ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Regulators and receptors of the complement system

Regulators and receptors of the complement system
Literature review current through: Jan 2024.
This topic last updated: Mar 30, 2022.

INTRODUCTION — Precise control of the complement system is necessary because of its potent proinflammatory and cellular destructive capabilities. The regulation of the complement system is reviewed here. In order to comprehend the functions of the various regulatory proteins more fully, it is helpful to be familiar with the complement pathways, which are reviewed in greater detail separately (figure 1). (See "Complement pathways".)

COMPLEMENT REGULATION — Nearly one-half of all complement proteins serve a regulatory function [1-4]. The goal of regulation is to prevent complement damage to normal host tissue (inappropriate or wrong target) and fluid-phase activation (no target) [5]. Deficiencies of control proteins lead to excessive complement activation and significant morbidity and mortality. (See "Inherited disorders of the complement system", section on 'Abnormalities in regulatory proteins'.)

Regulatory proteins inhibit the system by destabilizing activation complexes and by mediating specific proteolysis of activation-derived fragments. The complement pathways are regulated at the following critical steps:

Activation (initiation)

Amplification (convertase formation)

Membrane attack (lysis)

The complement regulatory proteins, their location (in plasma or on cell membranes), function, miscellaneous comments, and associated diseases are summarized in the table (table 1).

Control of activation/initiation — In the classical pathway, C1 inhibitor (C1Inh) prevents excessive complement activation on a target, as well as in plasma (figure 1). C1 inhibitor, a member of the "serpin" superfamily of serine protease inhibitors, binds to each C1r and C1s subcomponent of the C1 complex (figure 2). This causes their dissociation and release from C1q, which is commonly attached to the Fc portion of immunoglobulin G (IgG) and immunoglobulin M (IgM) in an immune complex [6,7].

C1 inhibitor performs a similar function in the lectin pathway. This pathway has an activation scheme comparable to that of the classical complement pathway, but lectins (ie, proteins that bind to sugars) substitute for antibodies and lectin-associated proteases replace C1r and C1s. The lectins (specifically ficolins and collectins) bind sugar residues on microbial surfaces. Mannose-binding lectin-associated serine proteases (MASPs) subsequently cleave C4 and C2 (figure 1). (See "Complement pathways".)

Activation/initiation is controlled by C1 inhibitor that blocks the active sites of these MASPs, analogous to the classical pathway where it inhibits C1r and C1s [6,7]. (See "Inherited disorders of the complement system", section on 'Lectin pathway deficiencies'.)

C1 inhibitor has other biologic functions, in addition to control of the early steps in complement activation:

C1 inhibitor regulates three other interrelated pathways: the coagulation (contact), fibrinolytic, and kinin-generating systems. The role of C1 inhibitor in limiting the generation of kinins is central to the pathogenesis of hereditary angioedema, a condition caused by a deficit or dysfunction of C1 inhibitor. Hereditary angioedema is reviewed in detail elsewhere. (See "Hereditary angioedema (due to C1 inhibitor deficiency): Pathogenesis and diagnosis".)

In animals, treatment with purified C1 inhibitor improves survival in experimental models of bacterial- and endotoxin-induced septic shock [8,9]. The effect was also demonstrated with "inactivated" C1 inhibitor, suggesting that inhibition of the complement system was not the primary mechanism [8]. The use of C1 inhibitor therapy in human sepsis has been studied primarily in lab-based studies, anecdotal reports, and small, randomized trials in humans [10]. A possible role of complement in the pathophysiology of sepsis is discussed separately. (See "Pathophysiology of sepsis", section on 'Complement activation'.)

Control of amplification — The C3 convertases are powerful amplifiers of the complement system. The convertase steps are regulated by a family of complement-binding proteins (table 1) [1,11-16]:

Membrane proteins – Decay-accelerating factor (DAF; CD55), membrane cofactor protein (MCP; CD46), and CUB and sushi multiple domains protein 1 (CSMD1).

Plasma proteins – C4b binding protein (C4BP) regulates C4b and C4b-containing classical and lectin pathway convertases. Factor H regulates C3b, C3b-containing alternative pathway convertases, and C5 convertases that contain either one C3b (from the classical pathway/lectin pathway) or two C3bs (alternative pathway).

