Role and cellular source of nicotinamide adenine dinucleotide phosphate oxidase in hepatic fibrosis.

Standard

Role and cellular source of nicotinamide adenine dinucleotide phosphate oxidase in hepatic fibrosis. / Samuele, De Minicis; Seki, Ekihiro; Paik, Yong-Han; Osterreicher, Christoph H; Kodama, Yuzo; Kluwe, Johannes; Torozzi, Luciano; Miyai, Katsumi; Benedetti, Antonio; Schwabe, Robert F; Brenner, David A.

in: HEPATOLOGY, Jahrgang 52, Nr. 4, 4, 2010, S. 1420-1430.

Publikationen: SCORING: Beitrag in Fachzeitschrift/ZeitungSCORING: ZeitschriftenaufsatzForschungBegutachtung

Harvard

Samuele, DM, Seki, E, Paik, Y-H, Osterreicher, CH, Kodama, Y, Kluwe, J, Torozzi, L, Miyai, K, Benedetti, A, Schwabe, RF & Brenner, DA 2010, 'Role and cellular source of nicotinamide adenine dinucleotide phosphate oxidase in hepatic fibrosis.', HEPATOLOGY, Jg. 52, Nr. 4, 4, S. 1420-1430. <http://www.ncbi.nlm.nih.gov/pubmed/20690191?dopt=Citation>

APA

Samuele, D. M., Seki, E., Paik, Y-H., Osterreicher, C. H., Kodama, Y., Kluwe, J., Torozzi, L., Miyai, K., Benedetti, A., Schwabe, R. F., & Brenner, D. A. (2010). Role and cellular source of nicotinamide adenine dinucleotide phosphate oxidase in hepatic fibrosis. HEPATOLOGY, 52(4), 1420-1430. [4]. http://www.ncbi.nlm.nih.gov/pubmed/20690191?dopt=Citation

Vancouver

Samuele DM, Seki E, Paik Y-H, Osterreicher CH, Kodama Y, Kluwe J et al. Role and cellular source of nicotinamide adenine dinucleotide phosphate oxidase in hepatic fibrosis. HEPATOLOGY. 2010;52(4):1420-1430. 4.

Bibtex

@article{ec3af8d0796f47579fbb255cc4343a24,
title = "Role and cellular source of nicotinamide adenine dinucleotide phosphate oxidase in hepatic fibrosis.",
abstract = "Reactive oxygen species (ROS) generated by nicotinamide adenine dinucleotide phosphate oxidase (NOX) is required for liver fibrosis. This study investigates the role of NOX in ROS production and the differential contribution of NOX from bone marrow (BM)-derived and non-BM-derived liver cells. Hepatic fibrosis was induced by bile duct ligation (BDL) for 21 days or by methionine-choline-deficient (MCD) diet for 10 weeks in wild-type (WT) mice and mice deficient in p47phox (p47phox knockout [KO]), a component of NOX. The p47phox KO chimeric mice were generated by the combination of liposomal clodronate injection, irradiation, and BM transplantation of p47phox KO BM into WT recipients and vice versa. Upon BDL, chimeric mice with p47phox KO BM-derived cells, including Kupffer cells, and WT endogenous liver cells showed a 25% reduction of fibrosis, whereas chimeric mice with WT BM-derived cells and p47phox KO endogenous liver cells, including hepatic stellate cells, showed a 60% reduction of fibrosis. In addition, p47phox KO compared to WT mice treated with an MCD diet showed no significant changes in steatosis and hepatocellular injury, but a 50% reduction in fibrosis. Cultured WT and p47phox KO hepatocytes treated with free fatty acids had a similar increase in lipid accumulation. Free fatty acids promoted a 1.5-fold increase in ROS production both in p47phox KO and in WT hepatocytes. CONCLUSION: NOX in both BM-derived and non-BM-derived cells contributes to liver fibrosis. NOX does not play a role in experimental steatosis and the generation of ROS in hepatocytes, but exerts a key role in fibrosis.",
keywords = "Animals, Male, Hepatic Stellate Cells enzymology, Mice, Mice, Inbred C57BL, Ligation, Mice, Knockout, Liver cytology, Bile Ducts surgery, Bone Marrow Transplantation, Carbon Tetrachloride Poisoning complications, Choline Deficiency physiopathology, Kupffer Cells enzymology, Lipid Peroxidation, Liver Cirrhosis enzymology, Methionine deficiency, NADPH Oxidase metabolism, Reactive Oxygen Species metabolism, Transplantation Chimera, Animals, Male, Hepatic Stellate Cells enzymology, Mice, Mice, Inbred C57BL, Ligation, Mice, Knockout, Liver cytology, Bile Ducts surgery, Bone Marrow Transplantation, Carbon Tetrachloride Poisoning complications, Choline Deficiency physiopathology, Kupffer Cells enzymology, Lipid Peroxidation, Liver Cirrhosis enzymology, Methionine deficiency, NADPH Oxidase metabolism, Reactive Oxygen Species metabolism, Transplantation Chimera",
author = "Samuele, {De Minicis} and Ekihiro Seki and Yong-Han Paik and Osterreicher, {Christoph H} and Yuzo Kodama and Johannes Kluwe and Luciano Torozzi and Katsumi Miyai and Antonio Benedetti and Schwabe, {Robert F} and Brenner, {David A}",
year = "2010",
language = "Deutsch",
volume = "52",
pages = "1420--1430",
journal = "HEPATOLOGY",
issn = "0270-9139",
publisher = "John Wiley and Sons Ltd",
number = "4",

}

RIS

TY - JOUR

T1 - Role and cellular source of nicotinamide adenine dinucleotide phosphate oxidase in hepatic fibrosis.

