Cathepsin S inhibition combines control of systemic and peripheral pathomechanisms of autoimmune tissue injury

Standard

Cathepsin S inhibition combines control of systemic and peripheral pathomechanisms of autoimmune tissue injury. / Tato, Maia; Kumar, Santhosh V; Liu, Yajuan; Mulay, Shrikant R; Moll, Solange; Popper, Bastian; Eberhard, Jonathan N; Thomasova, Dana; Rufer, Arne Christian; Gruner, Sabine; Haap, Wolfgang; Hartmann, Guido; Anders, Hans-Joachim.

In: SCI REP-UK, Vol. 7, No. 1, 05.06.2017, p. 2775.

Research output: SCORING: Contribution to journalSCORING: Journal articleResearchpeer-review

Harvard

Tato, M, Kumar, SV, Liu, Y, Mulay, SR, Moll, S, Popper, B, Eberhard, JN, Thomasova, D, Rufer, AC, Gruner, S, Haap, W, Hartmann, G & Anders, H-J 2017, 'Cathepsin S inhibition combines control of systemic and peripheral pathomechanisms of autoimmune tissue injury', SCI REP-UK, vol. 7, no. 1, pp. 2775. https://doi.org/10.1038/s41598-017-01894-y

APA

Tato, M., Kumar, S. V., Liu, Y., Mulay, S. R., Moll, S., Popper, B., Eberhard, J. N., Thomasova, D., Rufer, A. C., Gruner, S., Haap, W., Hartmann, G., & Anders, H-J. (2017). Cathepsin S inhibition combines control of systemic and peripheral pathomechanisms of autoimmune tissue injury. SCI REP-UK, 7(1), 2775. https://doi.org/10.1038/s41598-017-01894-y

Vancouver

Bibtex

@article{cdbb365dadeb4523b107b8db9c55e246,
title = "Cathepsin S inhibition combines control of systemic and peripheral pathomechanisms of autoimmune tissue injury",
abstract = "Cathepsin(Cat)-S processing of the invariant chain-MHC-II complex inside antigen presenting cells is a central pathomechanism of autoimmune-diseases. Additionally, Cat-S is released by activated-myeloid cells and was recently described to activate protease-activated-receptor-(PAR)-2 in extracellular compartments. We hypothesized that Cat-S blockade targets both mechanisms and elicits synergistic therapeutic effects on autoimmune tissue injury. MRL-(Fas)lpr mice with spontaneous autoimmune tissue injury were treated with different doses of Cat-S inhibitor RO5459072, mycophenolate mofetil or vehicle. Further, female MRL-(Fas)lpr mice were injected with recombinant Cat-S with/without concomitant Cat-S or PAR-2 blockade. Cat-S blockade dose-dependently reversed aberrant systemic autoimmunity, e.g. plasma cytokines, activation of myeloid cells and hypergammaglobulinemia. Especially IgG autoantibody production was suppressed. Of note (MHC-II-independent) IgM were unaffected by Cat-S blockade while they were suppressed by MMF. Cat-S blockade dose-dependently suppressed immune-complex glomerulonephritis together with a profound and early effect on proteinuria, which was not shared by MMF. In fact, intravenous Cat-S injection induced severe glomerular endothelial injury and albuminuria, which was entirely prevented by Cat-S or PAR-2 blockade. In-vitro studies confirm that Cat-S induces endothelial activation and injury via PAR-2. Therapeutic Cat-S blockade suppresses systemic and peripheral pathomechanisms of autoimmune tissue injury, hence, Cat-S is a promising therapeutic target in lupus nephritis.",
keywords = "Journal Article",
author = "Maia Tato and Kumar, {Santhosh V} and Yajuan Liu and Mulay, {Shrikant R} and Solange Moll and Bastian Popper and Eberhard, {Jonathan N} and Dana Thomasova and Rufer, {Arne Christian} and Sabine Gruner and Wolfgang Haap and Guido Hartmann and Hans-Joachim Anders",
year = "2017",
month = jun,
day = "5",
doi = "10.1038/s41598-017-01894-y",
language = "English",
volume = "7",
pages = "2775",
journal = "SCI REP-UK",
issn = "2045-2322",
publisher = "NATURE PUBLISHING GROUP",
number = "1",

