Innate immune cells in liver inflammation

Evaggelia Liaskou, Daisy V Wilson, Ye H Oo, Evaggelia Liaskou, Daisy V Wilson, Ye H Oo

Abstract

Innate immune system is the first line of defence against invading pathogens that is critical for the overall survival of the host. Human liver is characterised by a dual blood supply, with 80% of blood entering through the portal vein carrying nutrients and bacterial endotoxin from the gastrointestinal tract. The liver is thus constantly exposed to antigenic loads. Therefore, pathogenic microorganism must be efficiently eliminated whilst harmless antigens derived from the gastrointestinal tract need to be tolerized in the liver. In order to achieve this, the liver innate immune system is equipped with multiple cellular components; monocytes, macrophages, granulocytes, natural killer cells, and dendritic cells which coordinate to exert tolerogenic environment at the same time detect, respond, and eliminate invading pathogens, infected or transformed self to mount immunity. This paper will discuss the innate immune cells that take part in human liver inflammation, and their roles in both resolution of inflammation and tissue repair.

Figures

Figure 1
Figure 1
Innate immune cells in liver inflammation. During an infectious insult in the liver (1) resident macrophages, Kupffer cells, are the first immune cells to detect the presence of invading pathogens (bacteria, parasites, viruses, damaged, and/or necrotic cells) via PRRs/PAMPs. (2) Upon activation Kupffer cells release cytokines TNFα, IL-1, and IL-6 as well as chemokines CXCL 1–3, CXCL-8, CCL-2–4 that initiate (3) the acute-phase response and inflammation. Acute inflammation is characterized by the rise in plasma proteins, collectively named acute-phase proteins (APPs) that include C-reactive protein (CRP) and complement components. (4) Proinflammatory cytokines released from activated Kupffer cells can activate hepatic sinusoidal endothelial cells to upregulate adhesion molecules (ICAM1 and 2, VCAM-1, MAdCAM etc.) and in combination with the chemokines secreted from Kupffer cells can stimulate the recruitment of neutrophils and monocytes to the liver. (5) Neutrophils are the initial phagocytes to arrive at the site of microbial invasion, where (6) they change their phenotype, they become activated and release powerful and cytotoxic antimicrobial molecules such as reactive oxygen species (ROS), oxidants, defensins, as well as chemokines to attract more neutrophils and monocytes. (7) Following their recruitment to the tissue, monocytes undergo differentiation into (8) tissue macrophages (MDMφ), which release TNFα, IL-1β, G-CSF, and GM-CSF factors that can extend the lifespan of neutrophils thus sustaining their presence at the site of inflammation. (9) In order for inflammation to be resolved, the dangerous neutrophils at the inflammatory loci undergo apoptosis and terminate the inflammatory process quickly. Apoptotic neutrophils represent an important anti-inflammatory stimulus to other cells involved in the resolution of inflammation by producing “eat-me” signals recognised by the surrounding phagocytes. Phosphatidylserine (PS) residues on the apoptotic neutrophil membrane allow recognition by its receptor on macrophages, which not only initiates phagocytosis but also modifies the transcriptional profile of the Mφ, increasing the production of IL-10 and TGF-b, cytokines associated with resolution of inflammatory response and tissue repair. Basophils are short-lived cells that express MHC II and CD80/CD86 costimulatory molecules, thus are able to present antigens to CD4+ T cells promoting their differentiation into Th2 cells via release of IL-4 and IL-13. Eosinophils recruited to the liver release proinflammatory mediators including granule-stored cationic proteins, cytokines, and chemokines. They also express MHC II, CD80/CD86, CD40, and ICAM-1; thus they are able to present antigens to T cells initiating or amplifying antigenic-specific immune responses.
Figure 2
Figure 2
Monocyte and macrophage heterogeneity. Monocytes originate in the bone marrow where they develop from hematopoietic stem cells (HSCs) via several differentiation steps and intermediate progenitor stages that pass through the common myeloid progenitor (CMP), the granulocyte/macrophage progenitor (GMP), and the macrophage/DC progenitor (MDP) stages. The MDP gives rise to monocytes, which are released in blood circulation where they remain for 1–3 days. In peripheral blood, circulating monocytes represent ~5–10% of peripheral blood white blood cells (WBCs) and are a highly heterogenic population. Three main subtypes have been described based on the expression of CD14 and CD16 receptors: the classical CD14++CD16, intermediate CD14++CD16+, and nonclassical CD14 low CD16++ monocytes. In general, circulation monocytes are recruited to tissues where they can differentiate into dendritic cells or tissue macrophages (Kupffer cells in the liver; microglial cells in the brain, etc.), replenishing the existing populations. Additional heterogeneity also exists between the macrophages, with two major classes being identified: the classically activated (M1) and the alternatively activated (M2) macrophages. M1 macrophages are developed in response to TNFα and IFNγ as well as in response to microbial products such as LPS, and they produce in turn proinflammatory cytokines including IL-1, IL-23, IL-6, and IL-12. M2 macrophages can develop in response to IL-4 and IL-13 cytokines and play important roles in down-regulation of inflammation and tissue remodelling by releasing IL-10 and IL-1 receptor antagonist (IL-1Ra). They also produce high levels of arginase, fibronectin, and a matrix-associated protein, βIG-H3.
Figure 3
Figure 3
Innate immune cell (neutrophils, NK cells and monocytes) recruitment to hepatic inflammation. Human liver receives dual blood supply from both portal vein and hepatic artery. During the inital event of hepatic inflammation, innate immune cells such as neutrophils, monocytes and NK cells are recruited to the liver. Liver resident dendritic cells sample the foreign antigen and carry to local draining portal lymph nodes where antigens are presented to the adaptive naive T cells. Following the antigen presentation, different types of antigen-specific T effectors cells leave the nodes and drain back to systemic circulation. These T effector cells recruit via hepatic sinusoid towards the site of injury or inflammation. Th17 cells which secrete IL-17 attract neutrophils and also link innate and adaptive immunity.
Figure 4
Figure 4
Linking innate and adaptive immune system. Dendritic cells from innate immune system present their antigen to naive T cells at local draining lymph nodes. T-cell receptor (TCR) ligation to MHC class II associated peptides processed from pathogens (signal 1) and binding of costimulatory molecule CD28 on lymphocyte to CD80 and CD86 expressed by dendritic cells (signal 2) leads to T-cell lineages differentiation. Signal 3 is the polarizing cytokines signals from the innate immune cells. Depending on type of antigen which is presented and nature of cytokines in the microenvironment, innate DC cells direct the development of Th1, Th2, Th17 lymphocytes lineages which plays crucial role in adaptive immune system.

References

    1. O’Farrelly C, Doherty DG. Liver Immunology Principles and Practice. chapter 1. 2007. A short primer on fundamental immunology; pp. 15–24.
