunological Societies (IUIS), building on earlier proposals to address inconsistencies in describing activation products. Enzyme complexes known as convertases, which cleave key complement proteins like and , follow a composite naming system reflecting their subunit composition. The classical pathway is denoted as C4b2a (or C4bC2a), comprising the C4b fragment, (the activated smaller fragment of ), and sometimes associated with C1s. In contrast, the alternative pathway is named , consisting of C3b and the Bb fragment from factor B. These names adhere to the same fragment rules and were standardized in a 1981 WHO-IUIS report specifically for the pathway, with broader updates in 2014 by the Complement Nomenclature Committee. Among the fragments, the anaphylatoxins—C3a, C4a, and C5a—represent small peptides released from the N-terminal portions of their respective parent proteins upon cleavage by convertases or other proteases. These ~8–11 kDa molecules are potent mediators of , binding to specific receptors to induce release, , and . Their directly applies the "a" and was consistently defined in the 1981 standardization efforts to distinguish them from opsonizing "b" fragments. The overall system, including these specifics, underwent its first major revision since 1981 in 2014 to incorporate newly discovered proteins and resolve ambiguities, such as those in C2 fragment designations.

Activation Pathways

Classical pathway

The classical pathway of the complement system is triggered by the binding of the C1q subcomponent to the Fc regions of immunoglobulin M (IgM) or immunoglobulin G (IgG) antibodies that are complexed with antigens on the surface of pathogens or damaged cells. This antibody-dependent recognition provides a link to the adaptive immune response, ensuring targeted activation only at sites of immune complex formation, without spontaneous initiation in the absence of antibodies. Upon binding, typically requiring at least two Fc regions for IgG or one pentameric IgM molecule, C1q undergoes a conformational change that activates the associated serine proteases C1r and C1s within the C1 complex (C1qrs₂). Activated C1r then autoactivates and cleaves C1s to its active form, enabling the cascade to proceed. The activated C1s protease subsequently cleaves the complement protein into two fragments: the small anaphylatoxin C4a, which is released into the fluid phase, and the larger C4b fragment, which covalently attaches to nearby surfaces via a reactive bond, often anchoring near the immune . C1s also cleaves , a single-chain , into the smaller C2b fragment (released) and the larger C2a fragment, which binds non-covalently to the surface-bound C4b to form the classical pathway , C4b2a (also known as C4bC2a). This bimolecular is stabilized on the target surface and exhibits specificity for immune complexes due to the initial antibody-mediated localization of C1q. The C4b2a then proteolytically cleaves the central complement protein into C3a, another anaphylatoxin that promotes , and C3b, which serves as a key by binding covalently to the surface. Surface-bound C3b can associate with additional C4b2a molecules to form the , C4b2a3b, which initiates the terminal complement cascade by cleaving into C5a (an anaphylatoxin and chemoattractant) and C5b, the latter nucleating assembly of the membrane attack complex. This stepwise amplification ensures efficient deposition of complement fragments only at antibody-coated targets, enhancing pathogen clearance through opsonization and subsequent effector functions.

Alternative pathway

The alternative pathway of the complement system provides a continuous, antibody-independent mechanism for innate immune surveillance, initiated by the spontaneous of the bond within the central component , generating the metastable form C3(H₂O). This occurs at a low rate in , altering C3's conformation to expose binding sites for factor B, a . Factor B binds to C3(H₂O), forming the proenzyme C3(H₂O)B, which is then cleaved by factor D—a circulating —into Ba and Bb fragments, yielding the fluid-phase C3(H₂O)Bb. This initial convertase cleaves additional C3 molecules into C3a (an anaphylatoxin) and C3b, establishing a basal level of known as the tickover mechanism. The tickover mechanism maintains a steady, low-level generation of C3b in the fluid phase, preventing widespread activation on host cells while enabling rapid response to ; this discrimination relies on the absence of membrane-bound regulators like () and membrane cofactor protein (MCP) on non-self surfaces, which would otherwise disassemble the convertase. Upon contact with pathogen surfaces—such as bacterial cell walls or foreign materials—C3b deposits covalently via its reactive , recruiting factor B to form the surface-bound complex C3bB. Factor D cleaves factor B in this complex, producing the active C3bBb, which amplifies C3b production exponentially by cleaving more C3. , the only known positive regulator in the pathway, binds to and stabilizes C3bBb, extending its approximately 5- to 10-fold, from ~90 seconds to 7–15 minutes and directing activation toward target surfaces. As an amplification loop, the alternative pathway generates the majority of C3b molecules during complement activation, regardless of the initiating pathway, thereby enhancing opsonization, , and membrane attack complex formation across the system; this role underscores its function as a and booster mechanism for innate immunity. The concept of "protected surfaces," where alternative pathway convertases form stably on non-host materials due to resistance to inactivation, was first elucidated in seminal work demonstrating selective activation on rabbit erythrocytes versus zymosan particles.

Lectin pathway

The lectin pathway of the complement system is initiated by the recognition of specific carbohydrate patterns on microbial surfaces through molecules, primarily mannose-binding (MBL) and ficolins. These soluble proteins circulate in as complexes with mannose-binding lectin-associated serine proteases (MASPs), including MASP-1, MASP-2, and MASP-3. Upon binding to mannose or residues on pathogens, conformational changes in MBL or ficolins trigger autoactivation of MASP-1, which in turn activates MASP-2. This process is analogous to the activation of C1r and C1s in the classical pathway, reflecting structural and functional similarities among these serine proteases. Activated MASP-2 then cleaves complement component into C4a and C4b fragments, followed by cleavage of into C2a and C2b. The resulting C4b and C2a fragments associate to form the C4b2a, which is identical to the convertase in the classical pathway and proceeds to cleave , amplifying the complement response. MASP-1 enhances this efficiency by facilitating MASP-2 activation under physiological conditions, ensuring robust pathway initiation without reliance on antibodies. Variants of the initiating complexes include those formed by ficolins (H-ficolin, L-ficolin, and M-ficolin), which recognize acetylated groups on microbes and associate with the same MASPs as MBL. Additionally, collectin-11 (also known as CL-K1) functions as another recognition molecule, forming complexes with MASPs to bind pathogen-associated molecular patterns and initiate the pathway. These diverse initiators broaden the lectin pathway's ability to detect a range of microbial threats in an antibody-independent manner. Evolutionarily, the lectin pathway shares a common ancestry with the classical pathway, as evidenced by the homology between MASPs and the C1r/C1s proteases; both families belong to the C1r/C1s/sea urchin VEGF/plasminogen () superfamily, with conserved modular structures including , EGF, and domains. This structural similarity underscores the pathways' parallel mechanisms for downstream activation while highlighting the pathway's role in innate immunity through direct carbohydrate recognition.