These proteins function in three ways (figure 3):

By preventing formation of the convertases.

By disassembling or disassociating the convertases (known as decay-accelerating activity [DAA]).

By limited proteolytic cleavage of C4b and/or C3b. This process, called cofactor activity, requires collaboration between the plasma-serine protease known as factor I and a cofactor protein, such as MCP or factor H.

These widely expressed membrane regulators inhibit complement on host tissue, while the plasma inhibitors primarily prevent activation in the fluid phase. However, at sites of injury, the plasma inhibitors can interact with structures, such as the exposed basement membrane. For example, anionic or heparin-binding sites in factor H and C4BP allow those plasma regulators, in essence, to act like a membrane protein at inflammatory sites and in areas of cellular injury (eg, apoptotic and necrotic cells) [17]. Regulatory proteins are a target of pathogen inactivation or highjacking [18]. Indeed, CD46 has been called a pathogen magnet [19].

Deficiencies in regulators — Regulator deficiencies are associated with the following disorders:

Age-related macular degeneration [20]

Atypical hemolytic uremic syndrome [21,22] (see "Overview of hemolytic uremic syndrome in children")

C3 glomerulopathy (C3G; formerly dense deposit disease) [23] (see "C3 glomerulopathies: Dense deposit disease and C3 glomerulonephritis")

CD55 deficiency with hyperactivation of complement, angiopathic thrombosis, and protein-losing enteropathy (CHAPLE) syndrome [24] (see "Inherited disorders of the complement system")

Control of membrane attack — The membrane attack complex (MAC) is also regulated both in the fluid phase and on cells [25,26]. Control in plasma prevents diffusion from the activation site. S protein (also known as vitronectin) controls fluid-phase MAC by binding to a site on the C5b-7 complex, thereby preventing its attachment to cell membranes. MACs that deposit on self-tissue are inhibited by CD59 (also called protectin, membrane inhibitor of reactive lysis, or MAC inhibitory factor). This widely expressed glycolipid-anchored membrane protein has binding sites for both C8 and C9 and thereby inhibits the final steps of MAC assembly.

A deficiency of CD59 and DAF is the pathophysiologic basis of paroxysmal nocturnal hemoglobinuria (PNH). The deficiency is caused by a mutation in the PIGA gene (phosphatidylinositol glycan anchor biosynthesis, class A) that prevents the formation of a glycosylphosphatidylinositol anchor, such that CD59 and DAF are not expressed on the cell surface. This results in an increased sensitivity to lysis. Additionally, several cases with only loss-of-function CD59 deficiency have been described with all such patients (12 of 12) demonstrating neurologic symptoms, 92 percent (11 of 12) recurrent peripheral neuropathy, 50 percent (6 of 12) with recurrent strokes, and 8 percent (1 of 12) with retinal involvement [27]. (See "Pathogenesis of paroxysmal nocturnal hemoglobinuria", section on 'PIGA gene mutation'.)  

Control of anaphylatoxins — When complement proteins C3, C4, and C5 are activated, small peptides of 74 to 77 amino acids in length (C3a, C4a, and C5a) are released from the amino-terminus of the alpha chain and bind to their cognate receptors or are inactivated by a plasma enzymes (carboxypeptidase-N and carboxypeptidase-R) that remove the carboxyl-terminal arginyl residue [28-30].

COMPLEMENT RECEPTORS — Many host cells, especially human peripheral blood cells, possess receptors for complement activation fragments that promote the adherence and ingestion of microorganisms and immune complexes (table 2). Upon engagement, these receptors, which are expressed on most inflammatory and immunocompetent cells, induce cellular responses that trigger inflammatory and immune responses [6,12,31].

Complement receptor 1 — Most peripheral blood cells express complement receptor 1 (also called CR1, CD35, C3b/C4b receptor, and immune adherence receptor) [32]. CR1 plays a critical role in the clearance of C4b- and C3b-coated particles (eg, immune complexes) on which the complement system has been activated. Deposition of C3b and C4b on a target is particularly efficacious in promoting attachment to cells bearing complement receptors. This phenomenon is known as "immune adherence."

