Aggregation of thrombin-derived C-terminal fragments as a previously undisclosed host defense mechanism

Research output: Contribution to journalJournal articleResearchpeer-review

Standard

Aggregation of thrombin-derived C-terminal fragments as a previously undisclosed host defense mechanism. / Petrlova, Jitka; Hansen, Finja C; van der Plas, Mariena J A; Huber, Roland G; Mörgelin, Matthias; Malmsten, Martin; Bond, Peter J; Schmidtchen, Artur.

In: Proceedings of the National Academy of Sciences of the United States of America, Vol. 114, No. 21, 23.05.2017, p. E4213-E4222.

Research output: Contribution to journalJournal articleResearchpeer-review

Harvard

Petrlova, J, Hansen, FC, van der Plas, MJA, Huber, RG, Mörgelin, M, Malmsten, M, Bond, PJ & Schmidtchen, A 2017, 'Aggregation of thrombin-derived C-terminal fragments as a previously undisclosed host defense mechanism', Proceedings of the National Academy of Sciences of the United States of America, vol. 114, no. 21, pp. E4213-E4222. https://doi.org/10.1073/pnas.1619609114

APA

Petrlova, J., Hansen, F. C., van der Plas, M. J. A., Huber, R. G., Mörgelin, M., Malmsten, M., Bond, P. J., & Schmidtchen, A. (2017). Aggregation of thrombin-derived C-terminal fragments as a previously undisclosed host defense mechanism. Proceedings of the National Academy of Sciences of the United States of America, 114(21), E4213-E4222. https://doi.org/10.1073/pnas.1619609114

Vancouver

Petrlova J, Hansen FC, van der Plas MJA, Huber RG, Mörgelin M, Malmsten M et al. Aggregation of thrombin-derived C-terminal fragments as a previously undisclosed host defense mechanism. Proceedings of the National Academy of Sciences of the United States of America. 2017 May 23;114(21):E4213-E4222. https://doi.org/10.1073/pnas.1619609114

Author

Petrlova, Jitka ; Hansen, Finja C ; van der Plas, Mariena J A ; Huber, Roland G ; Mörgelin, Matthias ; Malmsten, Martin ; Bond, Peter J ; Schmidtchen, Artur. / Aggregation of thrombin-derived C-terminal fragments as a previously undisclosed host defense mechanism. In: Proceedings of the National Academy of Sciences of the United States of America. 2017 ; Vol. 114, No. 21. pp. E4213-E4222.

Bibtex

@article{03feea1a629042fbacd4e4def1f0fa6e,
title = "Aggregation of thrombin-derived C-terminal fragments as a previously undisclosed host defense mechanism",
abstract = "Effective control of endotoxins and bacteria is crucial for normal wound healing. During injury, the key enzyme thrombin is formed, leading to generation of fibrin. Here, we show that human neutrophil elastase cleaves thrombin, generating 11-kDa thrombin-derived C-terminal peptides (TCPs), which bind to and form amorphous amyloid-like aggregates with both bacterial lipopolysaccharide (LPS) and gram-negative bacteria. In silico molecular modeling using atomic resolution and coarse-grained simulations corroborates our experimental observations, altogether indicating increased aggregation through LPS-mediated intermolecular contacts between clusters of TCP molecules. Upon bacterial aggregation, recombinantly produced TCPs induce permeabilization of Escherichia coli and phagocytic uptake. TCPs of about 11 kDa are present in acute wound fluids as well as in fibrin sloughs from patients with infected wounds. We noted aggregation and colocalization of LPS with TCPs in such fibrin material, which indicates the presence of TCP-LPS aggregates under physiological conditions. Apart from identifying a function of proteolyzed thrombin and its fragments, our findings provide an interesting link between the coagulation system, innate immunity, LPS scavenging, and protein aggregation/amyloid formation.",
keywords = "Journal Article",
author = "Jitka Petrlova and Hansen, {Finja C} and {van der Plas}, {Mariena J A} and Huber, {Roland G} and Matthias M{\"o}rgelin and Martin Malmsten and Bond, {Peter J} and Artur Schmidtchen",
year = "2017",
month = may,
day = "23",
doi = "10.1073/pnas.1619609114",
language = "English",
volume = "114",
pages = "E4213--E4222",
journal = "Proceedings of the National Academy of Sciences of the United States of America",
issn = "0027-8424",
publisher = "The National Academy of Sciences of the United States of America",
number = "21",

}

RIS

TY - JOUR

T1 - Aggregation of thrombin-derived C-terminal fragments as a previously undisclosed host defense mechanism

AU - Petrlova, Jitka

AU - Hansen, Finja C

AU - van der Plas, Mariena J A

AU - Huber, Roland G

AU - Mörgelin, Matthias

AU - Malmsten, Martin

AU - Bond, Peter J

AU - Schmidtchen, Artur

PY - 2017/5/23

Y1 - 2017/5/23

N2 - Effective control of endotoxins and bacteria is crucial for normal wound healing. During injury, the key enzyme thrombin is formed, leading to generation of fibrin. Here, we show that human neutrophil elastase cleaves thrombin, generating 11-kDa thrombin-derived C-terminal peptides (TCPs), which bind to and form amorphous amyloid-like aggregates with both bacterial lipopolysaccharide (LPS) and gram-negative bacteria. In silico molecular modeling using atomic resolution and coarse-grained simulations corroborates our experimental observations, altogether indicating increased aggregation through LPS-mediated intermolecular contacts between clusters of TCP molecules. Upon bacterial aggregation, recombinantly produced TCPs induce permeabilization of Escherichia coli and phagocytic uptake. TCPs of about 11 kDa are present in acute wound fluids as well as in fibrin sloughs from patients with infected wounds. We noted aggregation and colocalization of LPS with TCPs in such fibrin material, which indicates the presence of TCP-LPS aggregates under physiological conditions. Apart from identifying a function of proteolyzed thrombin and its fragments, our findings provide an interesting link between the coagulation system, innate immunity, LPS scavenging, and protein aggregation/amyloid formation.

AB - Effective control of endotoxins and bacteria is crucial for normal wound healing. During injury, the key enzyme thrombin is formed, leading to generation of fibrin. Here, we show that human neutrophil elastase cleaves thrombin, generating 11-kDa thrombin-derived C-terminal peptides (TCPs), which bind to and form amorphous amyloid-like aggregates with both bacterial lipopolysaccharide (LPS) and gram-negative bacteria. In silico molecular modeling using atomic resolution and coarse-grained simulations corroborates our experimental observations, altogether indicating increased aggregation through LPS-mediated intermolecular contacts between clusters of TCP molecules. Upon bacterial aggregation, recombinantly produced TCPs induce permeabilization of Escherichia coli and phagocytic uptake. TCPs of about 11 kDa are present in acute wound fluids as well as in fibrin sloughs from patients with infected wounds. We noted aggregation and colocalization of LPS with TCPs in such fibrin material, which indicates the presence of TCP-LPS aggregates under physiological conditions. Apart from identifying a function of proteolyzed thrombin and its fragments, our findings provide an interesting link between the coagulation system, innate immunity, LPS scavenging, and protein aggregation/amyloid formation.

KW - Journal Article

U2 - 10.1073/pnas.1619609114

DO - 10.1073/pnas.1619609114

M3 - Journal article

C2 - 28473418

VL - 114

SP - E4213-E4222

JO - Proceedings of the National Academy of Sciences of the United States of America

JF - Proceedings of the National Academy of Sciences of the United States of America

SN - 0027-8424

IS - 21

ER -

ID: 185031218