AU - Samuele, De Minicis

AU - Seki, Ekihiro

AU - Paik, Yong-Han

AU - Osterreicher, Christoph H

AU - Kodama, Yuzo

AU - Kluwe, Johannes

AU - Torozzi, Luciano

AU - Miyai, Katsumi

AU - Benedetti, Antonio

AU - Schwabe, Robert F

AU - Brenner, David A

PY - 2010

Y1 - 2010

N2 - Reactive oxygen species (ROS) generated by nicotinamide adenine dinucleotide phosphate oxidase (NOX) is required for liver fibrosis. This study investigates the role of NOX in ROS production and the differential contribution of NOX from bone marrow (BM)-derived and non-BM-derived liver cells. Hepatic fibrosis was induced by bile duct ligation (BDL) for 21 days or by methionine-choline-deficient (MCD) diet for 10 weeks in wild-type (WT) mice and mice deficient in p47phox (p47phox knockout [KO]), a component of NOX. The p47phox KO chimeric mice were generated by the combination of liposomal clodronate injection, irradiation, and BM transplantation of p47phox KO BM into WT recipients and vice versa. Upon BDL, chimeric mice with p47phox KO BM-derived cells, including Kupffer cells, and WT endogenous liver cells showed a 25% reduction of fibrosis, whereas chimeric mice with WT BM-derived cells and p47phox KO endogenous liver cells, including hepatic stellate cells, showed a 60% reduction of fibrosis. In addition, p47phox KO compared to WT mice treated with an MCD diet showed no significant changes in steatosis and hepatocellular injury, but a 50% reduction in fibrosis. Cultured WT and p47phox KO hepatocytes treated with free fatty acids had a similar increase in lipid accumulation. Free fatty acids promoted a 1.5-fold increase in ROS production both in p47phox KO and in WT hepatocytes. CONCLUSION: NOX in both BM-derived and non-BM-derived cells contributes to liver fibrosis. NOX does not play a role in experimental steatosis and the generation of ROS in hepatocytes, but exerts a key role in fibrosis.

AB - Reactive oxygen species (ROS) generated by nicotinamide adenine dinucleotide phosphate oxidase (NOX) is required for liver fibrosis. This study investigates the role of NOX in ROS production and the differential contribution of NOX from bone marrow (BM)-derived and non-BM-derived liver cells. Hepatic fibrosis was induced by bile duct ligation (BDL) for 21 days or by methionine-choline-deficient (MCD) diet for 10 weeks in wild-type (WT) mice and mice deficient in p47phox (p47phox knockout [KO]), a component of NOX. The p47phox KO chimeric mice were generated by the combination of liposomal clodronate injection, irradiation, and BM transplantation of p47phox KO BM into WT recipients and vice versa. Upon BDL, chimeric mice with p47phox KO BM-derived cells, including Kupffer cells, and WT endogenous liver cells showed a 25% reduction of fibrosis, whereas chimeric mice with WT BM-derived cells and p47phox KO endogenous liver cells, including hepatic stellate cells, showed a 60% reduction of fibrosis. In addition, p47phox KO compared to WT mice treated with an MCD diet showed no significant changes in steatosis and hepatocellular injury, but a 50% reduction in fibrosis. Cultured WT and p47phox KO hepatocytes treated with free fatty acids had a similar increase in lipid accumulation. Free fatty acids promoted a 1.5-fold increase in ROS production both in p47phox KO and in WT hepatocytes. CONCLUSION: NOX in both BM-derived and non-BM-derived cells contributes to liver fibrosis. NOX does not play a role in experimental steatosis and the generation of ROS in hepatocytes, but exerts a key role in fibrosis.

KW - Animals

KW - Male

KW - Hepatic Stellate Cells enzymology

KW - Mice

KW - Mice, Inbred C57BL

KW - Ligation

KW - Mice, Knockout

KW - Liver cytology

KW - Bile Ducts surgery

KW - Bone Marrow Transplantation

KW - Carbon Tetrachloride Poisoning complications

KW - Choline Deficiency physiopathology

KW - Kupffer Cells enzymology

KW - Lipid Peroxidation

KW - Liver Cirrhosis enzymology

KW - Methionine deficiency

KW - NADPH Oxidase metabolism

KW - Reactive Oxygen Species metabolism

KW - Transplantation Chimera

KW - Animals

KW - Male

KW - Hepatic Stellate Cells enzymology

KW - Mice

KW - Mice, Inbred C57BL

KW - Ligation

KW - Mice, Knockout

KW - Liver cytology

KW - Bile Ducts surgery

KW - Bone Marrow Transplantation

KW - Carbon Tetrachloride Poisoning complications

KW - Choline Deficiency physiopathology

KW - Kupffer Cells enzymology

KW - Lipid Peroxidation

KW - Liver Cirrhosis enzymology

KW - Methionine deficiency

KW - NADPH Oxidase metabolism

KW - Reactive Oxygen Species metabolism

KW - Transplantation Chimera

M3 - SCORING: Zeitschriftenaufsatz

VL - 52

SP - 1420

EP - 1430

JO - HEPATOLOGY

JF - HEPATOLOGY

SN - 0270-9139

IS - 4

M1 - 4

ER -