    1. Triantafilou M, Lepper PM, Olden R, Dias IS, Triantafilou K. Location, location, location: is membrane partitioning everything when it comes to innate immune activation? Mediators of Inflammation. 2011;2011:10 pages.186093
    1. Akira S, Uematsu S, Takeuchi O. Pathogen recognition and innate immunity. Cell. 2006;124(4):783–801.
    1. Lalor PF, Adams DH. The liver: a model of organ-specific lymphocyte recruitment. Expert Reviews in Molecular Medicine. 2002;4(2):1–16.
    1. Gao B, Jeong WI, Tian Z. Liver: an organ with predominant innate immunity. Hepatology. 2008;47(2):729–736.
    1. Billiar TR, Curran RD, Williams DL, Kispert PH. Liver nonparenchymal cells are stimulated to provide interleukin 6 for induction of the hepatic acute-phase response in endotoxemia but not in remote localized inflammation. Archives of Surgery. 1992;127(1):31–37.
    1. Wagner S, Lynch NJ, Walter W, Schwaeble WJ, Loos M. Differential expression of the murine mannose-binding lectins A and C in lymphoid and nonlymphoid organs and tissues. Journal of Immunology. 2003;170(3):1462–1465.
    1. Gabay C, Kushner I. Acute-phase proteins and other systemic responses to inflammation. The New England Journal of Medicine. 1999;340(6):448–454.
    1. Nielsen SS, Grøofte T, Tygstrup N, Vilstrup H. Synthesis of acute phase proteins in rats with cirrhosis exposed to lipopolysaccharide. Comparative Hepatology. 2006;5, article 3
    1. Moshage H. Cytokines and the hepatic acute phase response. The Journal of Pathology. 1997;181(3):257–266.
    1. Park WB, Lee KD, Lee CS, et al. Production of C-reactive protein in Escherichia coli-infected patients with liver dysfunction due to liver cirrhosis. Diagnostic Microbiology and Infectious Disease. 2005;51(4):227–230.
    1. Gabay C, Roux-Lombard P, De Moerloose P, Dayer JM, Vischer T, Guerne PA. Absence of correlation between interleukin 6 and C-reactive protein blood levels in systemic lupus erythematosus compared with rheumatoid arthritis. Journal of Rheumatology. 1993;20(5):815–821.
    1. Du Clos TW. Function of C-reactive protein. Annals of Medicine. 2000;32(4):274–278.
    1. Cohen JI, Roychowdhury S, McMullen MR, Stavitsky AB, Nagy LE. Complement and alcoholic liver disease: role of C1q in the pathogenesis of ethanol-induced liver injury in mice. Gastroenterology. 2010;139(2):664–674.
    1. Rutkowski MJ, Sughrue ME, Kane AJ, Ahn BJ, Fang S, Parsa AT. The complement cascade as a mediator of tissue growth and regeneration. Inflammation Research. 2010;59(11):897–905.
    1. Heijnen BHM, Straatsburg IH, Padilla ND, Van Mierlo GJ, Hack CE, Van Gulik TM. Inhibition of classical complement activation attenuates liver ischaemia and reperfusion injury in a rat model. Clinical and Experimental Immunology. 2006;143(1):15–23.
    1. Strey CW, Markiewski M, Mastellos D, et al. The proinflammatory mediators C3a and C5a are essential for liver regeneration. Journal of Experimental Medicine. 2003;198(6):913–923.
    1. Mastellos D, Papadimitriou JC, Franchini S, Tsonis PA, Lambris JD. A novel role of complement: mice deficient in the fifth component of complement (C5) exhibit impaired liver regeneration. Journal of Immunology. 2001;166(4):2479–2486.
    1. Markiewski MM, Mastellos D, Tudoran R, et al. C3a and C3b activation products of the third component of complement (C3) are critical for normal liver recovery after toxic injury. Journal of Immunology. 2004;173(2):747–754.
    1. Jaeschke H, Farhood A, Bautista AP, Spolarics Z, Spitzer JJ. Complement activates Kupffer cells and neutrophils during reperfusion after hepatic ischemia. American Journal of Physiology. 1993;264(4):G801–G809.
    1. Summers C, Rankin SM, Condliffe AM, Singh N, Peters AM, Chilvers ER. Neutrophil kinetics in health and disease. Trends in Immunology. 2010;31(8):318–324.
    1. Rankin SM. The bone marrow: a site of neutrophil clearance. Journal of Leukocyte Biology. 2010;88(2):241–251.
    1. Borregaard N. Neutrophils, from marrow to microbes. Immunity. 2010;33(5):657–670.
    1. Sadik CD, Kim ND, Luster AD. Neutrophils cascading their way to inflammation. Trends in Immunology. 2011;32(10):452–460.
    1. Martin C, Burdon PCE, Bridger G, Gutierrez-Ramos JC, Williams TJ, Rankin SM. Chemokines acting via CXCR2 and CXCR4 control the release of neutrophils from the bone marrow and their return following senescence. Immunity. 2003;19(4):583–593.
    1. Edwards S, Lalor PF, Nash GB, Rainger GE, Adams DH. Lymphocyte traffic through sinusoidal endothelial cells is regulated by hepatocytes. Hepatology. 2005;41(3):451–459.
    1. Braet F, Wisse E. Structural and functional aspects of liver sinusoidal endothelial cell fenestrae: a review. Comparative Hepatology. 2002;1, article 1
    1. Yadav SS, Howell DN, Gao W, Steeber DA, Harland RC, Clavien PA. L-selectin and ICAM-1 mediate reperfusion injury and neutrophil adhesion in the warm ischemic mouse liver. American Journal of Physiology. 1998;275(6):G1341–G1352.
    1. Adams DH, Hubscher SG, Fisher NC, Williams A, Robinson M. Expression of E-selectin and E-selectin ligands in human liver inflammation. Hepatology. 1996;24(3):533–538.
    1. Adams DH, Shaw S. Leucocyte-endothelial interactions and regulation of leucocyte migration. The Lancet. 1994;343(8901):831–836.
    1. Ley K, Laudanna C, Cybulsky MI, Nourshargh S. Getting to the site of inflammation: the leukocyte adhesion cascade updated. Nature Reviews Immunology. 2007;7(9):678–689.
    1. Jaeschke H, Smith CW, Clemens MG, Ganey PE, Roth RA. Mechanisms of inflammatory liver injury: adhesion molecules and cytotoxicity of neutrophils. Toxicology and Applied Pharmacology. 1996;139(2):213–226.
    1. Williams MR, Azcutia V, Newton G, Alcaide P, Luscinskas FW. Emerging mechanisms of neutrophil recruitment across endothelium. Trends in Immunology. 2011;32(10):461–469.
    1. Lee WY, Kubes P. Leukocyte adhesion in the liver: distinct adhesion paradigm from other organs. Journal of Hepatology. 2008;48(3):504–512.
    1. Jaeschke H, Hasegawa T. Role of neutrophils in acute inflammatory liver injury. Liver International. 2006;26(8):912–919.
    1. Wong J, Johnston B, Lee SS, et al. A minimal role for selectins in the recruitment of leukocytes into the inflamed liver microvasculature. Journal of Clinical Investigation. 1997;99(11):2782–2790.
