Chemokine receptor CCR6-dependent accumulation of γδ T cells in injured liver restricts hepatic inflammation and fibrosis

Standard

Chemokine receptor CCR6-dependent accumulation of γδ T cells in injured liver restricts hepatic inflammation and fibrosis. / Hammerich, Linda; Bangen, Jörg M; Govaere, Olivier; Zimmermann, Henning W; Gassler, Nikolaus; Huss, Sebastian; Liedtke, Christian; Prinz, Immo; Lira, Sergio A; Luedde, Tom; Roskams, Tania; Trautwein, Christian; Heymann, Felix; Tacke, Frank.

in: HEPATOLOGY, Jahrgang 59, Nr. 2, 02.2014, S. 630-42.

Publikationen: SCORING: Beitrag in Fachzeitschrift/ZeitungSCORING: ZeitschriftenaufsatzForschungBegutachtung

Harvard

Hammerich, L, Bangen, JM, Govaere, O, Zimmermann, HW, Gassler, N, Huss, S, Liedtke, C, Prinz, I, Lira, SA, Luedde, T, Roskams, T, Trautwein, C, Heymann, F & Tacke, F 2014, 'Chemokine receptor CCR6-dependent accumulation of γδ T cells in injured liver restricts hepatic inflammation and fibrosis', HEPATOLOGY, Jg. 59, Nr. 2, S. 630-42. https://doi.org/10.1002/hep.26697

APA

Hammerich, L., Bangen, J. M., Govaere, O., Zimmermann, H. W., Gassler, N., Huss, S., Liedtke, C., Prinz, I., Lira, S. A., Luedde, T., Roskams, T., Trautwein, C., Heymann, F., & Tacke, F. (2014). Chemokine receptor CCR6-dependent accumulation of γδ T cells in injured liver restricts hepatic inflammation and fibrosis. HEPATOLOGY, 59(2), 630-42. https://doi.org/10.1002/hep.26697

Vancouver

Bibtex

@article{498eb691fef447bc87e99dc6f70f4ed2,
title = "Chemokine receptor CCR6-dependent accumulation of γδ T cells in injured liver restricts hepatic inflammation and fibrosis",
abstract = "UNLABELLED: Chronic liver injury promotes hepatic inflammation, representing a prerequisite for organ fibrosis. We hypothesized a contribution of chemokine receptor CCR6 and its ligand, CCL20, which may regulate migration of T-helper (Th)17, regulatory, and gamma-delta (γδ) T cells. CCR6 and CCL20 expression was intrahepatically up-regulated in patients with chronic liver diseases (n = 50), compared to control liver (n = 5). Immunohistochemistry revealed the periportal accumulation of CCR6(+) mononuclear cells and CCL20 induction by hepatic parenchymal cells in liver disease patients. Similarly, in murine livers, CCR6 was expressed by macrophages, CD4 and γδ T-cells, and up-regulated in fibrosis, whereas primary hepatocytes induced CCL20 upon experimental injury. In two murine models of chronic liver injury (CCl4 and methionine-choline-deficient diet), Ccr6(-/-) mice developed more severe fibrosis with strongly enhanced hepatic immune cell infiltration, compared to wild-type (WT) mice. Although CCR6 did not affect hepatic Th-cell subtype composition, CCR6 was explicitly required by the subset of interleukin (IL)-17- and IL-22-expressing γδ T cells for accumulation in injured liver. The adoptive transfer of WT γδ, but not CD4 T cells, into Ccr6(-/-) mice reduced hepatic inflammation and fibrosis in chronic injury to WT level. The anti-inflammatory function of hepatic γδ T cells was independent of IL-17, as evidenced by transfer of Il-17(-/-) cells. Instead, hepatic γδ T cells colocalized with hepatic stellate cells (HSCs) in vivo and promoted apoptosis of primary murine HSCs in a cell-cell contact-dependent manner, involving Fas-ligand (CD95L). Consistent with γδ T-cell-induced HSC apoptosis, activated myofibroblasts were more frequent in fibrotic livers of Ccr6(-/-) than in WT mice.CONCLUSION: γδ T cells are recruited to the liver by CCR6 upon chronic injury and protect the liver from excessive inflammation and fibrosis by inhibiting HSCs.",
keywords = "Animals, Apoptosis, Case-Control Studies, Cell Movement, Chemokine CCL20/metabolism, Disease Models, Animal, Female, Hepatitis/metabolism, Humans, Interleukin-17/metabolism, Liver Cirrhosis/metabolism, Liver Diseases/metabolism, Male, Mice, Inbred C57BL, Mice, Knockout, Receptors, Antigen, T-Cell, gamma-delta/metabolism, Receptors, CCR6/deficiency, T-Lymphocytes/metabolism, T-Lymphocytes, Regulatory/metabolism, Th17 Cells/metabolism, Up-Regulation",
author = "Linda Hammerich and Bangen, {J{\"o}rg M} and Olivier Govaere and Zimmermann, {Henning W} and Nikolaus Gassler and Sebastian Huss and Christian Liedtke and Immo Prinz and Lira, {Sergio A} and Tom Luedde and Tania Roskams and Christian Trautwein and Felix Heymann and Frank Tacke",
note = "{\textcopyright} 2013 by the American Association for the Study of Liver Diseases.",
year = "2014",
month = feb,
doi = "10.1002/hep.26697",
language = "English",
volume = "59",
pages = "630--42",
journal = "HEPATOLOGY",
issn = "0270-9139",
publisher = "John Wiley and Sons Ltd",
number = "2",

}

RIS

TY - JOUR

T1 - Chemokine receptor CCR6-dependent accumulation of γδ T cells in injured liver restricts hepatic inflammation and fibrosis

AU - Hammerich, Linda

AU - Bangen, Jörg M

AU - Govaere, Olivier

AU - Zimmermann, Henning W

AU - Gassler, Nikolaus

AU - Huss, Sebastian

AU - Liedtke, Christian

AU - Prinz, Immo

AU - Lira, Sergio A

AU - Luedde, Tom

AU - Roskams, Tania

AU - Trautwein, Christian

AU - Heymann, Felix

AU - Tacke, Frank

N1 - © 2013 by the American Association for the Study of Liver Diseases.