Erythrocytes express approximately 500 copies of CR1 per cell. This allows the erythrocyte to bind intravascular immune complexes and then serve as a vehicle (taxi) to transport immune complexes to the liver and spleen. Hepatic and splenic macrophages then "clear" such immune complexes by "stripping" them from the erythrocyte, destroying the antigen (often viruses or bacteria), and facilitating an immune response (antigen presentation). The erythrocyte may then return to the circulation for another round of immune complex clearance.

The function of CR1 depends in part upon the type of cell on which it is expressed. The primary function of CR1 on erythrocytes is to clear circulating immune complexes. In comparison, CR1 on neutrophils and monocytes binds C3b- and C4b-bearing immune complexes, resulting in a cellular response that often includes internalization and digestion. In addition, CR1 is expressed on B lymphocytes, a subset of T lymphocytes, and on follicular dendritic cells where it facilitates the localization of complement-bearing antigens to lymphoid follicles. On peripheral blood cells, CR1 is primarily an immune adherence receptor. However, in a highly inflammatory milieu with engagement of C3aR and C5aR, CR1 becomes capable of ingesting/internalizing such coated particles. It also serves a regulatory role by preventing further complement activation and by converting C3b to hemolytically inactive fragments (iC3b and C3dg), which then serve as ligands for additional complement receptors.

CR1 is a receptor for the malaria parasite [33,34]. It also has been implicated as a risk factor for Alzheimer disease and schizophrenia [35-37]. Levels of CR1 are reduced in diseases, such as systemic lupus erythematosus (SLE), glomerulonephritides, and human immunodeficiency virus (HIV) [32]. These diseases feature immune complexes that may reduce levels of CR1, and in the case of SLE, such reductions parallel disease activity [32]. Additionally, a study found that CR1 protein levels and genetic variants were associated with chronic Chagas disease in a Brazilian cohort [34].

Complement receptor 2 — Complement receptor 2 (also called CR2, C3d or C3dg receptor, and CD21) is expressed on B lymphocytes, follicular dendritic cells, epithelial cells in the pharynx and upper airway, and in low amounts on peripheral blood T cells. It is not found on monocytes, granulocytes, or erythrocytes.

CR2 serves to localize complement-bearing immune complexes to B lymphocyte-rich areas of the spleen and lymph nodes. In this way, CR2, as well as CR1, promotes antigen-driven activation of B cells. This "adjuvant" function of the complement system is being exploited by attaching C3 fragments to vaccines in order to enhance their antigenicity. CR2, through its association with other membrane proteins, is also an important coreceptor in the signaling of B cells [1,38]. CR2 is also the receptor for the Epstein-Barr virus (EBV). (See "Virology of Epstein-Barr virus".)

Patients with the rare heritable disorder X-linked agammaglobulinemia lack mature B lymphocytes. Consequently, EBV cannot infect the B cells of these patients [39,40]. (See "Agammaglobulinemia".)

Complement receptor 3 — Complement receptor 3 (also called CR3, Mac-1, and CD11b/18) binds and promotes the opsonization of particles bearing fragments of C3, especially iC3b [41]. CR3 is expressed by macrophages/monocytes and certain lymphocytes and is part of the integrin family of adherence-promoting proteins. (See "Leukocyte-endothelial adhesion in the pathogenesis of inflammation".)

CR3 promotes ingestion of iC3b-coated targets. Deficiency of CR3 is associated with delayed separation of the umbilical cord, omphalitis, and severe childhood infections. This disorder is known as leukocyte-adhesion deficiency syndrome. (See "Leukocyte-adhesion deficiency".)

Since immune complexes possess variable quantities of C3-derived fragments (C3b, iC3b, C3dg, and C3d), multiple complement receptors usually cooperate to help clear immune complexes and facilitate adaptive immune responses to antigens. For example, CR1 promotes initial adherence, CR3 facilitates internalization, and CR2 transmits cellular signals to facilitate the adaptive immune response [41].

Complement receptor 4 — Complement receptor 4 (CR4, CD11c/18) has a similar function and tissue distribution as CR3. However, CR4 may also play an important role in neutrophil and monocyte adhesion to endothelium [41]. (See "Leukocyte-endothelial adhesion in the pathogenesis of inflammation".)