Effector Mechanisms

Membrane attack complex formation

The terminal phase of complement activation, common to all three pathways, begins with the formation of enzymes that cleave the central complement protein into its fragments C5a and C5b. In the classical and pathways, the C5 convertase is the complex C4b2a3b, where C3b associates with the C4b2a to enable C5 cleavage. In the alternative pathway, the convertase is C3bBb3b, stabilized by and similarly cleaving C5. This cleavage step marks the irreversible commitment to the terminal pathway, with C5b serving as the initiating subunit for downstream assembly while C5a acts as an anaphylatoxin. Assembly of the , also known as C5b-9, proceeds sequentially in the fluid phase before insertion. C5b rapidly binds to form the metastable C5b6 intermediate, which then associates with C7 to generate C5b67; the latter complex exposes hydrophobic domains on C7 that facilitate binding and penetration into the of the target . C8 subsequently binds to C5b67, forming C5b-8, which further embeds into the and creates a low-affinity for C9. Multiple C9 monomers (typically 10 to 18) then polymerize onto C5b-8 in a unidirectional, manner, forming a β-barrel transmembrane channel approximately 100 (10 ) in . This poly-C9 structure completes the MAC pore, with the number of C9 units determining the pore's size and lytic efficiency. The primary lytic mechanism of the MAC involves disruption of target cell membrane integrity, leading to uncontrolled influx of water and ions that causes colloid osmotic and . The pore's large diameter allows passage of small molecules and ions, rapidly depolarizing the and compromising cellular , particularly effective against and enveloped viruses. MAC stability is enhanced by the cylindrical arrangement of C9 monomers, which resists , although the complex can disassemble over time if not fully polymerized. In quantitative terms, complete pores with 12-18 C9 units exhibit maximal cytolytic activity, while incomplete assemblies may be less stable. Beyond , sublytic concentrations of can elicit non-lytic signaling in host cells, promoting inflammatory responses without cell death. Insertion of partial pores triggers calcium influx and activation of pathways such as and the , leading to production and enhanced immune cell recruitment. These effects are particularly relevant in endothelial and epithelial cells, where sublytic contributes to vascular inflammation and tissue remodeling during immune responses.

Opsonization and chemotaxis

Opsonization is a key effector mechanism of the complement system, whereby activated complement proteins tag pathogens and immune complexes for enhanced recognition and uptake by phagocytic cells. The primary opsonin is C3b, generated through cleavage of C3 during complement activation, which covalently binds to the surface of microbes or altered host cells, marking them for phagocytosis. This C3b coating facilitates binding to complement receptor 1 (CR1, also known as CD35) on phagocytes such as macrophages and neutrophils, promoting efficient engulfment. Further proteolytic processing of C3b yields iC3b, which binds to complement receptor 3 (CR3, CD11b/CD18) and complement receptor 4 (CR4), extending the opsonization window and enabling phagocytosis even after initial C3b decay. In contrast, C4b, produced in the classical and lectin pathways, plays a minor opsonizing role by binding to CR1, but its contribution is less pronounced compared to C3b/iC3b due to lower deposition efficiency and rapid inactivation. Complement-mediated opsonization dramatically boosts phagocytic efficiency, often enhancing uptake by 10- to 100-fold compared to non-opsonized targets, underscoring its in innate immunity. This process not only accelerates clearance but also synergizes with antibody-mediated opsonization via Fcγ receptors, where CR1 acts as a co-receptor to lower the for engulfment. For instance, C3b/iC3b-opsonized are rapidly internalized by professional , preventing dissemination and limiting . Beyond opsonization, complement fragments drive and through anaphylatoxins C3a and C5a, small peptides released upon C3 and C5 . These anaphylatoxins bind to G-protein-coupled receptors—C3a to C3aR and C5a to C5aR1 (CD88)—on immune cells including , , and neutrophils, triggering intracellular signaling via Gαi proteins. This binding induces degranulation, releasing and other mediators that increase and promote local . Concurrently, C3a and C5a act as potent chemoattractants, directing leukocyte migration to infection sites by activating chemotactic responses and enhancing adhesion molecule expression on endothelial cells. Complement also facilitates the non-inflammatory clearance of apoptotic cells, maintaining tissue homeostasis without triggering damaging responses. C1q binds directly to exposed phospholipids or altered surface molecules on apoptotic cells, initiating classical pathway activation and localized C3b deposition for opsonization. Phagocytes then recognize these opsonized cells via CR1 and CR3, leading to silent engulfment that suppresses pro-inflammatory cytokine release, such as IL-12, and promotes anti-inflammatory signals like TGF-β production. This mechanism prevents secondary necrosis and autoimmunity by efficiently removing over 10^11 apoptotic cells daily in humans.

Regulation

Soluble regulatory proteins

The soluble regulatory proteins of the complement system are circulating components that prevent uncontrolled in the fluid phase, thereby limiting and tissue damage while allowing targeted responses on surfaces. These inhibitors act at various steps of the pathways, primarily by inhibiting activity, accelerating the of convertase enzymes, or serving as cofactors for proteolytic inactivation of complement fragments. Key examples include , , factor I, and C4-binding protein, each with distinct roles in regulating the classical, , and pathways. C1 inhibitor (C1-INH), a serine protease inhibitor (serpin) family member, is the primary regulator of the classical and lectin pathways by irreversibly binding and inhibiting the activated serine proteases C1r and C1s of the C1 complex, as well as MBL-associated serine proteases (MASPs) in the lectin pathway, thereby preventing spontaneous C4 and C2 cleavage. This inhibition occurs through formation of a covalent complex that sterically blocks substrate access, maintaining complement in a quiescent state in plasma. C1-INH circulates at a plasma concentration of approximately 0.25 g/L (range: 0.15–0.35 g/L), with a half-life of 67–72 hours, and is primarily synthesized by hepatocytes. Deficiency or dysfunction of C1-INH, as seen in hereditary angioedema (HAE) types I and II, leads to unchecked bradykinin production via dysregulation of the kallikrein-kinin system and uncontrolled activation of the classical complement pathway (evidenced by low C4 and C2 levels), but the recurrent episodes of subcutaneous and mucosal edema result primarily from bradykinin-induced vascular permeability. Factor H, a 155-kDa glycoprotein composed of 20 short consensus repeats (SCRs), serves as the principal soluble regulator of the alternative pathway by binding to C3b, accelerating the decay of the C3 convertase (C3bBb) through displacement of Bb, and acting as a cofactor for factor I-mediated cleavage of C3b to iC3b. Its affinity for C3b is enhanced on host surfaces via interactions with sialic acid and glycosaminoglycans, distinguishing self from non-self. Factor H is present in plasma at concentrations of 250–600 μg/mL and is mainly liver-derived, though extrahepatic production occurs in fibroblasts and endothelial cells. Inherited deficiencies or mutations in factor H, often involving the C-terminal SCRs 19–20, are strongly associated with atypical hemolytic uremic syndrome (aHUS) and age-related macular degeneration, where impaired regulation leads to excessive C3 activation and endothelial damage. Factor I, a soluble of approximately 88 kDa, functions as the central inactivator of complement by proteolytically cleaving b to iC3b and further to C3dg, as well as C4b to C4d, but only in the presence of cofactors such as or C4-binding protein. This cofactor-dependent activity limits the amplification loop of the alternative pathway and inhibits classical/ convertases, preventing widespread opsonization in plasma. Plasma levels of factor I are typically 20–50 μg/mL, with synthesis occurring in the liver and monocytes. Complete or partial deficiencies in factor I result in uncontrolled consumption, recurrent pyogenic infections (e.g., with species), and susceptibility to autoimmune conditions like systemic , as the lack of inactivation allows persistent complement activation. C4-binding protein (C4BP), a large oligomeric (molecular mass ~570 kDa) consisting of seven α-chains and one β-chain linked to , regulates the classical and pathways by binding C4b, dissociating the (C4b2a), and serving as a cofactor for factor I-mediated degradation of C4b. This action curtails activation downstream of C1-INH, particularly in fluid phase where C4BP concentrations exceed those of C4b. C4BP circulates at ~200 μg/mL (range: 150–300 μg/mL) and is predominantly hepatic in origin, with levels increasing as an acute-phase reactant. Although rare, C4BP deficiency has been linked to increased autoimmune manifestations, such as and , due to dysregulated classical pathway activity, though it often co-occurs with alterations affecting .