    1. Jaeschke H, Farhood A, Fisher MA, Smith CW. Sequestration of neutrophils in the hepatic vasculature during endotoxemia is independent of β2 integrins and intercellular adhesion molecule-1. Shock. 1996;6(5):351–356.
    1. Jaeschke H, Smith CW. Mechanisms of neutrophil-induced parenchymal cell injury. Journal of Leukocyte Biology. 1997;61(6):647–653.
    1. McDonald B, McAvoy EF, Lam F, et al. Interaction of CD44 and hyaluronan is the dominant mechanism for neutrophil sequestration in inflamed liver sinusoids. Journal of Experimental Medicine. 2008;205(4):915–927.
    1. Declèves AE, Caron N, Nonclercq D, et al. Dynamics of hyaluronan, CD44, and inflammatory cells in the rat kidney after ischemia/reperfusion injury. International Journal of Molecular Medicine. 2006;18(1):83–94.
    1. Bajt ML, Farhood A, Jaeschke H. Effects of CXC chemokines on neutrophil activation and sequestration in hepatic vasculature. American Journal of Physiology. 2001;281(5):G1188–G1195.
    1. Borregaard N, Cowland JB. Granules of the human neutrophilic polymorphonuclear leukocyte. Blood. 1997;89(10):3503–3521.
    1. Silva MT, do Vale A, dos Santos NMN. Secondary necrosis in multicellular animals: an outcome of apoptosis with pathogenic implications. Apoptosis. 2008;13(4):463–482.
    1. Nathan C. Neutrophils and immunity: challenges and opportunities. Nature Reviews Immunology. 2006;6(3):173–182.
    1. Yamashiro S, Kamohara H, Wang JM, Yang D, Gong WH, Yoshimura T. Phenotypic and functional change of cytokine-activated neutrophils: inflammatory neutrophils are heterogeneous and enhance adaptive immune responses. Journal of Leukocyte Biology. 2001;69(5):698–704.
    1. De Filippo K, Henderson RB, Laschinger M, Hogg N. Neutrophil chemokines KC and macrophage-inflammatory protein-2 are newly synthesized by tissue macrophages using distinct TLR signaling pathways. Journal of Immunology. 2008;180(6):4308–4315.
    1. Cailhier JF, Partolina M, Vuthoori S, et al. Conditional macrophage ablation demonstrates that resident macrophages initiate acute peritoneal inflammation. Journal of Immunology. 2005;174(4):2336–2342.
    1. Selsted ME, Ouellette AJ. Mammalian defensins in the antimicrobial immune response. Nature Immunology. 2005;6(6):551–557.
    1. Levy O. Antimicrobial proteins and peptides: anti-infective molecules of mammalian leukocytes. Journal of Leukocyte Biology. 2004;76(5):909–925.
    1. Levay PF, Viljoen M. Lactoferrin: a general review. Haematologica. 1995;80(3):252–267.
    1. Zanetti M. Cathelicidins, multifunctional peptides of the innate immunity. Journal of Leukocyte Biology. 2004;75(1):39–48.
    1. Lee A, Whyte MKB, Haslett C. Inhibition of apoptosis and prolongation of neutrophil functional longevity by inflammatory mediators. Journal of Leukocyte Biology. 1993;54(4):283–288.
    1. Miller LS, Pietras EM, Uricchio LH, et al. Inflammasome-mediated production of IL-1β is required for neutrophil recruitment against Staphylococcus aureus in vivo. Journal of Immunology. 2007;179(10):6933–6942.
    1. Scannell M, Flanagan MB, DeStefani A, et al. Annexin-1 and peptide derivatives are released by apoptotic cells and stimulate phagocytosis of apoptotic neutrophils by macrophages. Journal of Immunology. 2007;178(7):4595–4605.
    1. Miller LS, O’Connell RM, Gutierrez MA, et al. MyD88 mediates neutrophil recruitment initiated by IL-1R but not TLR2 activation in immunity against Staphylococcus aureus. Immunity. 2006;24(1):79–91.
    1. Kennedy AD, Deleo FR. Neutrophil apoptosis and the resolution of infection. Immunologic Research. 2009;43(1–3):25–61.
    1. Bonder CS, Ajuebor MN, Zbytnuik LD, Kubes P, Swain MG. Essential role for neutrophil recruitment to the liver in concanavalin A-induced hepatitis. Journal of Immunology. 2004;172(1):45–53.
    1. Tiegs G, Hentschel J, Wendel A. A T cell-dependent experimental liver injury in mice inducible by concanavalin A. Journal of Clinical Investigation. 1992;90(1):196–203.
    1. Jaeschke H, Farhood A, Smith CW. Neutrophils contribute to ischemia/reperfusion injury in rat liver in vivo. FASEB Journal. 1990;4(15):3355–3359.
    1. Suzuki S, Toledo-Pereyra LH, Rodriguez FJ, Cejalvo D. Neutrophil infiltration as an important factor in liver ischemia and reperfusion injury: modulating effects of FK506 and cyclosporine. Transplantation. 1993;55(6):1265–1272.
    1. Bautista AP. Neutrophilic infiltration in alcoholic hepatitis. Alcohol. 2002;27(1):17–21.
    1. Jaeschke H. Neutrophil-mediated tissue injury in alcoholic hepatitis. Alcohol. 2002;27(1):23–27.
    1. Abraham E, Carmody A, Shenkar R, Arcaroli J. Neutrophils as early immunologic effectors in hemorrhage- or endotoxemia-induced acute lung injury. American Journal of Physiology. 2000;279(6):L1137–L1145.
    1. Molnar RG, Wang P, Chaudry IH. Does neutrophil-mediated oxidative stress play any significant role in producing hepatocellular dysfunction during early sepsis? Journal of Surgical Research. 1998;80(1):75–79.
    1. Chosay JG, Essani NA, Dunn CJ, Jaeschke H. Neutrophil margination and extravasation in sinusoids and venules of liver during endotoxin-induced injury. American Journal of Physiology. 1997;272(5):G1195–G1200.
    1. Gujral JS, Farhood A, Jaeschke H. Oncotic necrosis and caspase-dependent apoptosis during galactosamine-induced liver injury in rats. Toxicology and Applied Pharmacology. 2003;190(1):37–46.
    1. Gujral JS, Liu J, Farhood A, Hinson JA, Jaeschke H. Functional importance of ICAM-1 in the mechanism of neutrophil-induced liver injury in bile duct-ligated mice. American Journal of Physiology. 2004;286(3):G499–G507.
    1. Li XK, Matin AFM, Suzuki H, Uno T, Yamaguchi T, Harada Y. Effect of protease inhibitor on ischemia/reperfusion injury of the rat liver. Transplantation. 1993;56(6):1331–1336.
    1. Lemmers A, Moreno C, Gustot T, et al. The interleukin-17 pathway is involved in human alcoholic liver disease. Hepatology. 2009;49(2):646–657.
    1. Tacke F, Randolph GJ. Migratory fate and differentiation of blood monocyte subsets. Immunobiology. 2006;211(6–8):609–618.
