review article

The Role of Complement in Alcoholic Liver Disease

Chengjie Lin1,2,3, Zhigao Hu1,2,3, Biao Lei2, Bo Tang2,Junfei Jin2, Songqing He1,2*

1Department of Hepatopancreatobiliary Surgery, the first affiliated hospital of Guangxi Medical University, China.

2Departmentof Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, P.R. China; Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Guilin Medical University, P.R. China.

3Departmentof General Surgery, Xiangya Hospital, Central South University, China.

Authorship Note:Chengjie Lin and Zhigao Hu contributed equally to this work.
*Corresponding Author: Dr. Songqing He, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, P.R. China. Tel:+86773-2809503; E-mail: hesongqing@aliyun.com.


Received Date: 29 December, 2016; Accepted Date: 16 January, 2017; Published Date:23 January, 2017

Citation: Lin C, Hu Z, Lei B, Tang B, Jin J, He S (2017) TheRole of Complement in Alcoholic Liver Disease. . J Dig Dis Hepatol 2016: JDDH-123.

The complement system is a key component of the body’s immune system. When abnormally activated, the system can induce inflammation and damage to normal body tissues and participate in the development and progression of a variety of diseases. In the past, most scholars believed that alcoholic liver disease(ALD) is due to the stress of ethanol on liver cells, including oxidative stress and dysfunction of mitochondria and protease bodies, causing hepatocyte injury and apoptosis.Recently, studies have shown that complement activation is also involved in the genesis and development of ALD.This article reviews the roles of complement activation in ALD and of therapeutic intervention in complement-activation pathways. We intend to provide new ideas on the diagnosis and treatment of ALD.

Introduction

Liver disease caused by long-term excessive ethanol drinking is a major cause of chronic liver disease. As the global incidence of alcoholic liver disease (ALD) increases year by year, it has become a serious threat to human health. Almost all heavy drinkers have fatty liver, 10%–20% of which develop into alcoholic hepatitis, cirrhosis, and even hepatocellular carcinoma  Exploration of the mechanisms of alcohol-induced liver injury and repair is extremely important in developing methods for preventing and treating ALD.

The complement system plays an important beneficial role in the immune apparatus: complement activation promotes target-cell lysis, with the associated elimination of exogenous pathogens. Yet, the complement system is a "double-edged sword", as the excessive activation of complement can induce inflammation and lead to autoimmune diseases, such as autoimmune kidney disease, glomerular nephritis, acute lung injury, and others . Most plasma complement components are synthesized in liver cells. Thus, the liver becomes the main target of damage by complement activation. Several studies have illustrated that complement activation is involved in the development of ALD 

Metabolic pathways of ethanol

 and most is metabolized by alcohol dehydrogenase and aldehyde dehydrogenase to form acetic acid, which can be used as substrate in the tricarboxylic acid cycle to produce energy. With excessive drinking, the body can activate another metabolic pathway, i.e., the microsomal ethanol oxidation system (MEOS), which catalyzes ethanol mainlyby cytochrome P450 2El (CYP2E1) in Kupffer cells. The MEOS can lead to the overproduction of reactive oxygen species and reactive nitrogen species, which may exceed the body’s antioxidant capacity. Free radicals produced via the MEOS pathway promote a series of toxic effects: membrane-lipid peroxidation, intracellular protease degeneration, oxidative modification of DNA, and others, which eventually lead to necrosis or apoptosis of hepatocytes .A small percentage of ethanol is metabolized by fatty acid ethyl ester synthase to produce fatty acid ethyl ester through the non-oxidative pathway.Fatty acid ethyl ester has cytotoxicity, which can further injure the liver and pancreas[16]. Thus the liver becomes the main organ damaged by ethanol. Chronic ethanol exposure results in decreased protease activityin liver cells, imbalanceof the liver’sdetoxification function, and overproduction of acetaldehyde, thus inducing hepatic oxidative stress and complement activation; all these activities can injure hepatocytes [17].

Complement activation pathway

Most complement proteins are synthesized in liver cells; only a small portion is synthesized in endothelial cells, intestinal epithelial cells, and glomerular cells.