Membrane-associated regulators

Membrane-associated regulators of the complement system are cell surface proteins that locally inhibit complement activation to protect host tissues from inadvertent damage during immune responses. These proteins, anchored to the plasma membrane via transmembrane domains or glycosylphosphatidylinositol (GPI) linkages, act at various stages of the complement cascade, primarily on self cells to prevent amplification of C3 convertases, degradation of opsonins, and assembly of the membrane attack complex (MAC). By restricting complement activity to pathogen surfaces or altered self cells that express lower levels of these regulators, they contribute to self/non-self discrimination, ensuring that autologous cells are spared while foreign entities are targeted. Decay-accelerating factor (DAF, CD55) is a GPI-anchored expressed on the surface of most cells, including erythrocytes, leukocytes, endothelial cells, and epithelial cells. It accelerates the decay of classical and pathway and convertases by binding to and dissociating their components, thereby limiting C3b deposition and downstream effector functions on host membranes. This protective mechanism is crucial for preventing complement-mediated of self cells in contact with . Membrane cofactor protein (MCP, ) is a ubiquitously distributed on nucleated , such as endothelial cells, fibroblasts, and leukocytes, but absent on erythrocytes. It serves as a cofactor for the factor I, facilitating the proteolytic cleavage and inactivation of C3b and C4b deposited on cell surfaces, which halts the of complement in both classical and alternative pathways. MCP's broad tissue expression ensures targeted regulation at sites of potential complement exposure. Complement receptor 1 (CR1, CD35) is a transmembrane predominantly found on erythrocytes, monocytes, neutrophils, and B cells, with lower expression on other leukocytes and tissue cells. It combines decay-accelerating activity, similar to , by promoting the dissociation of and convertases, and cofactor activity for factor I-mediated cleavage of C3b and C4b. On erythrocytes, CR1 plays a key role in immune complex clearance while protecting the carrier cells from complement damage. Protectin (CD59) is a GPI-anchored protein widely expressed on human cells, including hematopoietic cells, endothelial cells, and epithelial surfaces. It binds to the partially formed C5b-8 complex and inhibits the recruitment and polymerization of C9, thereby preventing the formation of the pore-forming MAC on host cell membranes. This terminal pathway inhibition provides a final safeguard against complement-mediated cytolysis. Collectively, these regulators are constitutively expressed at varying densities on host cells, with higher levels on cells frequently exposed to complement, such as and vascular elements. Their absence or downregulation on pathogens or stressed self cells allows unchecked complement activation, facilitating immune discrimination between self and non-self. Seminal studies on their structures and functions, such as those elucidating DAF's decay mechanism and CD59's MAC inhibition, have established their essential roles in complement .

Physiological Roles

Role in innate immunity

The complement system serves as a cornerstone of innate immunity by providing rapid, antibody-independent defense against through direct recognition, activation of effector mechanisms, and coordination of inflammatory responses. In this capacity, it operates primarily via the and pathways, which enable surveillance and elimination of invading microbes without prior sensitization. These pathways initiate a proteolytic cascade that converges on the central component , leading to opsonization, inflammation, and cytolytic activity, thereby bridging pathogen detection with immune clearance. A key aspect of the complement system's innate role is its direct antimicrobial activity, achieved through formation of the (MAC) that lyses susceptible . In the pathway, spontaneous of exposes a bond, allowing low-level deposition on surfaces and amplification via factor B and D, culminating in MAC assembly (C5b-9) that perforates bacterial membranes. For instance, this pathway is critical for lysing such as Neisseria meningitidis, where complement activation on the bacterial surface drives rapid killing in serum, as evidenced by heightened susceptibility of capsule-deficient mutants to alternative pathway-mediated . The complements this by recognizing microbial glycans through pattern recognition molecules like mannose-binding (MBL) and ficolins, which bind motifs on surfaces and trigger MASP-mediated and cleavage, leading to activation and MAC formation. This mechanism contributes to the of enveloped viruses, such as , by targeting viral glycoproteins, and certain parasites, including Trypanosoma cruzi, where lectin-initiated MAC insertion disrupts parasite membranes and limits infection propagation. Beyond lysis, the complement system facilitates innate immune surveillance by recruiting to sites. Cleavage of during pathway activation generates C5a, a potent anaphylatoxin that acts as a chemoattractant for neutrophils, promoting their migration across endothelial barriers via C5a receptor (C5aR1) signaling. This enhances engulfment and clearance at localized inflammatory foci, amplifying the innate response without adaptive involvement. The lectin pathway's further underscores this surveillance function, as MBL and ficolins detect evolutionarily conserved microbial glycans—such as mannose-rich structures on bacterial lipopolysaccharides or fungal mannans—that differ from host sialylated glycans, thereby distinguishing self from non-self and initiating targeted complement deposition. The complement system's innate functions reflect its evolutionary conservation, predating adaptive immunity and extending to . Homologs of , the pivotal and anaphylatoxin precursor, have been identified in non-vertebrate deuterostomes, including the purple sea urchin (Strongylocentrotus purpuratus), where C3-like proteins in coelomocytes mediate proteolytic cascades for defense. This ancient presence, traceable to early metazoans like sponges, highlights complement's role as a foundational innate mechanism, with expansions in vertebrates enhancing its efficiency.

Interaction with adaptive immunity

The complement system bridges innate and adaptive immunity by enhancing B-cell activation through the deposition of fragments on antigens. Specifically, C3d-tagged antigens bind to (CR2, also known as CD21) on B cells, which co-ligates with the (BCR) via the CD19/CD81 complex, delivering a costimulatory signal that amplifies BCR signaling 10- to 100-fold and lowers the activation threshold. This mechanism facilitates more efficient antigen-specific B-cell responses, particularly for low-avidity interactions, and is crucial for . Antibodies provide feedback to the complement system primarily via the classical pathway, where IgM and certain IgG subclasses (IgG1 and IgG3 in humans) bind s and recruit C1q to initiate complement activation. This leads to C3b and C3d deposition on immune complexes, which not only promotes opsonization but also supports B-cell affinity maturation in germinal centers by facilitating antigen delivery to (FDCs) and B cells. Complement-mediated enhancement of antibody responses can increase titers by 10- to 1000-fold in model systems, underscoring its role in amplifying adaptive . Complement anaphylatoxins C3a and C5a further integrate with cellular adaptive responses by modulating (DC) function and T-cell . C3a and C5a, generated locally during immune activation, bind to receptors on DCs (C3aR and C5aR), promoting their maturation, production (e.g., IL-12), and migration to nodes, which enhances priming of naive T cells. These signals also influence T-helper cell polarization, with C5a favoring Th1 responses through DC activation while C3a can promote Th2 skewing in certain contexts, thereby fine-tuning adaptive immunity. In long-term adaptive immunity, complement contributes to immunological by aiding the clearance of immune complexes via CR1 (CD35) on erythrocytes and FDCs, which prevents excessive and sustains for B-cell maintenance. This process ensures efficient removal of circulating complexes while preserving depots on FDCs for secondary responses.

Pathophysiological Roles

Complement deficiencies

Complement deficiencies refer to inherited defects in the proteins of the complement system, which impair its and , leading to increased to infections and, in some cases, autoimmune conditions. These deficiencies are primarily genetic and affect specific components of the classical, , or pathways, resulting in recurrent bacterial infections due to compromised opsonization, , or membrane attack complex () formation. Most complement deficiencies follow an autosomal recessive inheritance pattern, requiring biallelic mutations for clinical manifestation, though exceptions like properdin deficiency are X-linked. The prevalence varies by component and population; for instance, C2 deficiency occurs in approximately 1 in 20,000 individuals of Caucasian descent, making it one of the more forms. Deficiencies in the terminal components (C5 through C9) disrupt formation, severely impairing the lytic killing of certain while leaving upstream functions like opsonization intact. Affected individuals face a dramatically elevated risk of invasive infections by species, such as , with the risk increased up to 10,000-fold compared to the general population. These patients often experience recurrent , though other infections are less frequent due to preserved early pathway activities. Early classical pathway deficiencies, involving C1, , or , compromise the initiation of complement activation triggered by immune complexes or antibodies. This leads to inefficient clearance of apoptotic cells and immune complexes, predisposing individuals to systemic lupus erythematosus (SLE)-like characterized by autoantibodies, , and . In addition to autoimmune risks, these deficiencies increase vulnerability to infections with encapsulated bacteria, though less severely than central pathway defects. Alternative pathway deficiencies, particularly in properdin or factor D, hinder the amplification loop that sustains complement activation on microbial surfaces. stabilizes the , and its absence results in fulminant meningococcal infections with high mortality, often presenting as severe or . Factor D deficiency similarly impairs alternative pathway initiation, leading to recurrent Neisseria infections, though cases are rarer and inheritance is autosomal recessive for factor D but X-linked for properdin. C3 deficiency represents a critical bottleneck, as is central to all complement pathways and essential for opsonization, , and downstream effector functions. Complete deficiency causes severe, recurrent pyogenic infections starting in early childhood, predominantly by and species, including , , and due to failed amplification and bacterial clearance. These patients may also develop immune complex-mediated diseases, but infections dominate the clinical picture.