    1. Stein M, Keshav S. The versatility of macrophages. Clinical and Experimental Allergy. 1992;22(1):19–27.
    1. Randolph GJ, Beaulieu S, Lebecque S, Steinman RM, Muller WA. Differentiation of monocytes into dendritic cells in a model of transendothelial trafficking. Science. 1998;282(5388):480–483.
    1. Sallusto F, Lanzavecchia A. Efficient presentation of soluble antigen by cultured human dendritic cells is maintained by granulocyte/macrophage colony-stimulating factor plus interleukin 4 and downregulated by tumor necrosis factor α . Journal of Experimental Medicine. 1994;179(4):1109–1118.
    1. Geissmann F, Auffray C, Palframan R, et al. Blood monocytes: distinct subsets, how they relate to dendritic cells, and their possible roles in the regulation of T-cell responses. Immunology and Cell Biology. 2008;86(5):398–408.
    1. Martinez FO, Helming L, Gordon S. Alternative activation of macrophages: an immunologic functional perspective. Annual Review of Immunology. 2009;27:451–483.
    1. Geissmann F, Jung S, Littman DR. Blood monocytes consist of two principal subsets with distinct migratory properties. Immunity. 2003;19(1):71–82.
    1. Gordon S, Taylor PR. Monocyte and macrophage heterogeneity. Nature Reviews Immunology. 2005;5(12):953–964.
    1. Grage-Griebenow E, Flad HD, Ernst M. Heterogeneity of human peripheral blood monocyte subsets. Journal of Leukocyte Biology. 2001;69(1):11–20.
    1. Passlick B, Flieger D, Loms Ziegler-Heitbrock HW. Identification and characterization of a novel monocyte subpopulation in human peripheral blood. Blood. 1989;74(7):2527–2534.
    1. Ziegler-Heitbrock HWL, Fingerle G, Strobel M, et al. The novel subset of CD14+/CD16+ blood monocytes exhibits features of tissue macrophages. European Journal of Immunology. 1993;23(9):2053–2058.
    1. Ziegler-Heitbrock L, Ancuta P, Crowe S, et al. Nomenclature of monocytes and dendritic cells in blood. Blood. 2010;116(16):e74–e80.
    1. Shi C, Pamer EG. Monocyte recruitment during infection and inflammation. Nature Reviews Immunology. 2011;11(11):762–774.
    1. Dolganiuc A, Oak S, Kodys K, et al. Hepatitis C core and nonstructural 3 proteins trigger toll-like receptor 2-mediated pathways and inflammatory activation. Gastroenterology. 2004;127(5):1513–1524.
    1. Geissmann F, Manz MG, Jung S, Sieweke MH, Merad M, Ley K. Development of monocytes, macrophages, and dendritic cells. Science. 2010;327(5966):656–661.
    1. Serbina NV, Jia T, Hohl TM, Pamer EG. Monocyte-mediated defense against microbial pathogens. Annual Review of Immunology. 2008;26:421–452.
    1. Weber C, Belge KU, Von Hundelshausen P, et al. Differential chemokine receptor expression and function in human monocyte subpopulations. Journal of Leukocyte Biology. 2000;67(5):699–704.
    1. Aspinall AI, Curbishley SM, Lalor PF, et al. CX3CR1 and vascular adhesion protein-1-dependent recruitment of CD16+ monocytes across human liver sinusoidal endothelium. Hepatology. 2010;51(6):2030–2039.
    1. Imhof BA, Aurrand-Lions M. Adhesion mechanisms regulating the migration of monocytes. Nature Reviews Immunology. 2004;4(6):432–444.
    1. Bardin N, Blot-Chabaud M, Despoix N, et al. CD146 and its soluble form regulate monocyte transendothelial migration. Arteriosclerosis, Thrombosis, and Vascular Biology. 2009;29(5):746–753.
    1. Gordon S. The role of the macrophage in immune regulation. Research in Immunology. 1998;149(7-8):685–688.
    1. Mosser DM, Edwards JP. Exploring the full spectrum of macrophage activation. Nature Reviews Immunology. 2008;8(12):958–969.
    1. Taylor PR, Martinez-Pomares L, Stacey M, Lin HH, Brown GD, Gordon S. Macrophage receptors and immune recognition. Annual Review of Immunology. 2005;23:901–944.
    1. Gordon S. Alternative activation of macrophages. Nature Reviews Immunology. 2003;3(1):23–35.
    1. Mosser DM. The many faces of macrophage activation. Journal of Leukocyte Biology. 2003;73(2):209–212.
    1. Gregory SH, Wing EJ. Neutrophil-Kupffer-cell interaction in host defenses to systemic infections. Immunology Today. 1998;19(11):507–510.
    1. Naito M, Hasegawa G, Takahashi K. Development, differentiation, and maturation of kupffer cells. Microscopy Research and Technique. 1997;39(4):350–364.
    1. Bilzer M, Roggel F, Gerbes AL. Role of Kupffer cells in host defense and liver disease. Liver International. 2006;26(10):1175–1186.
    1. Cousens LP, Wing EJ. Innate defenses in the liver during Listeria infection. Immunological Reviews. 2000;174:150–159.
    1. Morita A, Itoh Y, Toyama T, et al. Activated Kupffer cells play an important role in intra-hepatic Th1-associated necro-inflammation in Concanavalin A-induced hepatic injury in mice. Hepatology Research. 2003;27(2):143–150.
    1. Mosher B, Dean R, Harkema J, Remick D, Palma J, Crockett E. Inhibition of Kupffer cells reduced CXC chemokine production and liver injury. Journal of Surgical Research. 2001;99(2):201–210.
    1. Ross GD, Vetvicka V. CR3 (CD11b, CD18): a phagocyte and NK cell membrane receptor with multiple ligand specificities and functions. Clinical and Experimental Immunology. 1993;92(2):181–184.
    1. Yamasaki S, Ishikawa E, Sakuma M, Hara H, Ogata K, Saito T. Mincle is an ITAM-coupled activating receptor that senses damaged cells. Nature Immunology. 2008;9(10):1179–1188.
    1. Silva MT. When two is better than one: macrophages and neutrophils work in concert in innate immunity as complementary and cooperative partners of a myeloid phagocyte system. Journal of Leukocyte Biology. 2010;87(1):93–106.
    1. Soehnlein O, Lindbom L. Phagocyte partnership during the onset and resolution of inflammation. Nature Reviews Immunology. 2010;10(6):427–439.
    1. Jaeschke H, Farhood A. Neutrophil and Kupffer cell-induced oxidant stress and ischemia-reperfusion injury in rat liver. American Journal of Physiology. 1991;260(3):G355–G362.
    1. Jaeschke H. Enhanced sinusoidal glutathione efflux during endotoxin-induced oxidant stress in vivo. American Journal of Physiology. 1992;263(1):G60–G68.
    1. Stachlewitz RF, Seabra V, Bradford B, et al. Glycine and uridine prevent D-galactosamine hepatotoxicity in the rat: role of Kupffer cells. Hepatology. 1999;29(3):737–745.
    1. Arthur MJ, Bentley IS, Tanner AR, Saunders PK, Millward-Sadler GH, Wright R. Oxygen-derived free radicals promote hepatic injury in the rat. Gastroenterology. 1985;89(5):1114–1122.