Complement is activated by three pathways: the classical, mannan-binding lectin (MBL), and alternative pathways. The three pathways start with differentmechanisms, but they end with a common terminal pathway, asshown in Fig.1. The classical pathway is the main mechanism of immune responses. In it, C1q identifies immune complexes, followed by the activation of C1r and C1s. ActivatedC1scleaves C2 and C4 to form C3 convertase (C4bC2a), which cleaves C3 to form C5 convertase (C4bC2aC3b). In contrastto activation of the classical pathway, activation of the lectin pathway does not depend on immune complexes. In this pathway, the cascade ofenzymatic reactions proceeds in this sequence: MBL identifies the pathogens to form MBL-associated serine proteases (MASP1, MASP2); MASP1 directly cleaves C3 to form C3 convertase (C3bBb), which then is involved in the positive feedback loop of the alternative pathway. MASP2cleaves C4 and C2 in a manner similar to that of C1s, forming C3 convertase(C4bC2a), which continues to cleave C5 to form C5 convertase(C4bC2aC3b). Thus, this pathway can cross-promote the classical and alternative pathways.The alternative pathway is activated with C3, factor B and Factor D, the activation of which is also independent of immune complexes, and participates in the defense mechanisms of the early stage of inflammation The above three pathways  merge into the terminal pathway, in which C5 convertase cleaves C5 to form C5a and C5b, and C5b combines with C6, C7, C8 and C9 to form the membrane attack complex (MAC). Formation of the MAC leads to cell lysis and induces cells to release inflammatory cytokines.

Complement activation in alcoholic liver diseaseComplement activation in alcoholic fatty liver disease

but its pathogenesis is not clearly defined. Abnormal activation of complement reportedly enhances the sensitivity of the liver to fat, which leads to the development of fatty liver Järveläinen, et al.[6] found that deposition of complement C1, C3, and C8 was increased,and the expression of membrane-binding proteins complement receptor 1-related protein y(Crry) and CD59 was decreased in the liver cells of a mouse ALD model. These findings are evidence that alcohol-induced complement activation can result in ALD, at least in an experimental model.In other studies in mice chronically exposed to ethanol,Cohen, et al. found that lipid deposition in liver cells as well as values of liver-related serum enzymes (alanine aminotransferase and aspartate amino transferase) increased significantly; various degrees of liver cell apoptosis were also found. Moreover, with knock out of the C1q gene, hepatic steatosisin the mice was significantly decreased  This study illustrated that complement activation could be associated with ethanol-induced hepatic steatosis.

Bykov, et al.  fed C3+/+ and C3-/- mice a high fat and high alcohol diet, respectively. The authors found that hepatic steatosis and significant increases in triglyceride values developed in the C3+/+mice,whereas C3-/-mice were protected from ethanol-induced liver injury;research by Stewart,et al. [5] yielded similar results. Thus, activation of complement C1 and C3 appears to play a significant role in promoting fatty infiltration in the liver. Further definition of the relationship between activated complement C1, C3 and lipid metabolism in the liver may aid in the development of measures for intervention and treatment of alcoholic fatty liver disease.Besides C1 and C3,complement C5 also is involved in lipid metabolism.Bavia, et al. found the activation of complement C5 by high-dose ethanol exposure can affect the distribution of lipid in liver cells and serum: Less lipid and cholesterol is deposited in hepatocytes of C5- mice than  in hepatocytes of C5+mice, and values of IL-17, which is involved in the synthesis and metabolism of lipid and cholesterol,are  higher in C5- mice than  in C5+mice The above-cited reports indicate that activation of C5 likely plays a role in the development of alcoholic fatty liver.

Complement activation in alcoholic hepatitis

Cohen, et al.  found that long-term alcohol exposure can lead to apoptosis of liver cells, and the degree of apoptosis is positively correlated with liver injury. However,whether short-term alcohol exposure can cause hepatocyte apoptosis was not known. Further research resolved this issue: Short-term alcohol exposure did not cause hepatocyte apoptosis, but it did promote the deposition of complement C3b and the expression of inflammatory cytokines (tumor necrosis factor and IL6). After theCq gene was knocked out, the expression of inflammatory cytokines was significantly reduced compared to that in wild-type animals Experiments by Païdassi, et al. [31] and Lu, et al. [32] supported these observations.