Complement receptor of the immunoglobulin superfamily — Complement receptor of the immunoglobulin superfamily (CRIg) recognizes C3b and iC3b molecules covalently bound to particle (eg, pathogen) surfaces [12,13]. Functions of CRIg include inhibition of the alternative pathway, clearance of systemic pathogens, and regulation of the adaptive immune response. CRIg is widely expressed in lung, adipose tissue, spleen, adrenal gland, small intestine, bladder, colon, breast, and on macrophages associated with blood vessels. CRIg is the only complement receptor described thus far with immunoglobulin domains. No mutations in CRIg have been linked to disease [42].

Receptors for C5a and C3a — The complement system, which can be engaged in a few seconds, is likely the earliest response system to microbes and to tissue damage. Liberation of C3a and C5a and interaction with their respective receptors triggers the acute inflammatory reaction. C5a is a major chemotactic factor for neutrophils. Both C3a and C5a "activate" a wide variety of cell types.

C5a receptor — Two C5a receptors have been identified, C5aR1 and C5aR2. Both belong to the G protein-coupled receptor (GPCR) family [43]. C5aR1 is better defined, while C5aR2 is less well-understood and may be a decoy receptor for C5a [44]. C5aR1 is prominently expressed on neutrophils, macrophages, mast cells, and basophils. Also, it is a chemotactic factor for neutrophils and monocytes and causes release of their granular constituents. C5aR1 is expressed on a wide variety of epithelial and endothelial cells.

C5a has a spasmogenic effect upon various tissues by a direct action on smooth muscle cells bearing C5a receptors (C5aR) or secondarily by the release of mediators from mast cells [45].

Receptor binding of C5a may play a role in end-organ damage during sepsis [46]. This was illustrated in an animal model of intra-abdominal infection in which C5aR1 upregulation was found in lung, liver, kidney, and heart soon after onset of sepsis [47]. Administration of antibodies that blocked activation of C5a receptor was associated with improved survival. Another study comparing healthy volunteers to patients in septic shock suggested that septic shock in humans is associated with complement activation, C-reactive protein-dependent loss of C5aR on neutrophils, and appearance of a circulating C5aR in serum, which correlated with poor outcome [48]. (See "Pathophysiology of sepsis", section on 'Complement activation'.)

The binding of C5a to its receptor may also provide a mechanism by which the complement- and immunoglobulin-activated inflammatory systems interact. In a mouse model, binding of C5a to its receptor resulted in upregulation of Fc-gamma receptors. Binding of immune complexes to Fc-gamma receptors, in turn, leads to generation of more C5a, thus establishing a positive feedback loop [49]. This type of interaction also may be important in the pathogenesis of autoimmune diseases mediated by autoantibodies and immune complexes [50].

Note that in late 2021 the US Food and Drug Administration (FDA) approved the use of a small molecule inhibitor, avacopan, an inhibitor of C5aR, for the treatment of anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitis. (See "Granulomatosis with polyangiitis and microscopic polyangiitis: Induction and maintenance therapy", section on 'Induction therapy'.)

C3a receptor — The C3a receptor's major role appears to be in activating cells at sites of inflammation [51]. It is a member of the same G-coupled superfamily as the C5a receptor and has a broad tissue distribution. Receptors for C3a are present on endothelial, epithelial, and many types of peripheral blood cells, including neutrophils, monocytes, most lymphocytes, and basophils. Mast cell and basophil degranulation (similar to the C5a receptor) is mediated by C3a engaging its receptor on these cell types. Variations in this receptor may affect susceptibility to asthma [52,53].

CSMD1 — CUB and sushi domains protein 1 (CSMD1) is a transmembrane protein expressed in multiple tissues that serves as a cofactor in the factor I-mediated cleavage of C3b [54]. Four isoforms have been described. Changes in CSMD1 expression have been associated with several types of cancers, infertility, and disorders of cognitive function [55,56].