}

RIS

TY - JOUR

T1 - Cathepsin S inhibition combines control of systemic and peripheral pathomechanisms of autoimmune tissue injury

AU - Tato, Maia

AU - Kumar, Santhosh V

AU - Liu, Yajuan

AU - Mulay, Shrikant R

AU - Moll, Solange

AU - Popper, Bastian

AU - Eberhard, Jonathan N

AU - Thomasova, Dana

AU - Rufer, Arne Christian

AU - Gruner, Sabine

AU - Haap, Wolfgang

AU - Hartmann, Guido

AU - Anders, Hans-Joachim

PY - 2017/6/5

Y1 - 2017/6/5

N2 - Cathepsin(Cat)-S processing of the invariant chain-MHC-II complex inside antigen presenting cells is a central pathomechanism of autoimmune-diseases. Additionally, Cat-S is released by activated-myeloid cells and was recently described to activate protease-activated-receptor-(PAR)-2 in extracellular compartments. We hypothesized that Cat-S blockade targets both mechanisms and elicits synergistic therapeutic effects on autoimmune tissue injury. MRL-(Fas)lpr mice with spontaneous autoimmune tissue injury were treated with different doses of Cat-S inhibitor RO5459072, mycophenolate mofetil or vehicle. Further, female MRL-(Fas)lpr mice were injected with recombinant Cat-S with/without concomitant Cat-S or PAR-2 blockade. Cat-S blockade dose-dependently reversed aberrant systemic autoimmunity, e.g. plasma cytokines, activation of myeloid cells and hypergammaglobulinemia. Especially IgG autoantibody production was suppressed. Of note (MHC-II-independent) IgM were unaffected by Cat-S blockade while they were suppressed by MMF. Cat-S blockade dose-dependently suppressed immune-complex glomerulonephritis together with a profound and early effect on proteinuria, which was not shared by MMF. In fact, intravenous Cat-S injection induced severe glomerular endothelial injury and albuminuria, which was entirely prevented by Cat-S or PAR-2 blockade. In-vitro studies confirm that Cat-S induces endothelial activation and injury via PAR-2. Therapeutic Cat-S blockade suppresses systemic and peripheral pathomechanisms of autoimmune tissue injury, hence, Cat-S is a promising therapeutic target in lupus nephritis.

AB - Cathepsin(Cat)-S processing of the invariant chain-MHC-II complex inside antigen presenting cells is a central pathomechanism of autoimmune-diseases. Additionally, Cat-S is released by activated-myeloid cells and was recently described to activate protease-activated-receptor-(PAR)-2 in extracellular compartments. We hypothesized that Cat-S blockade targets both mechanisms and elicits synergistic therapeutic effects on autoimmune tissue injury. MRL-(Fas)lpr mice with spontaneous autoimmune tissue injury were treated with different doses of Cat-S inhibitor RO5459072, mycophenolate mofetil or vehicle. Further, female MRL-(Fas)lpr mice were injected with recombinant Cat-S with/without concomitant Cat-S or PAR-2 blockade. Cat-S blockade dose-dependently reversed aberrant systemic autoimmunity, e.g. plasma cytokines, activation of myeloid cells and hypergammaglobulinemia. Especially IgG autoantibody production was suppressed. Of note (MHC-II-independent) IgM were unaffected by Cat-S blockade while they were suppressed by MMF. Cat-S blockade dose-dependently suppressed immune-complex glomerulonephritis together with a profound and early effect on proteinuria, which was not shared by MMF. In fact, intravenous Cat-S injection induced severe glomerular endothelial injury and albuminuria, which was entirely prevented by Cat-S or PAR-2 blockade. In-vitro studies confirm that Cat-S induces endothelial activation and injury via PAR-2. Therapeutic Cat-S blockade suppresses systemic and peripheral pathomechanisms of autoimmune tissue injury, hence, Cat-S is a promising therapeutic target in lupus nephritis.

KW - Journal Article

U2 - 10.1038/s41598-017-01894-y

DO - 10.1038/s41598-017-01894-y

M3 - SCORING: Journal article

C2 - 28584258

VL - 7

SP - 2775

JO - SCI REP-UK

JF - SCI REP-UK

SN - 2045-2322

IS - 1

ER -