Complement C5, a core component of the complement activation pathway,is involved in the occurrence and development of alcoholic hepatitis, in addition to fatty liver .Bavia, et al.[33]documented thisina hepatitis model induced by alcohol; they found that values of pro-inflammatory cytokines(IL-6, IFN-γ, IL-1β, and others) in B6C5+ mice were significantly higher than those in B6C5- mice, and anti-inflammatory factors (IL10 and IL17) were secreted significantly more in B6C5- mice. These findings illustrated that activated C5 induced the expression of proinflammatory cytokines after alcohol exposure. Up-regulated expression of pro-inflammatory cytokines(IL-6, IFN-γ, IL-1β, and others) aids the body’sdefense against pathogenic microorganisms, but it also participates in the pathogenesis of alcoholic fatty liver and alcoholic hepatitis 

Complement activation in alcoholic hepatic fibrosis

 In recent years, the pathogenesis of alcoholic hepatic fibrosis has attracted wide attention internationally, but the cause of the fibrosis is still not fully defined .According to published reports, complement C3, C4 and activation of the MBL pathway are involved in the development of fibrosis. Bavia, et al.,[33] using the mouse model of ALD, found that values of TGF-β,which promotes hepatic fibrosis, were significantly higher in B6C5+ mice than in B6C5- mice Hillebradt, et al. found the C5 gene involved in the regulation of hepatic fibrosis on human chromosome, and further study found that C5- mice have decreased hepatic fibrosis. Thus, the evidence indicates that activation of complement C5 likely promotes hepatic fibrosis. Exploration of the relationship between complement activation and alcoholic hepatic fibrosis, and of possible intervention in ALD by reversing the progression of hepatic fibrosis in its early stage, seems worthwhile goals. 

Complement-induced Kupffer cells and alcoholic liver disease

Ethanol-induced activation of complement component C1q at the early stage of ALD promotes the release of inflammatory cytokines from Kupffer cells, which further promote alcoholic liver injury  Furthermore,Kupffer cells can express C3R and C5R,then induce prostaglandin release and synthesis of pro-inflammatory cytokines .However,in certain pathological conditions,activated C5 combines with C5R, inducing the upregulation of fibrinogenon Kupffer cells, an interaction that is believed to lead to hepatic fibrosis In addition,alcohol-induced upregulation of CD14 leads to Kupffer cells combining with lipopolysaccharide, which induces liver damage through the activation of TLR4 in Kupffer cells and inflammatory signaling pathways; these events can further aid in the development of hepatic fibrosis or cirrhosis [58]. Thus, Kupffer cells seem to be extensively involved in the development of ALD 

Complement regulation in alcoholic liver disease

Deficiency of CD55/DAF and complement regulatory factors aggravate liver injury  whereas FactorH can control the activity and stability of C3convertase via binding with C3b Also, defects in the Factor H gene can cause persistent activation of complement pathways and trigger various diseases . By contrast, Factor H-related proteins (FHRs), including FHR1–5, can either promote or inhibit complement activation. The degree of complement activation depends on the homeostasis betweenFactor H and FHR[67]. However, the relationship between Factor H and ALD has not been clarified and needs further research. In other complement regulatory activities, CD59, protein S and clusterininhibit the formation of the MAC through limiting the binding of complement C9 .Membrane cofactor protein (MCP) and Factor I can inhibit cells from binding with C3b and C4b 

Specific epitope structures of complement, such as anti-complement antibody, complement antisense strand, and complement mutants have been invented, with the intent of inhibiting complement activation. In addition, complement inhibitors and RNA aptamer are being used to inhibit progression of complement-related diseases andC1-INH and CR1 have been used in the treatment of ALD and other diseases[6].

Summary

Mounting evidence indicates that complement activation is involved in the development of ALD at all its stages—fatty liver, alcoholic hepatitis, and fibrosis/cirrhosis. Moreover, all three pathways of complement activation (classical, mannan-binding lectin, and alternative) promote the development of ALD.Therapeutic strategies, using various measures to inhibit complement activation, might prevent the development of ALD, but these strategies cannot be limited a single complement component. Thorough understanding of the relationships between complement activation and ALD may aid in developing new approaches for the treatment of ALD.

Disclosure of Potential Conflicts of Interest

The authors disclose no potential conflicts of interest.

Acknowledgments

This work was supported in part by The National Natural Science Foundation of China (No. 81430014 and No. 31370917), the Natural Science Foundation of Guangxi Province(2015GXNSFFA139004 and 2014GXNSFDA118019,The Bagui Scholarship Foundation.Guangxi health and Family planning commission “139”leading talents training plan.

 

Figure 1:Complement activation pathway and regulation. Three pathways (classical, pathway, MBL, and alternative) are involved in complement activation. Green arrows without a red line indicate the process of complement activation. Green arrows with red line indicate the complement regulatory proteins that inhibit complement activation.