Other receptors — Receptors for C1q [57], factor H [58], ficolins [59], and others have been noted on such cells as neutrophils, monocytes, and B cells. Their function is not as clearly defined as those described above. However, a protein, such as C1q, may serve both as a lectin to identify foreign materials and altered self as well as the link to proteases of the complement pathway. Additionally, protease-activated receptors 1 and 4 have been identified as receptors for the C4a anaphylatoxin [60].

SUMMARY

Importance of complement regulation – Nearly one-half of all complement proteins serve a regulatory function. Complement pathways are regulated at each important step: activation, amplification (convertase formation), and membrane attack (table 1). Precise control of the complement system is necessary because of its potent proinflammatory and cellular destructive capabilities. The goal of regulation is to minimize complement damage at sites of inflammation (inappropriate or wrong target) and fluid-phase activation (no target). (See 'Complement regulation' above.)

Complement receptors – Receptors for complement activation fragments are expressed on many host cells, including peripheral blood cells, endothelial cells, and epithelial cells (table 2). Receptors for C4b and C3b are present on most cells of the immune system and promote pathogen destruction and generation of the adaptive immune response. Receptors for C3a and C5a are widely distributed where they trigger the local inflammatory response (innate immunity) and also cell activation to prepare for the adaptive immune response. Together, these receptors promote the adherence and ingestion of microorganisms and immune complexes. (See 'Complement receptors' above.)

Deficiencies – Deficiencies of inhibitory proteins lead to excessive complement activation and significant morbidity and mortality (See 'Deficiencies in regulators' above.). Deficiencies of certain complement receptors, such as complement receptor 3 (CR3), result in severe infections in childhood (See "Inherited disorders of the complement system" and "Leukocyte-adhesion deficiency".)

ACKNOWLEDGMENT — The UpToDate editorial staff acknowledges E Richard Stiehm, MD, who contributed as a Section Editor to earlier versions of this topic review.