Involvement in diseases

The complement system contributes to pathology in various diseases through excessive activation or dysregulation, leading to tissue damage, inflammation, and immune-mediated injury. In autoimmune conditions, dysregulated complement components such as C3 and C5a exacerbate disease progression by amplifying inflammatory responses triggered by immune complexes. For instance, in rheumatoid arthritis (RA), immune complexes containing IgG deposit in synovial tissues, activating the classical pathway and resulting in C3 deposition in over 90% of RA synovial fluids, which promotes chronic joint inflammation and cartilage destruction. Similarly, in systemic lupus erythematosus (SLE), immune complexes activate the classical complement pathway via C1q binding, generating C3 fragments and C5a that drive neutrophil recruitment and tissue injury in organs like the kidneys and skin, contributing to lupus nephritis and vasculitis. Atypical hemolytic uremic syndrome (aHUS) exemplifies complement dysregulation leading to vascular pathology, where mutations in complement (CFH) impair alternative pathway regulation, causing uncontrolled activity and excessive endothelial cell damage. These CFH mutations, often loss-of-function variants, reduce the protein's ability to bind and inactivate C3b on host surfaces, resulting in persistent complement activation, , , and through endothelial lysis and thrombus formation. Paroxysmal nocturnal hemoglobinuria (PNH) involves complement-mediated due to deficiencies in membrane regulators CD55 and , rendering red blood cells highly susceptible to the membrane attack complex (). The absence of CD55, which accelerates decay of and convertases, combined with deficiency, which inhibits assembly, leads to chronic intravascular , nocturnal hemoglobinuria, and increased risk as affected erythrocytes are lysed by alternative and classical pathway activation in the bloodstream. In age-related macular degeneration (), chronic complement activation in the contributes to formation and photoreceptor degeneration, with variants in the CFH gene serving as major risk factors. , extracellular deposits beneath the , contain complement proteins including and C5b-9 (), indicating local alternative pathway overactivation that promotes and choroidal in late-stage ; the common Y402H polymorphism in CFH reduces its regulatory function, heightening susceptibility to this degenerative process. Recent studies have linked complement hyperactivation to the observed in severe cases, where infection triggers excessive classical and alternative pathway activation, amplifying proinflammatory responses. Post-2020 analyses show elevated plasma levels of complement activation products like C5a and C3a in critically ill patients, correlating with , , and multi-organ failure as these anaphylatoxins recruit neutrophils and exacerbate the hyperinflammatory state.

Diagnostic and therapeutic approaches

Diagnosis of complement system disorders relies on functional assays that evaluate pathway activity and specific protein quantification to identify deficiencies or dysregulation. The CH50 assay measures total hemolytic complement activity in the classical pathway by assessing the ability of patient serum to lyse antibody-sensitized sheep erythrocytes, serving as a screening tool for deficiencies in classical pathway components from C1 to C9. Similarly, the AH50 assay evaluates pathway function through of unsensitized rabbit erythrocytes, detecting abnormalities in factors B, D, , or C3. These hemolytic assays provide an overall assessment of pathway integrity but may miss isolated regulator defects. For precise identification of individual component levels, enzyme-linked immunosorbent assays (ELISAs) are employed, such as those quantifying concentrations in , which help confirm hereditary or acquired deficiencies associated with recurrent infections or autoimmune conditions. Functional assays for regulators, including cofactor activity tests for factor I or decay acceleration assays for , utilize ELISA-based detection of C3b deposition or formation to assess inhibitory protein efficacy. These methods enable targeted diagnosis, particularly in (aHUS) or (PNH), where pathway dysregulation predominates. Therapeutic modulation of the complement system primarily involves inhibitors targeting key activation points to mitigate excessive activity in diseases like PNH and aHUS. , a against , was approved by the FDA in 2007 for reducing in PNH and in 2011 for aHUS, preventing C5a and formation. , a longer-acting inhibitor with an extended half-life, received FDA approval in 2018 for PNH and later for aHUS, allowing less frequent dosing while maintaining efficacy. Proximal inhibition strategies include , a pegylated inhibitor approved by the FDA in 2021 for PNH, which blocks to address both intravascular and extravascular . Emerging therapies target alternative pathway amplifiers, such as danicopan, an oral factor D inhibitor approved by the FDA in 2024 as add-on therapy to C5 inhibitors for PNH patients with persistent extravascular hemolysis, enhancing hemoglobin levels in clinical trials. For complement deficiencies, gene therapy approaches are in preclinical development; for instance, adeno-associated virus (AAV)-mediated delivery of complement factor H has shown promise in resolving C3 glomerulopathy models by restoring regulation. Recent 2025 studies on truncated CFH AAV therapy have demonstrated long-term disease reversal in models. These strategies aim to provide durable correction, though clinical translation remains challenged by immune responses to vectors.

Emerging and Specialized Roles

Role in the central nervous system

The complement system contributes to brain development by facilitating , a process essential for refining neural circuits. During postnatal development, C1q, the initiating component of the , is expressed by neurons in response to signals from immature and localizes to developing s, particularly in the retinogeniculate pathway critical for maturation. This leads to the deposition of C3b opsonins on synapses, marking them for recognition and by via complement receptor 3 (CR3). Deficiency in C1q or results in impaired synapse elimination, with mice exhibiting persistent multiple innervation of axons onto thalamocortical neurons, underscoring complement's necessity for proper circuit refinement. Complement activity in the (CNS) is largely independent of systemic circulation due to the (BBB), which restricts serum protein leakage. Instead, complement components are synthesized locally by CNS-resident cells, primarily and , enabling rapid responses to developmental and homeostatic needs. produce key proteins such as and , while express terminal pathway components like C5-C9, supporting opsonization and phagocytic clearance without relying on peripheral sources. This localized synthesis maintains low baseline complement levels in the healthy while allowing upregulation during physiological remodeling. In neurodegenerative contexts, complement fragments C3a and C5a drive , exacerbating disease progression. In (MS), C5a and C3a anaphylatoxins activate and in cortical lesions, promoting proinflammatory release, blood-brain barrier disruption, and demyelination through classical and alternative pathway activation. Similarly, in (AD), these fragments amplify microglial responses around amyloid-β plaques, enhancing plaque compaction and associated synaptic loss while contributing to chronic . Complement opsonization of plaques, initiated by C1q, tags them for clearance but often leads to excessive glial activation and neurodegeneration when dysregulated. Preclinical studies in the 2020s highlight therapeutic potential for modulating complement in CNS disorders. C5aR antagonists, such as PMX205, reduce C5a-mediated microglial polarization toward proinflammatory states, decreasing pathology, , and cognitive deficits in AD mouse models. In neuroinflammatory models of hypoxia-ischemia and AD, these antagonists preserve synaptic integrity and mitigate plaque-associated without broadly impairing protective complement functions. Ongoing research emphasizes their ability to cross the , offering targeted intervention for complement-driven neurodegeneration.