 

Type of regulator

 

Regulator

 

Function

Complement regulatory protein

DAF/CD55, Crry, FH, FI

Inhibit C3, C5 convertase

Complement inhibitor

CD59, protein S, clusterin
C1-INH

Inhibit MAC

Targeted inhibitor

h5G1, 1-ScFv

Inhibit C1r, C1s

RNAaptamer

Specifically bind C1q, C5

Inhibit C5 activation

Table 1: Complementregulators

  1. Warren KR and Murray MM (2013) Alcoholic liver disease and pancreatitis: global health problems being addressed by the US National Institute on Alcohol Abuse and Alcoholism. J GastroenterolHepatol 28 Suppl 1: 4-6.
  2. Yang Y (2010) Mannose Binding Lectin and kidney disease. International Journal Of Urology and Nephrology 30: 241-244.
  3. Borza DB (2016) Glomerular basement membrane heparan sulfate in health and disease: A regulator of local complement activation. Matrix Biol.
  4. Russkamp NF, Ruemmler R, Roewe J, Moore BB, Ward PA et al. (2015) Experimental design of complement component 5a-induced acute lung injury (C5a-ALI): a role of CC-chemokine receptor type 5 during immune activation by anaphylatoxin. Faseb j 29: 3762-3772.
  5. Stewart S, Jones D, Day CP (2001) Alcoholic liver disease: new insights into mechanisms and preventative strategies. Trends Mol Med 7: 408-413.
  6. Järveläinen HA, Väkevä A, Lindros KO, Meri S (2002) Activation of Complement Components and Reduced Regulator Expression in Alcohol-Induced Liver Injury in the Rat. ClinImmunol 105: 57-63.
  7. Bykov I, Junnikkala S, Pekna M, Lindros KO, Meri S (2006) Complement C3 contributes to ethanol-induced liver steatosis in mice. See comment in PubMed Commons below Ann Med 38: 280-286.
  8. Pritchard MT, McMullen MR, Stavitsky AB, Cohen JI, Lin F et al. (2007) Differential contributions of C3, C5, and decayaccelerating factor to ethanol-induced fatty liver in mice. Gastroenterology 132: 1117-1126.
  9. Cohen JI, Roychowdhury S, DiBello PM, Jacobsen DW, Nagy LE (2009) Exogenous thioredoxin prevents ethanol-induced oxidative damage and apoptosis in mouse liver. Hepatology 49: 1709-1717.
  10. Cohen JI, Roychowdhury S, McMullen MR, Stavitsky AB, Nagy LE (2010) Complement and alcoholic liver disease: role of C1q in the pathogenesis of ethanol-induced liver injury in mice. Gastroenterology 139: 664-674, 674.
  11. Orman ES, Odena G, Bataller R (2013) Alcoholic liver disease: pathogenesis, management, and novel targets for therapy. J GastroenterolHepatol 28 Suppl 1: 77-84.
  12. Zhao J. The metabolism and harm of alcohol. Science technology and life. 2011: 195-198.
  13. Kawaratani H, Tsujimoto T, Douhara A, Takaya H, Moriya K et al. (2013) The effect of inflammatory cytokines in alcoholic liver disease. Mediators Inflamm 2013: 495156.
  14. Cohen JI, Chen X, Nagy LE (2011) Redox signaling and the innate immune system in alcoholic liver disease. See comment in PubMed Commons below Antioxid Redox Signal 15: 523-534.
  15. Leung TM and Nieto N (2013) CYP2E1 and oxidant stress in alcoholic and non-alcoholic fatty liver disease. J Hepatol 58: 395-398.
  16. Cohen JI and Nagy LE (2011) The pathogenesis of alcoholic liver disease:The interaction between the parenchyma cell and non parenchymal cells. Joumal of Digestive Disease 16: 131-134.
  17. Nagy LE1 (2015) The Role of Innate Immunity in Alcoholic Liver Disease. See comment in PubMed Commons below Alcohol Res 37: 237-250.
  18. Ricklin D, Hajishengallis G, Yang K, Lambris JD (2010) Complement: a key system for immune surveillance and homeostasis. Nat Immunol 11: 785-797.
  19. Qin X and Gao B (2006) The complement system in liver diseases. Cell MolImmunol 3: 333-340.
  20. Vieyra MB1, Heeger PS (2010) Novel aspects of complement in kidney injury. Kidney Int 77: 495-499.
  21. He W (2010) Medical immunology. Beijing: People’s Medical Publishing House 76-91 p.
  22. Roerecke M, Nanau R, Rehm J, Neuman M (2016) Ethnicity matters: A Systematic Review and Meta-Analysis of the Non-Linear Relationship Between Alcohol Consumption and Prevalence and Incidence of Hepatic Steatosis. E Bio Medicine 8: 317-330.
  23. He S, Atkinson C, Evans Z, Ellett JD, Southwood M et al. (2009) A role for complement in the enhanced susceptibility of steatotic livers to ischemia and reperfusion injury. J Immunol 183: 4764-4772. 24. Bavia L, de Castro IA, Massironi SM, Isaac L (2014) Basal physiological parameters of two congenic mice strains: C5 deficient C57BL/6 and C5 sufficient A/J. ImmunolLett 159: 47-54.