    1. Duffield JS, Forbes SJ, Constandinou CM, et al. Selective depletion of macrophages reveals distinct, opposing roles during liver injury and repair. Journal of Clinical Investigation. 2005;115(1):56–65.
    1. Laskin DL, Pendino KJ. Macrophages and inflammatory mediators in tissue injury. Annual Review of Pharmacology and Toxicology. 1995;35:655–677.
    1. Ju C, Reilly TP, Bourdi M, et al. Protective role of kupffer cells in acetaminophen-induced hepatic injury in mice. Chemical Research in Toxicology. 2002;15(12):1504–1513.
    1. James LP, McCullough SS, Knight TR, Jaeschke H, Hinson JA. Acetaminophen toxicity in mice lacking NADPH oxidase activity: role of peroxynitrite formation and mitochondrial oxidant stress. Free Radical Research. 2003;37(12):1289–1297.
    1. Holt MP, Cheng L, Ju C. Identification and characterization of infiltrating macrophages in acetaminophen-induced liver injury. Journal of Leukocyte Biology. 2008;84(6):1410–1421.
    1. Galli SJ, Tsai M. Mast cells in allergy and infection: versatile effector and regulatory cells in innate and adaptive immunity. European Journal of Immunology. 2010;40(7):1843–1851.
    1. Galli SJ, Borregaard N, Wynn TA. Phenotypic and functional plasticity of cells of innate immunity: macrophages, mast cells and neutrophils. Nature Immunology. 2011;12(11):1035–1044.
    1. Galli SJ, Kalesnikoff J, Grimbaldeston MA, Piliponsky AM, Williams CMM, Tsai M. Mast cells as “tunable” effector and immunoregulatory cells: recent advances. Annual Review of Immunology. 2005;23:749–786.
    1. Rao KN, Brown MA. Mast cells: multifaceted immune cells with diverse roles in health and disease. Annals of the New York Academy of Sciences. 2008;1143:83–104.
    1. Abraham SN, John ALS. Mast cell-orchestrated immunity to pathogens. Nature Reviews Immunology. 2010;10(6):440–452.
    1. Farreil DJ, Hines JE, Walls AF, Kelly PJ, Bennett MK, Burt AD. Intrahepatic mast cells in chronic liver diseases. Hepatology. 1995;22(4, part 1):1175–1181.
    1. Francis H, Meininger CJ. A review of mast cells and liver disease: what have we learned? Digestive and Liver Disease. 2010;42(8):529–536.
    1. Bardadin KA, Scheuer PJ. Mast cells in acute hepatitis. Journal of Pathology. 1986;149(4):315–325.
    1. Nakamura A, Yamazaki K, Suzuki K, Sato S. Increased portal tract infiltration of mast cells and eosinophils in primary biliary cirrhosis. American Journal of Gastroenterology. 1997;92(12):2245–2249.
    1. Ishii M, Iwai M, Harada Y, et al. A role of mast cells for hepatic fibrosis in primary sclerosing cholangitis. Hepatology Research. 2005;31(3):127–131.
    1. Terada T, Matsunaga Y. Increased mast cells in hepatocellular carcinoma and intrahepatic cholangiocarcinoma. Journal of Hepatology. 2000;33(6):961–966.
    1. Francis H, Alpini G, DeMorrow S. Recent advances in the regulation of cholangiocarcinoma growth. American Journal of Physiology. 2010;299(1):G1–G9.
    1. Sullivan BM, Locksley RM. Basophils: a nonredundant contributor to host immunity. Immunity. 2009;30(1):12–20.
    1. Min B. Basophils: what they “can do” versus what they ‘actually do’. Nature Immunology. 2008;9(12):1333–1339.
    1. Karasuyama H, Mukai K, Tsujimura Y, Obata K. Newly discovered roles for basophils: a neglected minority gains new respect. Nature Reviews Immunology. 2009;9(1):9–13.
    1. Lantz CS, Min B, Tsai M, Chatterjea D, Dranoff G, Galli SJ. IL-3 is required for increases in blood basophils in nematode infection in mice and can enhance IgE-dependent IL-4 production by basophils in vitro. Laboratory Investigation. 2008;88(11):1134–1142.
    1. Stone KD, Prussin C, Metcalfe DD. IgE, mast cells, basophils, and eosinophils. Journal of Allergy and Clinical Immunology. 2010;125(2, supplement 2):S73–S80.
    1. Siracusa MC, Perrigoue JG, Comeau MR, Artis D. New paradigms in basophil development, regulation and function. Immunology and Cell Biology. 2010;88(3):275–284.
    1. Siracusa MC, Comeau MR, Artis D. New insights into basophil biology: initiators, regulators, and effectors of type 2 inflammation. Annals of the New York Academy of Sciences. 2011;1217(1):166–177.
    1. Perrigoue JG, Saenz SA, Siracusa MC, et al. MHC class II-dependent basophil-CD4+ T cell interactions promote TH2 cytokine-dependent immunity. Nature Immunology. 2009;10(7):697–705.
    1. Charles N, Hardwick D, Daugas E, Illei GG, Rivera J. Basophils and the T helper 2 environment can promote the development of lupus nephritis. Nature Medicine. 2010;16(6):701–707.
    1. Min B, Prout M, Hu-Li J, et al. Basophils produce IL-4 and accumulate in tissues after infection with a Th2-inducing parasite. Journal of Experimental Medicine. 2004;200(4):507–517.
    1. Komiya A, Nagase H, Okugawa S, et al. Expression and function of toll-like receptors in human basophils. International Archives of Allergy and Immunology. 2006;140(1, supplement):23–27.
    1. Min B, Le Gros G, Paul WE. Basophils: a potential liaison between innate and adaptive immunity. Allergology International. 2006;55(2):99–104.
    1. Dahinden CA, Geiser T, Brunner T, et al. Monocyte chemotactic protein 3 is a most effective basophil- and eosinophil-activating chemokine. Journal of Experimental Medicine. 1994;179(2):751–756.
    1. Jinquan T, Jacobi HH, Jing C, et al. Chemokine stromal cell-derived factor 1α activates basophils by means of CXCR4. Journal of Allergy and Clinical Immunology. 2000;106(2):313–320.
    1. Tanimoto Y, Takahashi K, Kimura I. Effects of cytokines on human basophil chemotaxis. Clinical and Experimental Allergy. 1992;22(11):1020–1025.
    1. Forssmann U, Uguccioni M, Loetscher P, et al. Eotaxin-2, a novel CC chemokine that is selective for the chemokine receptor CCR3, and acts like eotaxin on human eosinophil and basophil leukocytes. Journal of Experimental Medicine. 1997;185(12):2171–2176.
    1. Eglite S, Pluss K, Dahinden CA. Requirements for C5a receptor-mediated IL-4 and IL-13 production and leukotriene C4 generation in human basophils. Journal of Immunology. 2000;165(4):2183–2189.