Non-immune functions and evolution

Beyond its classical roles in immunity, the complement system contributes to various non-immune processes essential for tissue and development. In , C1q plays a critical protective role in trophoblast function and embryo implantation by facilitating the of extravillous trophoblasts into the and promoting spiral artery remodeling, which ensures adequate placental blood flow. This local production of C1q by decidual endothelial cells and trophoblasts helps maintain feto-maternal tolerance without triggering inflammatory responses. Complement components also support wound healing through angiogenic mechanisms. The anaphylatoxin C3a, generated during complement activation, stimulates endothelial cell proliferation and vascular tube formation, thereby enhancing neovascularization at injury sites to promote tissue repair. This process is particularly evident in models of cutaneous , where C3a signaling recruits pro-angiogenic cells and modulates the to accelerate closure. In metabolic regulation, complement factor C3, produced by adipocytes, links adipose tissue dysfunction to insulin resistance and obesity. Elevated C3 levels in obese individuals correlate with impaired glucose uptake and hepatic steatosis, as adipocyte-derived C3 activates local inflammation and disrupts insulin signaling pathways. Recent studies further highlight C3's involvement in cancer metabolism, where activation in tumor microenvironments promotes lipid reprogramming that fuels tumor growth independently of immune surveillance. Emerging has identified intracellular complement activation, termed the "complosome," as a key non-immune function regulating cellular processes such as , , and mitochondrial function in various tissues, with implications for diseases like cancer and neurodegeneration as of 2023. The complement system represents an ancient phylogenetic , with origins tracing back to the emergence of multicellular animals over 500 million years ago. Homologs of , the central component, have been identified in cnidarians such as , indicating that a primitive thioester-containing protein capable of opsonization and existed in these early metazoans. Unlike vertebrates, lower lack the membrane attack complex (MAC), relying instead on pathways involving C3-like molecules and factor B for recognition and clearance. Adaptive radiation of the complement system occurred in vertebrates, where duplications expanded the classical, , and pathways, enabling more sophisticated immune and non-immune functions. This diversification, evident from jawed vertebrates onward, integrated complement with adaptive immunity while preserving ancestral roles in development and .