  24. Bavia L, de Castro IA, Massironi SM, Isaac L (2014) Basal physiological parameters of two congenic mice strains: C5 deficient C57BL/6 and C5 sufficient A/J. ImmunolLett 159: 47-54.
  25. Bavia L, de Castro IA, Isaac L (2015) C57BL/6 and A/J Mice Have Different Inflammatory Response and Liver Lipid Profile in Experimental Alcoholic Liver Disease. Mediators Inflamm 2015: 491641.
  26. Shi W, Zhu Q, Gu J, Liu X, Lu L et al. (2013) Anti-IL-17 antibody improves hepatic steatosis by suppressing interleukin-17-related fatty acid synthesis and metabolism. ClinDevImmunol 2013:253046.
  27. Hu X, Wang Y, Hao LY, Liu X, Lesch CA et al. (2015) Corrigendum: Sterol metabolism controls TH17 differentiation by generating endogenous RORgamma agonists. Nat ChemBiol 11: 741.
  28. Sun D and Hu S (2004) Relationship between endothelin -1 and neuropeptide Y and endotoxin-induced complement activation and systemic inflammatory response in healthy volunteers Chinese Critical Care Medicine 16: 48.
  29. Napier BA, Brubaker SW, Sweeney TE, Monette P, et al. (2016) Complement pathway amplifies caspase-11-dependent cell death and endotoxin-induced sepsis severity. See comment in PubMed Commons below J Exp Med 213: 2365-2382.
  30. Lachmann PJ, Lay E, Seilly DJ, Buchberger A, Schwaeble W et al. (2016) Further studies of the down-regulation by Factor I of the C3b feedback cycle using endotoxin as a soluble activator and red cells as a source of CR1 on sera of different complotype. ClinExpImmunol 183: 150-156.
  31. Païdassi H1, Tacnet-Delorme P, Garlatti V, Darnault C, Ghebrehiwet B, et al. (2008) C1q binds phosphatidylserine and likely acts as a multiligand-bridging molecule in apoptotic cell recognition. See comment in PubMed Commons below J Immunol 180: 2329-2338.
  32. Lu JH, Teh BK, Wang Ld, Wang YN, Tan YS, et al. (2008) The classical and regulatory functions of C1q in immunity and autoimmunity. See comment in PubMed Commons below Cell MolImmunol 5: 9-21.
  33. Bavia L, de Castro IA, Cogliati B, Dettoni JB, Alves VA (2016) Complement C5 controls liver lipid profile, promotes liver homeostasis and inflammation in C57BL/6 genetic background. Immunobiology 221: 822-832.
  34. Shen H1, French BA2, Liu H2, Tillman BC2, French SW3 (2014) Increased activity of the complement system in the liver of patients with alcoholic hepatitis. ExpMolPathol 97: 338-344.
  35. Petrasek J, Bala S, Csak T, Lippai D, KodysKet al. (2012) IL-1 receptor antagonist ameliorates inflammasome-dependent alcoholic steatohepatitis in mice. J Clin Invest 122:3476-3489.
  36. Martinon F1, Mayor A, Tschopp J (2009) Theinflammasomes: guardians of the body. See comment in PubMed Commons below Annu Rev Immunol 27: 229-265.
  37. MacLaren R1, Cui W, Cianflone K (2008) Adipokines and the immune system: an adipocentric view. See comment in PubMed Commons below AdvExp Med Biol 632: 1-21.
  38. Cohen JI, Nagy LE, Jiang X (2011) Pathogenesis of Alcoholic Liver Disease:Interactions between Parenchymal and Non-parenchymal Cells. Chinese Journal Of Gastroenterology 16: 131-134.
  39. Leeming DJ, Veidal SS, Karsdal MA, Nielsen MJ, Trebicka J et al. (2015) Pro-C5, a marker of true type V collagen formation and fibrilla-tion, correlates with portal hypertension in patients with alcoholic cirrhosis. Scand J Gastroenterol 50: 584-592.
  40. Jeong WI1, Gao B (2008) Innate immunity and alcoholic liver fibrosis. See comment in PubMed Commons below J GastroenterolHepatol 23 Suppl 1: S112-118.
  41. Duddempudi AT1 (2012) Immunology in alcoholic liver disease. See comment in PubMed Commons below Clin Liver Dis 16: 687-698.
  42. Baumann M, Witzke O, Canbay A, Patschan S, Treichel U et al. (2004) Serum C3 complement concentrations correlate with liver function in patients with liver cirrhosis. Hepatogastroenterology 51: 1451-1453.
  43. Chong WP, To YF, Ip WK, Yuen MF, Poon TP et al. (2005) Mannose-binding lectin in chronic hepatitis B virus infection. Hepatology 42:1037-1045.
  44. Karkampouna S1, Ten Dijke P, Dooley S, Julio MK (2012) TGFβ signaling in liver regeneration. See comment in PubMed Commons below Curr Pharm Des 18: 4103-4113.
  45. Hillebrandt S1, Wasmuth HE, Weiskirchen R, Hellerbrand C, Keppeler H, et al. (2005) Complement factor 5 is a quantitative trait gene that modifies liver fibrogenesis in mice and humans. See comment in PubMed Commons below Nat Genet 37: 835-843.
  46. Deaciuc IV, Fortunato F, D’Souza NB, Hill DB, McClain CJ (2001) Chronic alcohol exposure of rats exacerbates apoptosis in hepatocytes and sinusoidal endothelial cells. Hepatol Res 19: 306-324.