    1. de Paulis A, Prevete N, Fiorentino I, et al. Basophils infiltrate human gastric mucosa at sites of Helicobacter pylori infection, and exhibit chemotaxis in response to H. pylori-derived peptide Hp(2–20) Journal of Immunology. 2004;172(12):7734–7743.
    1. Brunner T, de Weck AL, Dahinden CA. Platelet-activating factor induces mediator release by human basophils primed with IL-3, granulocyte-macrophage colony-stimulating factor, or IL-5. Journal of Immunology. 1991;147(1):237–242.
    1. Bochner BS. Systemic activation of basophils and eosinophils: markers and consequences. Journal of Allergy and Clinical Immunology. 2000;106(5, supplement):S292–S302.
    1. Yoshimoto T, Yasuda K, Tanaka H, et al. Basophils contribute to TH2-IgE responses in vivo via IL-4 production and presentation of peptide-MHC class II complexes to CD4+ T cells. Nature Immunology. 2009;10(7):706–712.
    1. Nakanishi K. Basophils are potent antigen-presenting cells that selectively induce Th2 cells. European Journal of Immunology. 2010;40(7):1836–1842.
    1. Sokol CL, Chu NQ, Yu S, Nish SA, Laufer TM, Medzhitov R. Basophils function as antigen-presenting cells for an allergen-induced T helper type 2 response. Nature Immunology. 2009;10(7):713–720.
    1. Kim S, Prout M, Ramshaw H, Lopez AF, LeGros G, Min B. Cutting edge: basophils are transiently recruited into the draining lymph nodes during helminth infection via IL-3, but infection-induced Th2 immunity can develop without basophil lymph node recruitment or IL-3. Journal of Immunology. 2010;184(3):1143–1147.
    1. Hida S, Tadachi M, Saito T, Taki S. Negative control of basophil expansion by IRF-2 critical for the regulation of Th1/Th2 balance. Blood. 2005;106(6):2011–2017.
    1. Oh K, Shen T, Le Gros G, Min B. Induction of Th2 type immunity in a mouse system reveals a novel immunoregulatory role of basophils. Blood. 2007;109(7):2921–2927.
    1. Weller PF. The immunobiology of eosinophils. The New England Journal of Medicine. 1991;324(16):1110–1118.
    1. Foster PS, Mould AW, Yang M, et al. Elemental signals regulating eosinophil accumulation in the lung. Immunological Reviews. 2001;179:173–181.
    1. Rothenberg ME, Mishra A, Brandt EB, Hogan SP. Gastrointestinal eosinophils. Immunological Reviews. 2001;179:139–155.
    1. Gouon-Evans V, Pollard JW. Eotaxin is required for eosinophil homing into the stroma of the pubertal and cycling uterus. Endocrinology. 2001;142(10):4515–4521.
    1. DiScipio RG, Schraufstatter IU. The role of the complement anaphylatoxins in the recruitment of eosinophils. International Immunopharmacology. 2007;7(14):1909–1923.
    1. Cruikshank WW, Long A, Tarpy RE, et al. Early identification of interleukin-16 (lymphocyte chemoattractant factor) and macrophage inflammatory protein 1 alpha (MIP1 alpha) in bronchoalveolar lavage fluid of antigen-challenged asthmatics. American Journal of Respiratory Cell and Molecular Biology. 1995;13(6):738–747.
    1. Rot A, Krieger M, Brunner T, Bischoff SC, Schall TJ, Dahinden CA. RANTES and macrophage inflammatory protein 1α induce the migration and activation of normal human eosinophil granulocytes. Journal of Experimental Medicine. 1992;176(6):1489–1495.
    1. Badewa AP, Hudson CE, Heiman AS. Regulatory effects of eotaxin, eotaxin-2, and eotaxin-3 on eosinophil degranulation and superoxide anion generation. Experimental Biology and Medicine. 2002;227(8):645–651.
    1. Ponath PD, Qin S, Ringler DJ, et al. Cloning of the human eosinophil chemoattractant, eotaxin: expression, receptor binding, and functional properties suggest a mechanism for the selective recruitment of eosinophils. Journal of Clinical Investigation. 1996;97(3):604–612.
    1. Kitayama J, Fuhlbrigge RC, Puri KD, Springer TA. P-selectin, L-selectin, and alpha 4 integrin have distinct roles in eosinophil tethering and arrest on vascular endothelial cells under physiological flow conditions. Journal of Immunology. 1997;159(8):3929–3939.
    1. Patel KD, McEver RP. Comparison of tethering and rolling of eosinophils and neutrophils through selectins and P-selectin glycoprotein ligand-1. Journal of Immunology. 1997;159(9):4555–4565.
    1. Walsh GM, Symon FA, Lazarovits AI, Wardlaw AJ. Integrin α4β7 mediates human eosinophil interaction with MAdCAM-1, VCAM-1 and fibronectin. Immunology. 1996;89(1):112–119.
    1. Yamamoto H, Nagata M. Regulatory mechanisms of eosinophil adhesion to and transmigration across endothelial cells by α4 and β2 integrins. International Archives of Allergy and Immunology. 1999;120(1, supplement):24–26.
    1. Muller WA, Weigl SA, Deng X, Phillips DM. PECAM-1 is required for transendothelial migration of leukocytes. Journal of Experimental Medicine. 1993;178(2):449–460.
    1. Nagase H, Okugawa S, Ota Y, et al. Expression and function of toll-like receptors in eosinophils: activation by toll-like receptor 7 ligand. Journal of Immunology. 2003;171(8):3977–3982.
    1. Takafuji S, Tadokoro K, Ito K, Dahinden CA. Degranulation from human eosinophils stimulated with C3a and C5a. International Archives of Allergy and Immunology. 1994;104(1, supplement):27–29.
    1. Malm-Erjefält M, Persson CGA, Erjefält JS. Degranulation status of airway tissue eosinophils in mouse models of allergic airway inflammation. American Journal of Respiratory Cell and Molecular Biology. 2001;24(3):352–359.
    1. Hogan SP, Rosenberg HF, Moqbel R, et al. Eosinophils: biological properties and role in health and disease. Clinical and Experimental Allergy. 2008;38(5):709–750.
    1. Gauthier MC, Racine C, Ferland C, et al. Expression of membrane type-4 matrix metalloproteinase (metalloproteinase-17) by human eosinophils. International Journal of Biochemistry and Cell Biology. 2003;35(12):1667–1673.
    1. Puxeddu I, Berkman N, Ribatti D, et al. Osteopontin is expressed and functional in human eosinophils. Allergy. 2010;65(2):168–174.
    1. Horiuchi T, Weller PF. Expression of vascular endothelial growth factor by human eosinophils: upregulation by granulocyte macrophage colony-stimulating factor and interleukin-5. American Journal of Respiratory Cell and Molecular Biology. 1997;17(1):70–77.
    1. Kita H. Eosinophils: multifaceted biological properties and roles in health and disease. Immunological Reviews. 2011;242(1):161–177.
    1. Padigel UM, Lee JJ, Nolan TJ, Schad GA, Abraham D. Eosinophils can function as antigen-presenting cells to induce primary and secondary immune responses to Strongyloides stercoralis. Infection and Immunity. 2006;74(6):3232–3238.