References

  1. [1]
    COMPLEMENT: AN OVERVIEW FOR THE CLINICIAN - PMC
    The complement system is a major part of the innate immune system, protecting the host from pathogens, and made of 50 serum and membrane proteins.
  2. [2]
    The complement system and innate immunity - Immunobiology - NCBI
    The complement system, part of innate immunity, uses plasma proteins to opsonize pathogens, induce inflammation, and mark them for destruction by phagocytes.2-5. Complement is a system... · 2-9. Hydrolysis of C3 causes...
  3. [3]
    The complement system - PMC - NIH
    The complement system consists of a tightly regulated network of proteins that play an important role in host defense and inflammation.
  4. [4]
    The complement system: history, pathways, cascade and inhibitors
    There are three known pathways for complement activation: Classical, Alternative and Lectin pathway. Classical pathway. The classical pathway is initiated by ...
  5. [5]
    Overview of Complement Activation and Regulation - PMC
    Three main pathways can activate the complement system: classical, lectin, and alternative (Fig. 1). The classical pathway (CP) uses C1 and is triggered by ...
  6. [6]
    The complement system - PubMed - NIH
    The complement system is a network of proteins for host defense and inflammation, causing opsonization, pathogen removal, and cell lysis.
  7. [7]
    Complement and its role in innate and adaptive immune responses
    The complement system is crucial for innate defense, and it acts as a bridge between innate and adaptive immune responses, allowing integrated host defense.
  8. [8]
    Complement regulation: physiology and disease relevance - PMC
    Complement activation occurs in 4 main steps: (1) activation by one of three major pathways, (2) C3 convertase activation and amplification, (3) C5 convertase ...
  9. [9]
    Complement: an overview for the clinician - PubMed - NIH
    The complement system is an essential part of the immune system, involved in host defense, removing injured cells, and regulating adaptive immunity.
  10. [10]
    Complement and its role in innate and adaptive immune responses
    Dec 15, 2009 · Complement is a system of more than 30 proteins in the plasma and on cell surfaces, amounting to more than 3 g/L and constituting more than 15% ...
  11. [11]
    Complement System Part I – Molecular Mechanisms of Activation ...
    Complement is a complex innate immune surveillance system, playing a key role in defense against pathogens and in host homeostasis. The complement system is ...<|control11|><|separator|>
  12. [12]
    Complement System Part II: Role in Immunity - Frontiers
    Upon target recognition, C1q undergoes conformational change and activates the two serine proteases C1r and C1s, associated with it in the context of the C1 ...
  13. [13]
    A New Assessment of Thioester-Containing Proteins Diversity of the ...
    Evolution and Function of Thioester-Containing Proteins and the Complement System in the Innate Immune Response. ... C3, C4, C5 family of thioester-containing ...<|control11|><|separator|>
  14. [14]
    Complement Regulator - an overview | ScienceDirect Topics
    A network of proteins known as regulators of complement activation (RCA), which includes CR1, DAF, MCP, C4BP, FH, FHL-1, CSMD1 and FH-related family proteins.
  15. [15]
    Complement in disease: a defence system turning offensive - PMC
    For example, complement assists in the clearance of immune complexes, cellular debris, and apoptotic cells and has been associated with early development and ...
  16. [16]
    Complement as a regulator of adaptive immunity - PMC
    Complement plays a modulatory role in a variety of B cell functions, including activation and differentiation, antigen internalization and presentation, and ...
  17. [17]
    Complement and Humoral Immunity - PMC - NIH
    Three general pathways, i.e. classical, lectin and alternative, activate the complement system. The pathways are not distinct but overlap. For example, the ...
  18. [18]
    Complement System Part II: Role in Immunity - PMC - PubMed Central
    Complement is a complex innate immune surveillance system, playing a key role in host homeostasis, inflammation, and in the defense against pathogens.
  19. [19]
    Tolerance is dependent on complement C3 fragment iC3b binding ...
    We found that the development of antigen-specific tolerance is dependent on a complement activation product. The ligation of the complement C3 activation ...
  20. [20]
    100th Anniversary of Jules Bordet's Nobel Prize - Frontiers
    Sep 10, 2019 · Bordet favored the word alexin, coined by Hans Buchner (1850-1902) to characterize the substance responsible for the bacteriolysis.
  21. [21]
    Jules Bordet (1870-1961) | - Institut Pasteur
    "For his discoveries relating to immunity." In 1894 Jules Bordet joined the laboratory of Ilya Mechnikov under a grant from the Belgian government. He worked ...
  22. [22]
    Jules Bordet – Facts - NobelPrize.org
    Jules Bordet, Nobel Prize in Physiology or Medicine 1919. Born: 13 June 1870, Soignies, Belgium. Died: 6 April 1961, Brussels, Belgium.
  23. [23]
    Complement - SpringerLink
    Jun 19, 2018 · The complement system was initially viewed as an arm of innate immune defenses against invading pathogens by tagging them for elimination by professional ...Missing: milestones | Show results with:milestones
  24. [24]
    The complement system: History, pathways, cascade and inhibitors
    May 8, 2020 · During the late 19th and early 20th centuries, Jules Bordet, a Belgian scientist, identified a heat-sensitive component in fresh serum that ...Missing: Hans 1888-1890
  25. [25]
    Hans Joachim Muller-Eberhard | Biographical Memoirs: Volume 79
    In Uppsala Eberhard turned his attention to the nature of complement and determined the future direction of his research. While at Rockefeller he had reviewed ...
  26. [26]
    A nostalgic journey: The Hans J. Müller-Eberhard Memorial Lecture ...
    The scientific career and research contributions of Hans J. Müller-Eberhard to the field of complement research are presented in historical context, ...
  27. [27]
    Looking back on the alternative complement pathway - ScienceDirect
    The alternative pathway of complement originated from the Properdin pathway originally described by the Pillemer laboratory in the 1950s.
  28. [28]
    The Lectin Pathway of the Complement System—Activation ...
    The lectin pathway is the latest discovered activation route of the complement system (Figure 1). It was already observed in the 1980s that the MBL, in a ...
  29. [29]
    Analysis of the human regulators of complement activation (RCA ...
    Organization of the genes encoding complement receptors type 1 and 2, decay-accelerating factor, and C4-binding protein in the RCA locus on human chromosome 1.
  30. [30]
    Complement Protein Fragment Nomenclature | Sino Biological
    The components of complement were to be designated by the letter "C", the prime symbol, and numbers with activated products designated by the letter "a" added ...Missing: standardized | Show results with:standardized
  31. [31]
    Nomenclature of the alternative activating pathway of complement
    This terminology note outlines for the first time a standard nomenclature for the alternative activating pathway of complement. It was drafted by a group of ...
  32. [32]
    Complement Nomenclature—Deconvoluted - PMC - NIH
    Jun 7, 2019 · To facilitate communication in the field and better standardize complement nomenclature, we recommend adopting standard convention for C2 (C2 ...
  33. [33]
    Nomenclature of the alternative activating pathway of complement.
    Sep 1, 1981 · Nomenclature of the alternative activating pathway of complement. The Journal of Immunology, Volume 127, Issue 3, September 1981, Pages 1261 ...
  34. [34]
    Complement Nomenclature 2014 - ScienceDirect.com
    The first update since 1981 of the nomenclature used in the field of complement has been completed by the Complement Nomenclature Committee established ...
  35. [35]
    Early Components of the Complement Classical Activation Pathway ...
    Feb 15, 2016 · The activation pathways of the complement system converge at C3 and progress to the formation of the membrane attack complexes (MAC or C5b–C9) ...
  36. [36]
    Complement System Part I – Molecular Mechanisms of ... - Frontiers
    Following, the same enzyme will cleave C2 to generate the classical pathway C3 convertase C4b2a. (C) C4b2a will interact with C3 cleaving it and releasing the ...<|control11|><|separator|>
  37. [37]
    Alternative Pathway - Svar Life Science
    The hydrolysis changes the structure of C3 and increases its reactivity allowing for the binding of the serine protease Factor B, which in turn is cleaved by ...
  38. [38]
    The Tick-Over Theory Revisited: Formation and Regulation of the ...
    The complement system is activated by three pathways: the classical, lectin, and alternative pathways. Activation of any one of these pathways leads to the ...
  39. [39]
    Initiation of the alternative pathway of complement and the history of ...
    Sep 11, 2022 · Spontaneous activation of the alternative pathway via tickover may play a major role in human pathologies where tissue damage is complement- ...Missing: factor BD properdin
  40. [40]
    Alternative pathway of the complement system - Microbe Notes
    Aug 17, 2022 · The alternative pathway of the complement system is a cascade of enzymatic reactions which requires spontaneous hydrolysis of the internal C3 thioester bond.
  41. [41]
    The Influence of the Lectin Pathway of Complement Activation on ...
    Activation of complement via the lectin pathway is initiated by several pattern-recognition molecules (PRM), complexed with mannose-binding lectin-associated ...
  42. [42]
    Revised mechanism of complement lectin-pathway activation ...
    Jun 12, 2012 · The MASP-2–specific inhibitor SGMI-2 efficiently prevents C4 deposition from both normal (C) and preactivated (D) serum in the case of both MBL– ...
  43. [43]
    C1r, C1s and MASPs. A structural insight into activation and functions