  47. Miller AM1, Wang H, Park O, Horiguchi N, Lafdil F, et al. (2010) Anti-inflammatory and anti-apoptotic roles of endothelial cell STAT3 in alcoholic liver injury. See comment in PubMed Commons below Alcohol ClinExp Res 34: 719-725.
  48. Bajtay Z1, Józsi M, Bánki Z, Thiel S, Thielens N, et al. (2000) Mannan-binding lectin and C1q bind to distinct structures and exert differential effects on macrophages. See comment in PubMed Commons below Eur J Immunol 30: 1706-1713.
  49. Roychowdhury S, McMullen MR, Pritchard MT, Hise AG, van Rooijen N et al. (2009) An early complement-dependent and TLR-4-independent phase in the pathogenesis of ethanol-induced liver injury in mice. Hepatology 49: 1326-1334.
  50. Duffield JS, Forbes SJ, Constandinou CM, Clay S, Partolina M et al. (2005) Selective depletion of macrophages reveals distinct, opposing roles during liver injury and repair. J Clin Invest 115: 56-65.
  51. Fallowfield JA, Mizuno M, Kendall TJ, Constandinou CM, Benyon RC et al. (2007) Scar-associated macrophages are a major source of hepatic matrix metalloproteinase-13 and facilitate the resolution of murine hepatic fibrosis. J Immunol 178: 5288-5295.
  52. Nieto N1 (2006) Oxidative-stress and IL-6 mediate the fibrogenic effects of [corrected] Kupffer cells on stellate cells. See comment in PubMed Commons below Hepatology 44: 1487-1501.
  53. Helmy KY1, Katschke KJ Jr, Gorgani NN, Kljavin NM, Elliott JM, et al. (2006) CRIg: a macrophage complement receptor required for phagocytosis of circulating pathogens. See comment in PubMed Commons below Cell 124: 915-927.
  54. Hinglais N, Kazatchkine MD, Mandet C, Appay MD, Bariety J (1989) Human liver Kupffer cells express CR1, CR3, and CR4 complement receptor antigens. An immunohistochemical study. Lab Invest 61: 509- 514.
  55. Kumar V1, Ali SR, Konrad S, Zwirner J, Verbeek JS, et al. (2006) Cell-derived anaphylatoxins as key mediators of antibody-dependent type II autoimmunity in mice. See comment in PubMed Commons below J Clin Invest 116: 512-520.
  56. Schieferdecker HL, Rothermel E, Timmermann A, Gotze O, Jungermann K (1997) Anaphylatoxin C5a receptor mRNA is strongly expressed in Kupffer and stellate cells and weakly in sinusoidal endothelial cells but not in hepatocytes of normal rat liver. FEBS Lett 406: 305-309.
  57. Schlaf G, Schmitz M, Heine I, Demberg T, Schieferdecker HL (2004) Upregulation of fibronectin but not of entactin, collagen IV and smooth muscle actin by anaphylatoxin C5a in rat hepatic stellate cells. HistolHistopathol 19: 1165-1174.
  58. Guo SP, Zhang WF, Gong JP (2016) [Research advance in the origin and immunological function of kupffer cells]. See comment in PubMed Commons below Sheng Li KeXue Jin Zhan 47: 57-60.
  59. Enomoto N1, Ikejima K, Yamashina S, Hirose M, Shimizu H, et al. (2001) Kupffer cell sensitization by alcohol involves increased permeability to gut-derived endotoxin. See comment in PubMed Commons below Alcohol ClinExp Res 25: 51S-4S.
  60. Wheeler MD1, Thurman RG (2003) Up-regulation of CD14 in liver caused by acute ethanol involves oxidant-dependent AP-1 pathway. See comment in PubMed Commons below J BiolChem 278: 8435-8441.
  61. Schieferdecker HL1, Schlaf G, Jungermann K, Götze O (2001) Functions of anaphylatoxin C5a in rat liver: direct and indirect actions on nonparenchymal and parenchymal cells. See comment in PubMed Commons below IntImmunopharmacol 1: 469-481.
  62. Reid DT, Reyes JL, McDonald BA, Vo T, Reimer RA et al. (2016) Kupffer Cells Undergo Fundamental Changes during the Development of Experimental NASH and Are Critical in Initiating Liver Damage and Inflammation. PLoS One 11: e0159524.
  63. Barata L, Miwa T, Sato S, Kim D, Mohammed I (2013)Deletion of Crry and DAF on murine platelets stimulates thrombopoiesis and increases factor H-dependent resistance of peripheral platelets to complement attack. J Immunol 190: 2886-2895.
  64. Laskowski J, Renner B, Le Quintrec M, Panzer S, Hannan JP et al. (2016) Distinct roles for the complement regulators factor H and Crry in protection of the kidney from injury. Kidney Int 90: 109-122.
  65. Liu F, Wu L, Wu G, Wang C, Zhang L et al. (2014) Targeted mouse complement inhibitor CR2-Crry protects against the development of atherosclerosis in mice. Atherosclerosis 234: 237-243.
