Endotoxinemia Accelerates Atherosclerosis Through Electrostatic Charge-Mediated Monocyte Adhesion

Standard

Endotoxinemia Accelerates Atherosclerosis Through Electrostatic Charge-Mediated Monocyte Adhesion. / Schumski, Ariane; Ortega-Gómez, Almudena; Wichapong, Kanin; Winter, Carla; Lemnitzer, Patricia; Viola, Joana R; Pinilla-Vera, Mayra; Folco, Eduardo; Solis-Mezarino, Victor; Völker-Albert, Moritz; Maas, Sanne L; Pan, Chang; Perez Olivares, Laura; Winter, Janine; Hackeng, Tilman; Karlsson, Mikael C I; Zeller, Tanja; Imhof, Axel; Baron, Rebecca M; Nicolaes, Gerry A F; Libby, Peter; Maegdefessel, Lars; Kamp, Frits; Benoit, Martin; Döring, Yvonne; Soehnlein, Oliver.

in: CIRCULATION, Jahrgang 143, Nr. 3, 19.01.2021, S. 254-266.

Publikationen: SCORING: Beitrag in Fachzeitschrift/ZeitungSCORING: ZeitschriftenaufsatzForschungBegutachtung

Harvard

Schumski, A, Ortega-Gómez, A, Wichapong, K, Winter, C, Lemnitzer, P, Viola, JR, Pinilla-Vera, M, Folco, E, Solis-Mezarino, V, Völker-Albert, M, Maas, SL, Pan, C, Perez Olivares, L, Winter, J, Hackeng, T, Karlsson, MCI, Zeller, T, Imhof, A, Baron, RM, Nicolaes, GAF, Libby, P, Maegdefessel, L, Kamp, F, Benoit, M, Döring, Y & Soehnlein, O 2021, 'Endotoxinemia Accelerates Atherosclerosis Through Electrostatic Charge-Mediated Monocyte Adhesion', CIRCULATION, Jg. 143, Nr. 3, S. 254-266. https://doi.org/10.1161/CIRCULATIONAHA.120.046677

APA

Schumski, A., Ortega-Gómez, A., Wichapong, K., Winter, C., Lemnitzer, P., Viola, J. R., Pinilla-Vera, M., Folco, E., Solis-Mezarino, V., Völker-Albert, M., Maas, S. L., Pan, C., Perez Olivares, L., Winter, J., Hackeng, T., Karlsson, M. C. I., Zeller, T., Imhof, A., Baron, R. M., ... Soehnlein, O. (2021). Endotoxinemia Accelerates Atherosclerosis Through Electrostatic Charge-Mediated Monocyte Adhesion. CIRCULATION, 143(3), 254-266. https://doi.org/10.1161/CIRCULATIONAHA.120.046677

Vancouver

Schumski A, Ortega-Gómez A, Wichapong K, Winter C, Lemnitzer P, Viola JR et al. Endotoxinemia Accelerates Atherosclerosis Through Electrostatic Charge-Mediated Monocyte Adhesion. CIRCULATION. 2021 Jan 19;143(3):254-266. https://doi.org/10.1161/CIRCULATIONAHA.120.046677

Bibtex

@article{3925116eefa94ce0b830d348eb7406f2,
title = "Endotoxinemia Accelerates Atherosclerosis Through Electrostatic Charge-Mediated Monocyte Adhesion",
abstract = "BACKGROUND: Acute infection is a well-established risk factor of cardiovascular inflammation increasing the risk for a cardiovascular complication within the first weeks after infection. However, the nature of the processes underlying such aggravation remains unclear. Lipopolysaccharide derived from Gram-negative bacteria is a potent activator of circulating immune cells including neutrophils, which foster inflammation through discharge of neutrophil extracellular traps (NETs). Here, we use a model of endotoxinemia to link acute infection and subsequent neutrophil activation with acceleration of vascular inflammation Methods: Acute infection was mimicked by injection of a single dose of lipopolysaccharide into hypercholesterolemic mice. Atherosclerosis burden was studied by histomorphometric analysis of the aortic root. Arterial myeloid cell adhesion was quantified by intravital microscopy.RESULTS: Lipopolysaccharide treatment rapidly enhanced atherosclerotic lesion size by expansion of the lesional myeloid cell accumulation. Lipopolysaccharide treatment led to the deposition of NETs along the arterial lumen, and inhibition of NET release annulled lesion expansion during endotoxinemia, thus suggesting that NETs regulate myeloid cell recruitment. To study the mechanism of monocyte adhesion to NETs, we used in vitro adhesion assays and biophysical approaches. In these experiments, NET-resident histone H2a attracted monocytes in a receptor-independent, surface charge-dependent fashion. Therapeutic neutralization of histone H2a by antibodies or by in silico designed cyclic peptides enables us to reduce luminal monocyte adhesion and lesion expansion during endotoxinemia.CONCLUSIONS: Our study shows that NET-associated histone H2a mediates charge-dependent monocyte adhesion to NETs and accelerates atherosclerosis during endotoxinemia.",
author = "Ariane Schumski and Almudena Ortega-G{\'o}mez and Kanin Wichapong and Carla Winter and Patricia Lemnitzer and Viola, {Joana R} and Mayra Pinilla-Vera and Eduardo Folco and Victor Solis-Mezarino and Moritz V{\"o}lker-Albert and Maas, {Sanne L} and Chang Pan and {Perez Olivares}, Laura and Janine Winter and Tilman Hackeng and Karlsson, {Mikael C I} and Tanja Zeller and Axel Imhof and Baron, {Rebecca M} and Nicolaes, {Gerry A F} and Peter Libby and Lars Maegdefessel and Frits Kamp and Martin Benoit and Yvonne D{\"o}ring and Oliver Soehnlein",
year = "2021",
month = jan,
day = "19",
doi = "10.1161/CIRCULATIONAHA.120.046677",
language = "English",
volume = "143",
pages = "254--266",
journal = "CIRCULATION",
issn = "0009-7322",
publisher = "Lippincott Williams and Wilkins",
number = "3",