  1. Zipfel PF, Skerka C. Complement regulators and inhibitory proteins. Nat Rev Immunol 2009; 9:729.
  2. Dunkelberger JR, Song WC. Complement and its role in innate and adaptive immune responses. Cell Res 2010; 20:34.
  3. Lesher AM, Song WC. Review: Complement and its regulatory proteins in kidney diseases. Nephrology (Carlton) 2010; 15:663.
  4. Bajic G, Degn SE, Thiel S, Andersen GR. Complement activation, regulation, and molecular basis for complement-related diseases. EMBO J 2015; 34:2735.
  5. Sjöberg AP, Trouw LA, Blom AM. Complement activation and inhibition: a delicate balance. Trends Immunol 2009; 30:83.
  6. Ricklin D, Hajishengallis G, Yang K, Lambris JD. Complement: a key system for immune surveillance and homeostasis. Nat Immunol 2010; 11:785.
  7. Gaboriaud C, Ling WL, Thielens NM, et al. Deciphering the fine details of C1 assembly and activation mechanisms: "mission impossible"? Front Immunol 2014; 5:565.
  8. Liu D, Lu F, Qin G, et al. C1 inhibitor-mediated protection from sepsis. J Immunol 2007; 179:3966.
  9. Igonin AA, Protsenko DN, Galstyan GM, et al. C1-esterase inhibitor infusion increases survival rates for patients with sepsis*. Crit Care Med 2012; 40:770.
  10. Singer M, Jones AM. Bench-to-bedside review: the role of C1-esterase inhibitor in sepsis and other critical illnesses. Crit Care 2011; 15:203.
  11. Liszewski MK, Atkinson JP. Complement regulators in human disease: lessons from modern genetics. J Intern Med 2015; 277:294.
  12. Holers VM. Complement and its receptors: new insights into human disease. Annu Rev Immunol 2014; 32:433.
  13. Zeng Z, Surewaard BG, Wong CH, et al. CRIg Functions as a Macrophage Pattern Recognition Receptor to Directly Bind and Capture Blood-Borne Gram-Positive Bacteria. Cell Host Microbe 2016; 20:99.
  14. Irvine KM, Banh X, Gadd VL, et al. CRIg-expressing peritoneal macrophages are associated with disease severity in patients with cirrhosis and ascites. JCI Insight 2016; 1:e86914.
  15. Escudero-Esparza A, Kalchishkova N, Kurbasic E, et al. The novel complement inhibitor human CUB and Sushi multiple domains 1 (CSMD1) protein promotes factor I-mediated degradation of C4b and C3b and inhibits the membrane attack complex assembly. FASEB J 2013; 27:5083.
  16. Reis ES, Mastellos DC, Hajishengallis G, Lambris JD. New insights into the immune functions of complement. Nat Rev Immunol 2019; 19:503.
  17. Gullstrand B, Mårtensson U, Sturfelt G, et al. Complement classical pathway components are all important in clearance of apoptotic and secondary necrotic cells. Clin Exp Immunol 2009; 156:303.
  18. Hovingh ES, van den Broek B, Jongerius I. Hijacking Complement Regulatory Proteins for Bacterial Immune Evasion. Front Microbiol 2016; 7:2004.
  19. Liszewski MK, Atkinson JP. Complement regulator CD46: genetic variants and disease associations. Hum Genomics 2015; 9:7.
  20. Armento A, Ueffing M, Clark SJ. The complement system in age-related macular degeneration. Cell Mol Life Sci 2021; 78:4487.
  21. Fakhouri F, Frémeaux-Bacchi V. Thrombotic microangiopathy in aHUS and beyond: clinical clues from complement genetics. Nat Rev Nephrol 2021; 17:543.
  22. Wong EKS, Kavanagh D. Diseases of complement dysregulation-an overview. Semin Immunopathol 2018; 40:49.
  23. Smith RJH, Appel GB, Blom AM, et al. C3 glomerulopathy - understanding a rare complement-driven renal disease. Nat Rev Nephrol 2019; 15:129.
  24. Ozen A, Comrie WA, Ardy RC, et al. CD55 Deficiency, Early-Onset Protein-Losing Enteropathy, and Thrombosis. N Engl J Med 2017; 377:52.
  25. Serna M, Giles JL, Morgan BP, Bubeck D. Structural basis of complement membrane attack complex formation. Nat Commun 2016; 7:10587.
  26. Morgan BP, Boyd C, Bubeck D. Molecular cell biology of complement membrane attack. Semin Cell Dev Biol 2017; 72:124.
  27. Tabib A, Karbian N, Mevorach D. Demyelination, strokes, and eculizumab: Lessons from the congenital CD59 gene mutations. Mol Immunol 2017; 89:69.
  28. Haas PJ, van Strijp J. Anaphylatoxins: their role in bacterial infection and inflammation. Immunol Res 2007; 37:161.
  29. Matthews KW, Mueller-Ortiz SL, Wetsel RA. Carboxypeptidase N: a pleiotropic regulator of inflammation. Mol Immunol 2004; 40:785.
  30. Klos A, Tenner AJ, Johswich KO, et al. The role of the anaphylatoxins in health and disease. Mol Immunol 2009; 46:2753.
  31. Kemper C, Köhl J. Novel roles for complement receptors in T cell regulation and beyond. Mol Immunol 2013; 56:181.