  66. Vernon KA, Ruseva MM, Cook HT, Botto M, Malik TH et al. (2016) Partial Complement Factor H Deficiency Associates with C3 Glomerulopathy and Thrombotic Microangiopathy. J Am SocNephrol 27: 1334-1342.
  67. Medjeral-Thomas N1, Pickering MC2 (2016) The complement factor H-related proteins. See comment in PubMed Commons below Immunol Rev 274: 191-201.
  68. Faber C, Williams J, Juel HB, Greenwood J, Nissen MH et al. (2012) Complement factor H deficiency results in decreased neuroretinal expression of Cd59a in aged mice. Invest Ophthalmol Vis Sci 53: 6324- 6330.
  69. Wilson V1, Darlay R, Wong W, Wood KM, McFarlane J, et al. (2013) Genotype/phenotype correlations in complement factor H deficiency arising from uniparentalisodisomy. See comment in PubMed Commons below Am J Kidney Dis 62: 978-983.
  70. Schejbel L, Schmidt IM, Kirchhoff M, Andersen CB, Marquart HV et al. (2011) Complement factor H deficiency and endocapillary glomerulonephritis due to paternal isodisomy and a novel factor H mutation. Genes Immun 12: 90-99.
  71. Blackall DP and Marques MB (2004) Hemolytic uremic syndrome revisited: Shiga toxin, factor H, and fibrin generation. Am J ClinPathol 121 Suppl: S81-88.
  72. Wu G, Hu W, Shahsafaei A, Song W, Dobarro M et al. (2009) Complement regulator CD59 protects against atherosclerosis by restricting the formation of complement membrane attack complex. Circ Res 104: 550-558.
  73. Wickham SE, Hotze EM, Farrand AJ, Polekhina G, Nero TL et al. (2011) Mapping the intermedilysin-human CD59 receptor interface reveals a deep correspondence with the binding site on CD59 for complement binding proteins C8alpha and C9. J BiolChem 286: 20952-20962.
  74. Huang Y, Fedarovich A, Tomlinson S, Davies C (2007) Crystal structure of CD59: implications for molecular recognition of the complement proteins C8 and C9 in the membrane-attack complex. ActaCrystallogr D BiolCrystallogr 63: 714-721.
  75. Podack ER, Muller-Eberhard HJ (1978) Binding of desoxycholate, phosphatidylcholine vesicles, lipoprotein and of the S-protein to complexes of terminal complement components. J Immunol 121: 1025- 1030.
  76. McDonald JF, Nelsestuen GL (1997) Potent inhibition of terminal complement assembly by clusterin: characterization of its impact on C9 polymerization. Biochemistry 36: 7464-7473.
  77. Chu Q, Liu X, Xiang Y, Li Q (2014) study on the structure and function of complement C3. Chinese Journal of Immunology 30: 549-553.
  78. Liu Q and Xing G (2013) Complement regulatory proteins and IgA nephropathy. Chinese Journal of Practical Medicine 40: 122-124.
  79. de Haas CJ, Veldkamp KE, Peschel A, Weerkamp F, Van Wamel WJ et al. (2004) Chemotaxis inhibitory protein of Staphylococcus aureus, a bacterial antiinflammatory agent. J Exp Med 199: 687-695.
  80. Fujita E1, Farkas I, Campbell W, Baranyi L, Okada H, et al. (2004) Inactivation of C5a anaphylatoxin by a peptide that is complementary to a region of C5a. See comment in PubMed Commons below J Immunol 172: 6382-6387.
  81. Guo RF1, Ward PA (2005) Role of C5a in inflammatory responses. See comment in PubMed Commons below Annu Rev Immunol 23: 821-852.
  82. Nikiforovich GV and Baranski TJ (2009) Structural models for the complex of chemotaxis inhibitory protein of Staphylococcus aureus with the C5a receptor. BiochemBiophys Res Commun 390:481-484.
  83. Otto M, Hawlisch H, Monk PN, Muller M, Klos A et al. (2004) C5a mutants are potent antagonists of the C5a receptor (CD88) and of C5L2: position 69 is the locus that determines agonism or antagonism. J BiolChem 279: 142-151.
  84. Li B, Cui S, Lv F, Chen Y, Li Y et al. (2006) Protective effects of complementary peptide of anaphylatoxin C5a on lung injury of experimental sepsis mouse. ActaAcademiae Medicine MilitarisTertiae 28: 4.
  85. Wu Z, Lv F, Lv C, Wu Y (2003) Blocking effects of anti-sense peptide of C5a on the adhesion between pulmonary vascular endothelial cell and neutrophil. Chinese Journal Of Immunology 19: 5.
  86. Li L, Chen L, Zang J, Tang X, Liu Y et al. (2015) C3a and C5a receptor antagonists ameliorate endothelial-myofibroblast transition via the Wnt/beta-catenin signaling pathway in diabetic kidney disease. Metabolism 64: 597-610.
  87. Lee JH, Kim H, Ko J, Lee Y (2002) Interaction of C5 protein with RNA aptamers selected by SELEX. Nucleic Acids Res 30: 5360-5368.
  88. Biesecker G1, Dihel L, Enney K, Bendele RA (1999) Derivation of RNA aptamer inhibitors of human complement C5. See comment in PubMed Commons below Immunopharmacology 42: 219-230.