}

RIS

TY - JOUR

T1 - Endotoxinemia Accelerates Atherosclerosis Through Electrostatic Charge-Mediated Monocyte Adhesion

AU - Schumski, Ariane

AU - Ortega-Gómez, Almudena

AU - Wichapong, Kanin

AU - Winter, Carla

AU - Lemnitzer, Patricia

AU - Viola, Joana R

AU - Pinilla-Vera, Mayra

AU - Folco, Eduardo

AU - Solis-Mezarino, Victor

AU - Völker-Albert, Moritz

AU - Maas, Sanne L

AU - Pan, Chang

AU - Perez Olivares, Laura

AU - Winter, Janine

AU - Hackeng, Tilman

AU - Karlsson, Mikael C I

AU - Zeller, Tanja

AU - Imhof, Axel

AU - Baron, Rebecca M

AU - Nicolaes, Gerry A F

AU - Libby, Peter

AU - Maegdefessel, Lars

AU - Kamp, Frits

AU - Benoit, Martin

AU - Döring, Yvonne

AU - Soehnlein, Oliver

PY - 2021/1/19

Y1 - 2021/1/19

N2 - BACKGROUND: Acute infection is a well-established risk factor of cardiovascular inflammation increasing the risk for a cardiovascular complication within the first weeks after infection. However, the nature of the processes underlying such aggravation remains unclear. Lipopolysaccharide derived from Gram-negative bacteria is a potent activator of circulating immune cells including neutrophils, which foster inflammation through discharge of neutrophil extracellular traps (NETs). Here, we use a model of endotoxinemia to link acute infection and subsequent neutrophil activation with acceleration of vascular inflammation Methods: Acute infection was mimicked by injection of a single dose of lipopolysaccharide into hypercholesterolemic mice. Atherosclerosis burden was studied by histomorphometric analysis of the aortic root. Arterial myeloid cell adhesion was quantified by intravital microscopy.RESULTS: Lipopolysaccharide treatment rapidly enhanced atherosclerotic lesion size by expansion of the lesional myeloid cell accumulation. Lipopolysaccharide treatment led to the deposition of NETs along the arterial lumen, and inhibition of NET release annulled lesion expansion during endotoxinemia, thus suggesting that NETs regulate myeloid cell recruitment. To study the mechanism of monocyte adhesion to NETs, we used in vitro adhesion assays and biophysical approaches. In these experiments, NET-resident histone H2a attracted monocytes in a receptor-independent, surface charge-dependent fashion. Therapeutic neutralization of histone H2a by antibodies or by in silico designed cyclic peptides enables us to reduce luminal monocyte adhesion and lesion expansion during endotoxinemia.CONCLUSIONS: Our study shows that NET-associated histone H2a mediates charge-dependent monocyte adhesion to NETs and accelerates atherosclerosis during endotoxinemia.

AB - BACKGROUND: Acute infection is a well-established risk factor of cardiovascular inflammation increasing the risk for a cardiovascular complication within the first weeks after infection. However, the nature of the processes underlying such aggravation remains unclear. Lipopolysaccharide derived from Gram-negative bacteria is a potent activator of circulating immune cells including neutrophils, which foster inflammation through discharge of neutrophil extracellular traps (NETs). Here, we use a model of endotoxinemia to link acute infection and subsequent neutrophil activation with acceleration of vascular inflammation Methods: Acute infection was mimicked by injection of a single dose of lipopolysaccharide into hypercholesterolemic mice. Atherosclerosis burden was studied by histomorphometric analysis of the aortic root. Arterial myeloid cell adhesion was quantified by intravital microscopy.RESULTS: Lipopolysaccharide treatment rapidly enhanced atherosclerotic lesion size by expansion of the lesional myeloid cell accumulation. Lipopolysaccharide treatment led to the deposition of NETs along the arterial lumen, and inhibition of NET release annulled lesion expansion during endotoxinemia, thus suggesting that NETs regulate myeloid cell recruitment. To study the mechanism of monocyte adhesion to NETs, we used in vitro adhesion assays and biophysical approaches. In these experiments, NET-resident histone H2a attracted monocytes in a receptor-independent, surface charge-dependent fashion. Therapeutic neutralization of histone H2a by antibodies or by in silico designed cyclic peptides enables us to reduce luminal monocyte adhesion and lesion expansion during endotoxinemia.CONCLUSIONS: Our study shows that NET-associated histone H2a mediates charge-dependent monocyte adhesion to NETs and accelerates atherosclerosis during endotoxinemia.

U2 - 10.1161/CIRCULATIONAHA.120.046677

DO - 10.1161/CIRCULATIONAHA.120.046677

M3 - SCORING: Journal article

C2 - 33167684

VL - 143

SP - 254

EP - 266

JO - CIRCULATION

JF - CIRCULATION

SN - 0009-7322

IS - 3

ER -