    C1r, C1s, and MASPs initiate classical and lectin pathway activation of the complement system, forming complexes with pattern recognition molecules.Missing: homology | Show results with:homology
  44. [44]
    Molecular interactions between MASP-2, C4, and C2 and ... - PubMed
    In the lectin pathway, mannose-binding lectin (MBL) and ficolins bind to pathogens and activate MBL-associated serine protease-2 (MASP-2). MASP-2 cleaves C4 ...
  45. [45]
    Interactions between mannose-binding lectin and MASPs during ...
    Complement activation is initiated when complexes comprising mannose-binding lectin (MBL) or serum ficolins and MBL-associated serine protease-2 (MASP-2) bind ...
  46. [46]
    Mannan-binding lectin-associated serine protease (MASP) - PubMed
    Oct 15, 2012 · MASP-1 dramatically increases lectin pathway activity at physiological conditions through direct activation of MASP-2.
  47. [47]
    The Lectin Pathway of Complement and Rheumatic Heart Disease
    After the binding of MBL/MASPs, CL-K1/MASPs, or ficolin/MASPs complexes to their targets, MASP-1 can auto-activate and trigger MASP-2 (10), leading to C4 and C2 ...
  48. [48]
    Collectin-11/MASP complex: lectin complement pathway activation
    Dec 4, 2012 · Thus, we hypothesized that collectin-11 could be a novel initiation molecule in the lectin pathway of complement. We can show that collectin-11 ...
  49. [49]
    The Lectin Pathway of the Complement System—Activation ... - MDPI
    Plasma concentration of ficolins-1, -2, MBL, MASP-1 and MAP-19 significantly increased, while the level of MASP-3 decreased in IgAN patients. A low level of ...
  50. [50]
    Evolution of the initiating enzymes of the complement system
    Dec 14, 2001 · The domain structure and gene organization of the MASP/C1r/C1s serine protease family. The first line shows the domain structure of an ...
  51. [51]
    C1r, C1s and MASPs. A structural insight into activation and functions
    C1r, C1s and the mannose-binding lectin-associated serine proteases (MASPs) are responsible for the initiation of the classical- and lectin pathway activation ...
  52. [52]
    Classical Complement Pathway C3 C5 Convertase - an overview
    The classical complement pathway C3 C5 convertase is defined as a key enzymatic complex formed during complement activation that cleaves C3 into C3a and C3b ...
  53. [53]
    Functional Characterization of Alternative and Classical Pathway C3 ...
    C5 convertases cleave C5 into C5a, a potent mediator of leukocyte recruitment and inflammation, and C5b, the initiator of the membrane attack complex and cell ...
  54. [54]
    Assembly and Regulation of the Membrane Attack Complex Based ...
    Formation of the MAC is a sequential process. Upon complement activation, C5 is cleaved into C5a and C5b by the C5 convertase. C6 then captures a labile binding ...
  55. [55]
    Polymerization of C9 Enhances Bacterial Cell Damage & Killing
    MAC pores assemble by stepwise binding of C5b, C6, C7, C8 and finally C9, which can polymerize into a transmembrane ring of up to 18 C9 monomers. It is still ...
  56. [56]
    Structure of poly-C9 in complement membrane attack complex
    Feb 4, 2016 · 1a–d, Supplementary Figs 3–6). These data revealed a symmetrical assembly of 22 C9 monomers (Fig. 1a–c) that closely resembles the MAC4.
  57. [57]
    a review of the complement membrane attack complex - Journals
    Jun 19, 2017 · The membrane attack complex (MAC) is an important innate immune effector of the complement terminal pathway that forms cytotoxic pores on the surface of ...
  58. [58]
    p02748 · co9_human - UniProt
    Homooligomer; about 20 C9 chains oligomerize to give rise to a huge beta-barrel that forms a 100 Angstrom diameter pore in target membranes (PubMed:26841934, ...
  59. [59]
    Review Molecular cell biology of complement membrane attack
    The membrane attack complex (MAC) is the pore-forming toxin of the complement system, a relatively early evolutionary acquisition that confers upon complement ...
  60. [60]
    Complement membrane attack complexes activate noncanonical NF ...
    Jul 20, 2015 · Complement membrane attack complexes (MACs) promote inflammatory functions in endothelial cells (ECs) by stabilizing NF-κB–inducing kinase ...
  61. [61]
    The complement membrane attack complex triggers intracellular Ca ...
    Jul 1, 2013 · This is the first study that links non-lethal effects of sublytic MAC attack with inflammasome activation and provides a mechanism by which ...
  62. [62]
    The membrane attack complex as an inflammatory trigger - PubMed
    When assembly occurs on a target cell membrane the forming complex inserts into and through the bilayer to create a pore, the membrane attack complex (MAC). On ...
  63. [63]
    Structural implications for iC3b formation & function
    Complement-mediated opsonization, phagocytosis, and immune stimulation are critical processes in host defense and homeostasis, with the complement ...
  64. [64]
    Complement C4, C4A and C4a – What they do and how they differ
    Jun 11, 2024 · During this process, nascent C4b can bind covalently to surfaces and functions as an opsonin, which is to deposit on cell surfaces and make them ...1. Complement C4 Activation · Fig. 1 · 2. Complement C4, C4a And...
  65. [65]
    Complement Receptors and Their Role in Leukocyte Recruitment ...
    Feb 10, 2021 · ... 100 fold [reviewed in (Matthews et al., 2004)]. ... The importance of complement opsonization in phagocytosis has been demonstrated by a number of ...
  66. [66]
    Purified complement C3b triggers phagocytosis and activation ... - NIH
    Jan 6, 2023 · We show that purified C3b can potently trigger phagocytosis and killing of bacterial cells via Complement receptor 1.
  67. [67]
    The Role of the Anaphylatoxins in Health and Disease - PMC
    The anaphylatoxins (AT) C3a, C5a and C5a-desArg are generally considered pro-inflammatory polypeptides generated after proteolytic cleavage of C3 and C5 in ...
  68. [68]
    Complement - a key system for immune surveillance and homeostasis
    The functional repertoire of complement includes a major role in the resolution of inflammation through induction of regulatory T cells (Treg), non-inflammatory ...<|control11|><|separator|>
  69. [69]
    Complement in removal of the dead - balancing inflammation
    This triggers complement activation and opsonization of cells with fragments of C3b, which enhances phagocytosis and thus ensures silent removal.
  70. [70]
    Complement activation, regulation, and molecular basis for ...
    In humans, these are complement protein C1q, mannan‐binding lectin (MBL), collectin‐LK (CL‐LK), or the three ficolins L/M/H (also denoted ficolin‐1, ficolin‐2, ...
  71. [71]
    Factor H: A Complement Regulator in Health and Disease, and a ...
    Factor H is a major plasma regulator acting in the alternative complement pathway at the level of the central C3b component, facilitating C3b inactivation and ...
  72. [72]
    Regulation of Complement and Contact System Activation via C1 ...
    Oct 26, 2016 · The serpin C1 inhibitor (C1-INH) is the only regulator of classical complement activation as well as the major regulator of the contact system.
  73. [73]
    BIOLOGICAL ACTIVITIES OF C1 INHIBITOR - PMC - NIH
    Broadly speaking, C1 inhibitor plays important roles in the regulation of vascular permeability and in the suppression of inflammation.
  74. [74]
    Patterns of C1-Inhibitor/Plasma Serine Protease Complexes in ...
    May 5, 2020 · C1-INH formed the highest concentration complexes with C1r and C1s. We observed higher plasma kallikrein/C1-INH complex concentration in ...Purified Proteins And... · Protease/c1-Inh Complexes · Results
  75. [75]
    Hereditary angioedema: Linking complement regulation to the ...
    C1 inhibitor is not only a regulator of complement activity but also has a major role in the kallikrein‐kinin system, the contact system of coagulation, and ...
  76. [76]
    Proof of concept of a new plasma complement Factor H from waste ...
    Plasma exchange has been used to correct for FH deficiency and although the therapeutic potential of purified FH has been suggested by in vivo experiments in ...Abstract · Introduction · Materials and methods · Results
  77. [77]
    Complement factor H deficiency and endocapillary ... - Nature
    Jan 27, 2011 · Complement factor H (CFH) is a regulator of the alternative complement activation pathway. Mutations in the CFH gene are associated with ...
  78. [78]
    Complement factor I: Regulatory nexus, driver of immunopathology ...
    Complement factor I (FI) is the nexus for classical, lectin and alternative pathway complement regulation.
  79. [79]
    Complement factor I deficiency: a not so rare immune defect ...
    Jun 18, 2012 · Complete factor I deficiency is strongly associated with severe infections. Approximately 30 families with this deficiency have been described ...
  80. [80]
    Effect of rare coding variants in the CFI gene on Factor I expression ...
    ... complement factor I (CFI) gene on FI expression. We measured FI levels in ... The normal range was determined as mean FI ± 2 SD using of (36.4 ± 16.7 μg/ml) in ...<|control11|><|separator|>
  81. [81]
    Diverse functions of C4b-binding protein in health and disease - PMC
    Complement activation serves three primary functions in the control of infection. The first is opsonization, in which complement components coat the surface of ...
  82. [82]
    Complement Component C4 Binding Protein - ScienceDirect.com
    A single kindred with C4 binding protein deficiency has been described. The proband presented with angioedema, vasculitis, and arthritis. The manifestations ...
  83. [83]
    Membrane complement regulatory proteins - PubMed - NIH
    Membrane complement regulatory proteins, like DAF, MCP, CR1, and CD59, protect tissues from bystander injury when complement is activated.
  84. [84]
    Structure/function studies of human decay-accelerating factor - PMC
    The decay-accelerating factor (DAF or CD55) is a 70 000 MW membrane protein that protects cells from activation of autologous complement on their surfaces.
  85. [85]
    Structure and function of decay accelerating factor CD55 - PubMed