    1. Shi HZ. Eosinophils function as antigen-presenting cells. Journal of Leukocyte Biology. 2004;76(3):520–527.
    1. Terasaki S, Nakanuma Y, Yamazaki M, Unoura M. Eosinophilic infiltration of the liver in primary biliary cirrhosis: a morphological study. Hepatology. 1993;17(2):206–212.
    1. Martinez OM, Villanueva JC, Gershwin ME, Krams SM. Cytokine patterns and cytotoxic mediators in primary biliary cirrhosis. Hepatology. 1995;21(1):113–119.
    1. Hartleb M, Kajor M, Kaczor R, Nowak A. Hepatic eosinophilic infiltration in primary sclerosing cholangitis. Journal of Gastroenterology. 1998;33(1):134–135.
    1. Scheurlen M, Mork H, Weber P. Hypereosinophilic syndrome resembling chronic inflammatory bowel disease with primary sclerosing cholangitis. Journal of Clinical Gastroenterology. 1992;14(1):59–63.
    1. Dillon JF, Finlayson NDC. Idiopathic hypereosinophilic syndrome presenting as intrahepatic cholestatic jaundice. American Journal of Gastroenterology. 1994;89(8):1254–1255.
    1. Foong A, Scholes JV, Gleich GJ, Kephart GM, Holt PR. Eosinophil-induced chronic active hepatitis in the idiopathic hypereosinophilic syndrome. Hepatology. 1991;13(6):1090–1094.
    1. Navarro VJ, Senior JR. Drug-related hepatotoxicity. The New England Journal of Medicine. 2006;354(7):731–739.
    1. Björnsson E, Kalaitzakis E, Olsson R. The impact of eosinophilia and hepatic necrosis on prognosis in patients with drug-induced liver injury. Alimentary Pharmacology and Therapeutics. 2007;25(12):1411–1421.
    1. Nonomura A, Kono N, Mizukami Y, Nakanuma Y. Histological changes of the liver in experimental graft-versus-host disease across minor histocompatibility barriers. VIII. Role of eosinophil infiltration. Liver. 1996;16(1):42–47.
    1. Hughes VF, Trull AK, Joshi O, Alexander GJM. Monitoring eosinophil activation and liver function after liver transplantation. Transplantation. 1998;65(10):1334–1339.
    1. Martinez OM, Ascher NL, Ferrell L, et al. Evidence for a nonclassical pathway of graft rejection involving interleukin 5 and eosinophils. Transplantation. 1993;55(4):909–918.
    1. Foster PF, Bhattacharyya A, Sankary HN, Coleman J, Ashmann M, Williams JW. Eosinophil cationic protein’s role in human hepatic allograft rejection. Hepatology. 1991;13(6):1117–1125.
    1. Nagral A, Ben-Ari Z, Dhillon AP, Burroughs AK. Eosinophils in acute cellular rejection in liver allografts. Liver Transplantation and Surgery. 1998;4(5):355–362.
    1. Dollinger MM, Plevris JN, Bouchier IAD, Harrison DJ, Hayes PC. Peripheral eosinophil count both before and after liver transplantation predicts acute cellular rejection. Liver Transplantation and Surgery. 1997;3(2):112–117.
    1. Tsuda K, Maeda T, Tominaga A, et al. Eosinophil-induced liver injury: an experimental model using IL-5 transgenic mice. Journal of Hepatology. 2001;34(2):270–277.
    1. Takahashi Y, Fukusato T, Kobayashi Y, et al. High expression of eosinophil chemoattractant ecalectin/galectin-9 in drug-induced liver injury. Liver International. 2006;26(1):106–115.
    1. Tarantino G, Cabibi D, Cammà C, et al. Liver eosinophilic infiltrate is a significant finding in patients with chronic hepatitis C. Journal of Viral Hepatitis. 2008;15(7):523–530.
    1. Steinman RM, Cohn ZA. Identification of a novel cell type in peripheral lymphoid organs of mice. I. Morphology, quantitation, tissue distribution. Journal of Experimental Medicine. 1973;137(5):1142–1162.
    1. Liu YJ. Dendritic cell subsets and lineages, and their functions in innate and adaptive immunity. Cell. 2001;106(3):259–262.
    1. Liu Y-J, Kanzler H, Soumelis V, Gilliet M. Dendritic cell lineage, plasticity and cross-regulation. Nature Immunology. 2001;2(7):585–589.
    1. Ueno H, Klechevsky E, Morita R, et al. Dendritic cell subsets in health and disease. Immunological Reviews. 2007;219(1):118–142.
    1. Altfeld M, Fadda L, Frleta D, Bhardwaj N. DCs and NK cells: critical effectors in the immune response to HIV-1. Nature Reviews Immunology. 2011;11(3):176–186.
    1. Liu YJ. IPC: professional type 1 interferon-producing cells and plasmacytoid dendritic cell precursors. Annual Review of Immunology. 2005;23:275–306.
    1. Weiner HL. The mucosal milieu creates tolerogenic dendritic cells and TRl and TH3 regulatory cells. Nature Immunology. 2001;2(8):671–672.
    1. Mancuso G, Gambuzza M, Midiri A, et al. Bacterial recognition by TLR7 in the lysosomes of conventional dendritic cells. Nature Immunology. 2009;10(6):587–594.
    1. Siegal FP, Kadowaki N, Shodell M, et al. The nature of the principal Type 1 interferon-producing cells in human blood. Science. 1999;284(5421):1835–1837.
    1. Shaw AS. How T cells “find” the right dendritic cell. Nature Immunology. 2008;9(3):229–230.
    1. Hermans IF, Silk JD, Gileadi U, et al. NKT cells enhance CD4+ and CD8+ T cell responses to soluble antigen in vivo through direct interaction with dendritic cells. Journal of Immunology. 2003;171(10):5140–5147.
    1. Steinbrink K, Wölfl M, Jonuleit H, Knop J, Enk AH. Induction of tolerance by IL-10-treated dendritic cells. Journal of Immunology. 1997;159(10):4772–4780.
    1. Martín-Fontecha A, Thomsen LL, Brett S, et al. Induced recruitment of NK cells to lymph nodes provides IFN-γ for TH1 priming. Nature Immunology. 2004;5(12):1260–1265.
    1. Soumelis V, Reche PA, Kanzler H, et al. Human epithelial cells trigger dendritic cell-mediated allergic inflammation by producing TSLP. Nature Immunology. 2002;3(7):673–680.
    1. Goddard S, Youster J, Morgan E, Adams DH. Interleukin-10 secretion differentiates dendritic cells from human liver and skin. American Journal of Pathology. 2004;164(2):511–519.
    1. Colonna M, Trinchieri G, Liu YJ. Plasmacytoid dendritic cells in immunity. Nature Immunology. 2004;5(12):1219–1226.
    1. Yamazaki S, Patel M, Harper A, et al. Effective expansion of alloantigen-specific Foxp3+ CD25+ CD4+ regulatory T cells by dendritic cells during the mixed leukocyte reaction. Proceedings of the National Academy of Sciences of the United States of America. 2006;103(8):2758–2763.