  32. Khera R, Das N. Complement receptor 1: disease associations and therapeutic implications. Mol Immunol 2009; 46:761.
  33. Stoute JA. Complement receptor 1 and malaria. Cell Microbiol 2011; 13:1441.
  34. Sandri TL, Lidani KCF, Andrade FA, et al. Human complement receptor type 1 (CR1) protein levels and genetic variants in chronic Chagas Disease. Sci Rep 2018; 8:526.
  35. Zhu XC, Yu JT, Jiang T, et al. CR1 in Alzheimer's disease. Mol Neurobiol 2015; 51:753.
  36. Fonseca MI, Chu S, Pierce AL, et al. Analysis of the putative role of CR1 in Alzheimer's disease: Genetic association, expression and function. PLoS One 2016; 11:e0149792.
  37. Arakelyan A, Zakharyan R, Khoyetsyan A, et al. Functional characterization of the complement receptor type 1 and its circulating ligands in patients with schizophrenia. BMC Clin Pathol 2011; 11:10.
  38. Erdei A, Füst G, Gergely J. The role of C3 in the immune response. Immunol Today 1991; 12:332.
  39. Wentink MW, Lambeck AJ, van Zelm MC, et al. CD21 and CD19 deficiency: Two defects in the same complex leading to different disease modalities. Clin Immunol 2015; 161:120.
  40. Thiel J, Kimmig L, Salzer U, et al. Genetic CD21 deficiency is associated with hypogammaglobulinemia. J Allergy Clin Immunol 2012; 129:801.
  41. Vorup-Jensen T, Jensen RK. Structural immunology of complement receptors 3 and 4. Front Immunol 2018; 9:2716.
  42. Liu G, Fu Y, Yosri M, et al. CRIg plays an essential role in intravascular clearance of bloodborne parasites by interacting with complement. Proc Natl Acad Sci U S A 2019; 116:24214.
  43. Hawksworth OA, Li XX, Coulthard LG, et al. New concepts on the therapeutic control of complement anaphylatoxin receptors. Mol Immunol 2017; 89:36.
  44. Kemper C. Targeting the Dark Horse of complement: the first generation of functionally selective C5aR2 ligands. Immunol Cell Biol 2016; 94:717.
  45. Sarma JV, Ward PA. New developments in C5a receptor signaling. Cell Health Cytoskelet 2012; 4:73.
  46. Ward PA. The dark side of C5a in sepsis. Nat Rev Immunol 2004; 4:133.
  47. Riedemann NC, Guo RF, Neff TA, et al. Increased C5a receptor expression in sepsis. J Clin Invest 2002; 110:101.
  48. Unnewehr H, Rittirsch D, Sarma JV, et al. Changes and regulation of the C5a receptor on neutrophils during septic shock in humans. J Immunol 2013; 190:4215.
  49. Kumar V, Ali SR, Konrad S, et al. Cell-derived anaphylatoxins as key mediators of antibody-dependent type II autoimmunity in mice. J Clin Invest 2006; 116:512.
  50. Atkinson JP. C5a and Fcgamma receptors: a mutual admiration society. J Clin Invest 2006; 116:304.
  51. Boos L, Campbell IL, Ames R, et al. Deletion of the complement anaphylatoxin C3a receptor attenuates, whereas ectopic expression of C3a in the brain exacerbates, experimental autoimmune encephalomyelitis. J Immunol 2004; 173:4708.
  52. Hasegawa K, Tamari M, Shao C, et al. Variations in the C3, C3a receptor, and C5 genes affect susceptibility to bronchial asthma. Hum Genet 2004; 115:295.
  53. Laumonnier Y, Wiese AV, Figge J, Karsten C. Regulation and function of anaphylatoxins and their receptors in allergic asthma. Mol Immunol 2017; 84:51.
  54. Kraus DM, Elliott GS, Chute H, et al. CSMD1 is a novel multiple domain complement-regulatory protein highly expressed in the central nervous system and epithelial tissues. J Immunol 2006; 176:4419.
  55. Lee AS, Rusch J, Lima AC, et al. Rare mutations in the complement regulatory gene CSMD1 are associated with male and female infertility. Nat Commun 2019; 10:4626.
  56. Athanasiu L, Giddaluru S, Fernandes C, et al. A genetic association study of CSMD1 and CSMD2 with cognitive function. Brain Behav Immun 2017; 61:209.
  57. Ghebrehiwet B, Hosszu KK, Valentino A, et al. Monocyte expressed macromolecular C1 and C1q receptors as molecular sensors of danger: Implications in SLE. Front Immunol 2014; 5:278.
  58. Losse J, Zipfel PF, Józsi M. Factor H and factor H-related protein 1 bind to human neutrophils via complement receptor 3, mediate attachment to Candida albicans, and enhance neutrophil antimicrobial activity. J Immunol 2010; 184:912.
  59. Kuraya M, Ming Z, Liu X, et al. Specific binding of L-ficolin and H-ficolin to apoptotic cells leads to complement activation. Immunobiology 2005; 209:689.
  60. Wang H, Ricklin D, Lambris JD. Complement-activation fragment C4a mediates effector functions by binding as untethered agonist to protease-activated receptors 1 and 4. Proc Natl Acad Sci U S A 2017; 114:10948.
Topic 3967 Version 19.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