    Studies of decay-accelerating factor (DAF) function and structure are reviewed. DAF was first recognized as a species restricting factor operating at the ...
  86. [86]
    Membrane cofactor protein (MCP or CD46) - PubMed
    Membrane cofactor protein (MCP; CD46) is a widely distributed C3b/C4b-binding cell surface glycoprotein which serves as an inhibitor of complement activation ...
  87. [87]
    Dissecting sites important for complement regulatory activity in ...
    Dec 1, 2000 · Membrane cofactor protein (MCP; CD46), a widely distributed regulator of complement activation, is a cofactor for the factor I-mediated ...
  88. [88]
    Leucocyte complement receptor 1 (CR1/CD35) transcript and its ...
    Complement receptor 1 (CD35), also known as CR1, is a key complement regulatory protein that acts as a co-factor of factor I. CR1 accelerates the decay of C3 ...
  89. [89]
    Expression of the complement regulatory protein CD59 on human ...
    Protectin (CD59) is a complement regulatory protein which blocks the membrane attack complex during complement activation.
  90. [90]
    Levels of cell membrane CD59 regulate the extent of complement ...
    Normal and neoplastic cells are protected from autologous complement (C) attack by different cell-surface C-regulatory proteins including CD59 (protectin) ...
  91. [91]
    Mechanisms in Neisseria meningitidis for resistance against ...
    Here we discuss the mechanisms of meningococcal resistance against complement-mediated killing and the influence of both bacterial and host genetic factors.
  92. [92]
    Viral Evasion of the Complement System and Its Importance for ...
    Lectin Complement Pathway​​ The lectin pathway is activated in response to invading pathogens via direct binding of PAMPs by various C1q-like lectins, complexed ...
  93. [93]
    Complement Evasion: An Effective Strategy That Parasites Utilize to ...
    The complement system acts as the first line of immune defense to eliminate the invading parasites by forming the membrane attack complex (MAC) and promoting an ...
  94. [94]
    Interaction Between the Complement System and Infectious Agents
    Here we summarize the interactions between complement and infectious organisms, including bacteria, viruses, fungi, and parasites (Table 1), and discuss the ...
  95. [95]
    Role of C5a in inflammatory responses - PubMed - NIH
    C5a is a strong chemoattractant and is involved in the recruitment of inflammatory cells such as neutrophils, eosinophils, monocytes, and T lymphocytes.
  96. [96]
    Recognition of Microbial Glycans by Soluble Human Lectins - PMC
    Human innate immune lectins that recognize microbial glycans can conduct microbial surveillance and thereby help prevent infection.
  97. [97]
    Interaction of lectin pathway of complement-activating pattern ... - NIH
    Recognition resulted in activation of the lectin pathway (LP) of complement and an enhancement of phagocytosis (shown for M. tuberculosis). Although MBL may be ...
  98. [98]
    Evolution of the complement system: from defense of the single cell ...
    The complement system is an evolutionarily ancient component of immunity that revolves around the central component C3.
  99. [99]
    Complement Deficiency - StatPearls - NCBI Bookshelf
    [8] Deficiencies of the late common pathway (C5, C6, C7, C8, and C9) are associated with increased Neisseria infections, including Neisseria meningitidis and ...
  100. [100]
    Complement component 2 deficiency - Genetics - MedlinePlus
    Jun 1, 2014 · C2. Inheritance. Expand Section. This condition is inherited in an autosomal recessive pattern , which means both copies of the gene in each ...
  101. [101]
    The yin and the yang of early classical pathway complement disorders
    Loss-of-function variants in C1, C4, and C2 are most often associated with lupus and systemic infections with encapsulated bacteria. C3 deficiency varies ...
  102. [102]
    Terminal complement complexes with or without C9 potentiate ... - NIH
    In particular, deficiencies in C5 through C9 result in a 1,000-fold to 10,000-fold increased risk for invasive meningococcal disease by Neisseria meningitidis ...
  103. [103]
    The Brief Case: Meningococcemia Leading to a Diagnosis of ... - NIH
    Jan 30, 2019 · In particular, individuals with terminal complement deficiencies (C5 through C9, Fig. 2) have a 7,000- to 10,000-fold-higher risk of ...
  104. [104]
    Systemic Lupus Erythematosus and Deficiencies of Early ...
    Feb 24, 2016 · The deficiency of early complement proteins from the classical pathway (CP) is strongly associated with development of systemic lupus erythematous (SLE)
  105. [105]
    Meningococcal disease and the complement system - PubMed Central
    Complement activation on surfaces may be initiated through one or more of three pathways—the classical, lectin, or alternative pathways (Fig. 1). Table 1.
  106. [106]
    Properdin deficiency in a large Swiss family - PubMed Central
    Properdin deficiency was first described in a family with fulminant meningococcal infections [6]. Later studies have established a remarkably strong association ...
  107. [107]
    Infections of People with Complement Deficiencies and Patients ...
    Individuals with this disorder have a high risk of meningococcal disease (408), consistent with the observation that their serum shows decreased bactericidal ...
  108. [108]
    Comparison of Three Different Methods for Measuring Classical ...
    The standard 50% hemolytic complement (CH50) assay is the most commonly used method of screening patient sera for functional activity of the classical ...
  109. [109]
    Alternative Complement Pathway Activity (AH50) - ARUP Laboratories
    The Alternative Complement Pathway Activity (AH50) test is for initial screening of suspected deficiency in the alternative complement pathway.
  110. [110]
    Modern Complement Analysis - PMC - NIH
    In most cases, where total hemolytic activities (CH50 and AH50) indicate a complement deficiency, immunochemical assays can be performed as an alternative to ...<|separator|>
  111. [111]
    Complement Deficiency and Anticomplement Therapeutic ...
    Feb 3, 2025 · Complement testing, including total complement testing and testing for C3 and C4, may therefore be useful to support a diagnosis of SLE.
  112. [112]
    Functional Assays for Complement Regulators - PubMed
    MeSH terms · Animals · Biological Assay / methods* · Complement Activation · Complement Inactivator Proteins / analysis* · Humans. Substances. Complement ...
  113. [113]
    Functional Assessment - complement system - Svar Life Science
    Functional complement assays use ELISA to detect deficiencies, assess drug effects, and determine the activity of all three pathways separately.
  114. [114]
    [PDF] Eculizumab - accessdata.fda.gov
    Soliris is indicated for the treatment of patients with paroxysmal nocturnal hemoglobinuria (PNH) to reduce hemolysis. 1.2. Atypical Hemolytic Uremic Syndrome ( ...
  115. [115]
    Ravulizumab: First Global Approval - PubMed
    In December 2018, intravenous ravulizumab received its first global approval in the USA for the treatment of adults with PNH, and is under regulatory review.
  116. [116]
    FDA approves new treatment for adults with serious rare blood ...
    May 18, 2021 · FDA has approved Empaveli (pegcetacoplan) injection to treat adults with paroxysmal nocturnal hemoglobinuria (PNH), a rare, life-threatening blood disease.Missing: inhibitor | Show results with:inhibitor
  117. [117]
    Voydeya approved in the US as add-on therapy to ravulizumab or ...
    Apr 1, 2024 · Voydeya (danicopan) has been approved in the US as add-on therapy to ravulizumab or eculizumab for the treatment of extravascular haemolysis (EVH) in adults.
  118. [118]
    Functional expression of complement factor I following AAV ... - Nature
    Mar 10, 2021 · These encouraging pre-clinical studies have led to a first-in-human AAV gene therapy trial for late-stage AMD (NCT03846193). Material and ...Material And Methods · Codon Optimisation And... · C3b Cleavage AssayMissing: preclinical | Show results with:preclinical
  119. [119]
    Complement and microglia mediate early synapse loss in Alzheimer ...
    Mar 31, 2016 · Here we show in mouse models that complement and microglia mediate synaptic loss early in AD. C1q, the initiating protein of the classical ...Missing: visual seminal
  120. [120]
    The Complement System at the Embryo Implantation Site - NIH
    More specifically, C1q was found to play an important role in the replacement of DECs by endovascular trophoblasts migrating along the decidual spiral arteries ...
  121. [121]
    Emerging and Novel Functions of Complement Protein C1q - PMC
    Clearance by C1q of apoptotic bodies is regarded to be an important protective function, since its impairment is associated with renal, vascular, and other ...Role Of C1q In Pregnancy · Association Of C1q With... · Role Of C1q In The...<|control11|><|separator|>
  122. [122]
    The Role of Complement in Angiogenesis - PMC - PubMed Central
    Dec 1, 2020 · The complement system regulates physiological and pathological angiogenesis in models of wound healing, retinal regeneration, age-related macular degeneration, ...
  123. [123]
    Complement Activation and Inhibition in Wound Healing - PMC
    The complement system is activated by distinct carbohydrate and lipid residues on altered self-molecules and injured cells and the cellular inflammatory ...
  124. [124]
    Complement activation in obesity, insulin resistance, and type 2 ...
    In an aged population, complement C3 has been described as a strong marker of insulin resistance[42]. New studies have shown that adipose tissue can activate ...
  125. [125]
    Sphingolipid metabolism and complement signaling in cancer ...
    This forum article examines the evidence of sphingolipid metabolism and complement system crosstalk in inducing tumor cell survival, migration, and metastasis.Missing: post- | Show results with:post-
  126. [126]
  127. [127]
    Genomic view of the evolution of the complement system - PMC
    The initial phase of DNA level analysis revealed the presence of the complement genes in invertebrate deuterostomes such as sea urchins (Al-Sharif et al. 1998) ...