    1. Hadeiba H, Sato T, Habtezion A, Oderup C, Pan J, Butcher EC. CCR9 expression defines tolerogenic plasmacytoid dendritic cells able to suppress acute graft-versus-host disease. Nature Immunology. 2008;9(11):1253–1260.
    1. Lau AH, Thomson AW. Dendritic cells and immune regulation in the liver. Gut. 2003;52(2):307–314.
    1. Sumpter TL, Abe M, Tokita D, Thomson AW. Dendritic cells, the liver, and transplantation. Hepatology. 2007;46(6):2021–2031.
    1. Wisse E, van’t Noordende JM, van der Meulen J, Daems TW. The pit cell: description of a new type of cell occurring in rat liver sinusoids and peripheral blood. Cell and Tissue Research. 1976;173(4):423–435.
    1. Biron CA, Nguyen KB, Pien GC, Cousens LP, Salazar-Mather TP. Natural killer cells in antiviral defense: function and regulation by innate cytokines. Annual Review of Immunology. 1999;17:189–220.
    1. Orange JS, Salazar-Mather TP, Opal SM, Biron CA. Mechanisms for virus-induced liver disease: tumor necrosis factor- mediated pathology independent of natural killer and T cells during murine cytomegalovirus infection. Journal of Virology. 1997;71(12):9248–9258.
    1. Doherty DG, O’Farrelly C. Innate and adaptive lymphoid cells in the human liver. Immunological Reviews. 2000;174:5–20.
    1. Caligiuri MA. Human natural killer cells. Blood. 2008;112(3):461–469.
    1. Yu J, Mao HC, Wei M, et al. CD94 surface density identifies a functional intermediary between the CD56bright and CD56dim human NK-cell subsets. Blood. 2010;115(2):274–281.
    1. Mavilio D, Lombardo G, Benjamin J, et al. Characterization of CD56−/CD16+ natural killer (NK) cells: a highly dysfunctional NK subset expanded in HIV-infected viremic individuals. Proceedings of the National Academy of Sciences of the United States of America. 2005;102(8):2886–2891.
    1. Lanier LL. Up on the tightrope: natural killer cell activation and inhibition. Nature Immunology. 2008;9(5):495–502.
    1. Jinushi M, Takehara T, Tatsumi T, et al. Negative regulation of NK cell activities by inhibitory receptor CD94/NKG2A leads to altered NK cell-induced modulation of dendritic cell functions in chronic hepatitis C virus infection. Journal of Immunology. 2004;173(10):6072–6081.
    1. Krueger PD, Lassen MG, Qiao H, Hahn YS. Regulation of NK cell repertoire and function in the liver. Critical Reviews in Immunology. 2011;31(1):43–52.
    1. Hokeness KL, Kuziel WA, Biron CA, Salazar-Mather TP. Monocyte chemoattractant protein-1 and CCR2 interactions are required for IFN-α/β-induced inflammatory responses and antiviral defense in liver. Journal of Immunology. 2005;174(3):1549–1556.
    1. Salazar-Mather TP, Orange JS, Biron CA. Early murine cytomegalovirus (MCMV) infection induces liver natural killer (NK) cell inflammation and protection through macrophage inflammatory protein 1α (MIP-1α)-dependent pathways. Journal of Experimental Medicine. 1998;187(1):1–14.
    1. Curbishley SM, Eksteen B, Gladue RP, Lalor P, Adams DH. CXCR3 activation promotes lymphocyte transendothelial migration across human hepatic endothelium under fluid flow. American Journal of Pathology. 2005;167(3):887–899.
    1. Maghazachi AA. Role of chemokines in the biology of natural killer cells. Current Topics in Microbiology and Immunology. 2010;341:37–58.
    1. Lanier LL. NK cell recognition. Annual Review of Immunology. 2005;23:225–274.
    1. Jiang W, Sun R, Zhou R, Wei H, Tian Z. TLR-9 activation aggravates concanavalin A-induced hepatitis via promoting accumulation and activation of liver CD4+ NKT cells. Journal of Immunology. 2009;182(6):3768–3774.
    1. Seino KI, Taniguchi M. Functionally distinct NKT cell subsets and subtypes. Journal of Experimental Medicine. 2005;202(12):1623–1626.
    1. Gao B. Natural killer group 2 member D, its ligands, and liver disease: good or bad? Hepatology. 2010;51(1):8–11.
    1. Gao B, Radaeva S, Park O. Liver natural killer and natural killer T cells: immunobiology and emerging roles in liver diseases. Journal of Leukocyte Biology. 2009;86(3):513–528.
    1. Swain MG. Hepatic NKT cells: friend or foe? Clinical Science. 2008;114(7-8):457–466.
    1. Biburger M, Tiegs G. α-galactosylceramide-induced liver injury in mice is mediated by TNF-α but independent of Kupffer cells. Journal of Immunology. 2005;175(3):1540–1550.
    1. Syn WK, Oo YH, Pereira TA, et al. Accumulation of natural killer T cells in progressive nonalcoholic fatty liver disease. Hepatology. 2010;51(6):1998–2007.
    1. Santodomingo-Garzon T, Han J, Le T, Yang Y, Swain MG. Natural killer T cells regulate the homing of chemokine CXC receptor 3-positive regulatory T cells to the liver in mice. Hepatology. 2009;49(4):1267–1276.
    1. Liaskou E, Karikoski M, Reynolds GM, et al. Regulation of mucosal addressin cell adhesion molecule 1 expression in human and mice by vascular adhesion protein 1 amine oxidase activity. Hepatology. 2011;53(2):661–672.
    1. Ridker PM, Cannon CP, Morrow D, et al. C-reactive protein levels and outcomes after statin therapy. The New England Journal of Medicine. 2005;352(1):20–28.
    1. Ridker PM, Koenig W, Fuster V. C-reactive protein and coronary heart disease. The New England Journal of Medicine. 2004;351(3):295–295.
    1. Jones SA, Scheller J, Rose-John S. Therapeutic strategies for the clinical blockade of IL-6/gp130 signaling. Journal of Clinical Investigation. 2011;121(9):3375–3383.
    1. Shook DR, Campana D. Natural killer cell engineering for cellular therapy of cancer. Tissue Antigens. 2011;78(6):409–415.
    1. Smits ELJ, Lee C, Hardwick N, et al. Clinical evaluation of cellular immunotherapy in acute myeloid leukaemia. Cancer Immunology, Immunotherapy. 2011;60(6):757–769.
    1. Lu W, Arraes LC, Ferreira WT, Andrieu JM. Therapeutic dendritic-cell vaccine for chronic HIV-1 infection. Nature Medicine. 2004;10(12):1359–1365.
    1. Palmer DH, Midgley RS, Mirza N, et al. A phase II study of adoptive immunotherapy using dendritic cells pulsed with tumor lysate in patients with hepatocellular carcinoma. Hepatology. 2009;49(1):124–132.

Source: PubMed

3
Suscribir