PY - 2014/2

Y1 - 2014/2

N2 - UNLABELLED: Chronic liver injury promotes hepatic inflammation, representing a prerequisite for organ fibrosis. We hypothesized a contribution of chemokine receptor CCR6 and its ligand, CCL20, which may regulate migration of T-helper (Th)17, regulatory, and gamma-delta (γδ) T cells. CCR6 and CCL20 expression was intrahepatically up-regulated in patients with chronic liver diseases (n = 50), compared to control liver (n = 5). Immunohistochemistry revealed the periportal accumulation of CCR6(+) mononuclear cells and CCL20 induction by hepatic parenchymal cells in liver disease patients. Similarly, in murine livers, CCR6 was expressed by macrophages, CD4 and γδ T-cells, and up-regulated in fibrosis, whereas primary hepatocytes induced CCL20 upon experimental injury. In two murine models of chronic liver injury (CCl4 and methionine-choline-deficient diet), Ccr6(-/-) mice developed more severe fibrosis with strongly enhanced hepatic immune cell infiltration, compared to wild-type (WT) mice. Although CCR6 did not affect hepatic Th-cell subtype composition, CCR6 was explicitly required by the subset of interleukin (IL)-17- and IL-22-expressing γδ T cells for accumulation in injured liver. The adoptive transfer of WT γδ, but not CD4 T cells, into Ccr6(-/-) mice reduced hepatic inflammation and fibrosis in chronic injury to WT level. The anti-inflammatory function of hepatic γδ T cells was independent of IL-17, as evidenced by transfer of Il-17(-/-) cells. Instead, hepatic γδ T cells colocalized with hepatic stellate cells (HSCs) in vivo and promoted apoptosis of primary murine HSCs in a cell-cell contact-dependent manner, involving Fas-ligand (CD95L). Consistent with γδ T-cell-induced HSC apoptosis, activated myofibroblasts were more frequent in fibrotic livers of Ccr6(-/-) than in WT mice.CONCLUSION: γδ T cells are recruited to the liver by CCR6 upon chronic injury and protect the liver from excessive inflammation and fibrosis by inhibiting HSCs.

AB - UNLABELLED: Chronic liver injury promotes hepatic inflammation, representing a prerequisite for organ fibrosis. We hypothesized a contribution of chemokine receptor CCR6 and its ligand, CCL20, which may regulate migration of T-helper (Th)17, regulatory, and gamma-delta (γδ) T cells. CCR6 and CCL20 expression was intrahepatically up-regulated in patients with chronic liver diseases (n = 50), compared to control liver (n = 5). Immunohistochemistry revealed the periportal accumulation of CCR6(+) mononuclear cells and CCL20 induction by hepatic parenchymal cells in liver disease patients. Similarly, in murine livers, CCR6 was expressed by macrophages, CD4 and γδ T-cells, and up-regulated in fibrosis, whereas primary hepatocytes induced CCL20 upon experimental injury. In two murine models of chronic liver injury (CCl4 and methionine-choline-deficient diet), Ccr6(-/-) mice developed more severe fibrosis with strongly enhanced hepatic immune cell infiltration, compared to wild-type (WT) mice. Although CCR6 did not affect hepatic Th-cell subtype composition, CCR6 was explicitly required by the subset of interleukin (IL)-17- and IL-22-expressing γδ T cells for accumulation in injured liver. The adoptive transfer of WT γδ, but not CD4 T cells, into Ccr6(-/-) mice reduced hepatic inflammation and fibrosis in chronic injury to WT level. The anti-inflammatory function of hepatic γδ T cells was independent of IL-17, as evidenced by transfer of Il-17(-/-) cells. Instead, hepatic γδ T cells colocalized with hepatic stellate cells (HSCs) in vivo and promoted apoptosis of primary murine HSCs in a cell-cell contact-dependent manner, involving Fas-ligand (CD95L). Consistent with γδ T-cell-induced HSC apoptosis, activated myofibroblasts were more frequent in fibrotic livers of Ccr6(-/-) than in WT mice.CONCLUSION: γδ T cells are recruited to the liver by CCR6 upon chronic injury and protect the liver from excessive inflammation and fibrosis by inhibiting HSCs.

KW - Animals

KW - Apoptosis

KW - Case-Control Studies

KW - Cell Movement

KW - Chemokine CCL20/metabolism

KW - Disease Models, Animal

KW - Female

KW - Hepatitis/metabolism

KW - Humans

KW - Interleukin-17/metabolism

KW - Liver Cirrhosis/metabolism

KW - Liver Diseases/metabolism

KW - Male

KW - Mice, Inbred C57BL

KW - Mice, Knockout

KW - Receptors, Antigen, T-Cell, gamma-delta/metabolism

KW - Receptors, CCR6/deficiency

KW - T-Lymphocytes/metabolism

KW - T-Lymphocytes, Regulatory/metabolism

KW - Th17 Cells/metabolism

KW - Up-Regulation

U2 - 10.1002/hep.26697

DO - 10.1002/hep.26697

M3 - SCORING: Journal article

C2 - 23959575

VL - 59

SP - 630

EP - 642

JO - HEPATOLOGY

JF - HEPATOLOGY

SN - 0270-9139

IS - 2

ER -