 

© by the Authors & Gavin Publishers. This is an Open Access Journal Article Published Under Attribution-Share Alike CC BY-SA: Creative Commons Attribution-Share Alike 4.0 International License. With this license, readers can share, distribute, download, even commercially, as long as the original source is properly cited. Read More.

Journal of Digestive Diseases and Hepatology

cara menggunakan pola slot mahjongrtp tertinggi hari inislot mahjong ways 1pola gacor olympus hari inipola gacor starlight princessslot mahjong ways 2strategi olympustrik mahjong ways 2trik olympus hari inirtp koi gatertp pragmatic tertinggicheat jackpot mahjongpg soft link gamertp jackpotelemen sakti mahjongpola maxwin mahjongslot olympus mudah mainrtp live starlightrumus slot mahjongmahjong scatter hitamslot pragmaticjam gacor mahjongpola gacor mahjongstrategi maxwin olympusslot jamin menangrtp slot gacorscatter wild banditopola slot mahjongstrategi maxwin sweet bonanzartp slot terakuratkejutan scatter hitamslot88 resmimaxwin olympuspola mahjong pgsoftretas mahjong waystrik mahjongtrik slot olympusewallet modal recehpanduan pemula slotpg soft primadona slottercheat mahjong androidtips dewa slot mahjongslot demo mahjonghujan scatter olympusrtp caishen winsrtp sweet bonanzamahjong vs qilinmaxwin x5000 starlight princessmahjong wins x1000rtp baru wild scatterpg soft trik maxwinamantotorm1131