Rapid Inflammasome Activation Is Attenuated in Post-Myocardial Infarction Monocytes

Standard

Rapid Inflammasome Activation Is Attenuated in Post-Myocardial Infarction Monocytes. / Giral, Hector; Franke, Vedran; Moobed, Minoo; Müller, Maja F; Lübking, Laura; James, Divya Maria; Hartung, Johannes; Kuschnerus, Kira; Meteva, Denitsa; Seppelt, Claudio; Jakob, Philipp; Klingenberg, Roland; Kränkel, Nicolle; Leistner, David; Zeller, Tanja; Blankenberg, Stefan; Zimmermann, Friederike; Haghikia, Arash; Lüscher, Thomas F; Akalin, Altuna; Landmesser, Ulf; Kratzer, Adelheid.

In: FRONT IMMUNOL, Vol. 13, 857455, 2022.

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

Harvard

Giral, H, Franke, V, Moobed, M, Müller, MF, Lübking, L, James, DM, Hartung, J, Kuschnerus, K, Meteva, D, Seppelt, C, Jakob, P, Klingenberg, R, Kränkel, N, Leistner, D, Zeller, T, Blankenberg, S, Zimmermann, F, Haghikia, A, Lüscher, TF, Akalin, A, Landmesser, U & Kratzer, A 2022, 'Rapid Inflammasome Activation Is Attenuated in Post-Myocardial Infarction Monocytes', FRONT IMMUNOL, vol. 13, 857455. https://doi.org/10.3389/fimmu.2022.857455

APA

Giral, H., Franke, V., Moobed, M., Müller, M. F., Lübking, L., James, D. M., Hartung, J., Kuschnerus, K., Meteva, D., Seppelt, C., Jakob, P., Klingenberg, R., Kränkel, N., Leistner, D., Zeller, T., Blankenberg, S., Zimmermann, F., Haghikia, A., Lüscher, T. F., ... Kratzer, A. (2022). Rapid Inflammasome Activation Is Attenuated in Post-Myocardial Infarction Monocytes. FRONT IMMUNOL, 13, [857455]. https://doi.org/10.3389/fimmu.2022.857455

Vancouver

Bibtex

@article{ec9f17e0279b4812b713d00361d59549,
title = "Rapid Inflammasome Activation Is Attenuated in Post-Myocardial Infarction Monocytes",
abstract = "Inflammasomes are crucial gatekeepers of the immune response, but their maladaptive activation associates with inflammatory pathologies. Besides canonical activation, monocytes can trigger non-transcriptional or rapid inflammasome activation that has not been well defined in the context of acute myocardial infarction (AMI). Rapid transcription-independent inflammasome activation induced by simultaneous TLR priming and triggering stimulus was measured by caspase-1 (CASP1) activity and interleukin release. Both classical and intermediate monocytes from healthy donors exhibited robust CASP1 activation, but only classical monocytes produced high mature interleukin-18 (IL18) release. We also recruited a limited number of coronary artery disease (CAD, n=31) and AMI (n=29) patients to evaluate their inflammasome function and expression profiles. Surprisingly, monocyte subpopulations isolated from blood collected during percutaneous coronary intervention (PCI) from AMI patients presented diminished CASP1 activity and abrogated IL18 release despite increased NLRP3 gene expression. This unexpected attenuated rapid inflammasome activation was accompanied by a significant increase of TNFAIP3 and IRAKM expression. Moreover, TNFAIP3 protein levels of circulating monocytes showed positive correlation with high sensitive troponin T (hsTnT), implying an association between TNFAIP3 upregulation and the severity of tissue injury. We suggest this monocyte attenuation to be a protective phenotype aftermath following a very early inflammatory wave in the ischemic area. Damage-associated molecular patterns (DAMPs) or other signals trigger a transitory negative feedback loop within newly recruited circulating monocytes as a mechanism to reduce post-injury tissue damage.",
keywords = "Humans, Inflammasomes/metabolism, Interleukin-18/metabolism, Interleukin-1beta/metabolism, Monocytes, Myocardial Infarction/pathology, NLR Family, Pyrin Domain-Containing 3 Protein/metabolism, Percutaneous Coronary Intervention",
author = "Hector Giral and Vedran Franke and Minoo Moobed and M{\"u}ller, {Maja F} and Laura L{\"u}bking and James, {Divya Maria} and Johannes Hartung and Kira Kuschnerus and Denitsa Meteva and Claudio Seppelt and Philipp Jakob and Roland Klingenberg and Nicolle Kr{\"a}nkel and David Leistner and Tanja Zeller and Stefan Blankenberg and Friederike Zimmermann and Arash Haghikia and L{\"u}scher, {Thomas F} and Altuna Akalin and Ulf Landmesser and Adelheid Kratzer",
note = "Copyright {\textcopyright} 2022 Giral, Franke, Moobed, M{\"u}ller, L{\"u}bking, James, Hartung, Kuschnerus, Meteva, Seppelt, Jakob, Klingenberg, Kr{\"a}nkel, Leistner, Zeller, Blankenberg, Zimmermann, Haghikia, L{\"u}scher, Akalin, Landmesser and Kratzer.",
year = "2022",
doi = "10.3389/fimmu.2022.857455",
language = "English",
volume = "13",
journal = "FRONT IMMUNOL",
issn = "1664-3224",
publisher = "Lausanne : Frontiers Research Foundation",

}

RIS

TY - JOUR

T1 - Rapid Inflammasome Activation Is Attenuated in Post-Myocardial Infarction Monocytes

AU - Giral, Hector

AU - Franke, Vedran

AU - Moobed, Minoo

AU - Müller, Maja F

AU - Lübking, Laura

AU - James, Divya Maria

AU - Hartung, Johannes

AU - Kuschnerus, Kira

AU - Meteva, Denitsa

AU - Seppelt, Claudio

AU - Jakob, Philipp

AU - Klingenberg, Roland

AU - Kränkel, Nicolle

AU - Leistner, David

AU - Zeller, Tanja

AU - Blankenberg, Stefan

AU - Zimmermann, Friederike

AU - Haghikia, Arash

AU - Lüscher, Thomas F

AU - Akalin, Altuna

AU - Landmesser, Ulf

AU - Kratzer, Adelheid

N1 - Copyright © 2022 Giral, Franke, Moobed, Müller, Lübking, James, Hartung, Kuschnerus, Meteva, Seppelt, Jakob, Klingenberg, Kränkel, Leistner, Zeller, Blankenberg, Zimmermann, Haghikia, Lüscher, Akalin, Landmesser and Kratzer.

PY - 2022

Y1 - 2022

N2 - Inflammasomes are crucial gatekeepers of the immune response, but their maladaptive activation associates with inflammatory pathologies. Besides canonical activation, monocytes can trigger non-transcriptional or rapid inflammasome activation that has not been well defined in the context of acute myocardial infarction (AMI). Rapid transcription-independent inflammasome activation induced by simultaneous TLR priming and triggering stimulus was measured by caspase-1 (CASP1) activity and interleukin release. Both classical and intermediate monocytes from healthy donors exhibited robust CASP1 activation, but only classical monocytes produced high mature interleukin-18 (IL18) release. We also recruited a limited number of coronary artery disease (CAD, n=31) and AMI (n=29) patients to evaluate their inflammasome function and expression profiles. Surprisingly, monocyte subpopulations isolated from blood collected during percutaneous coronary intervention (PCI) from AMI patients presented diminished CASP1 activity and abrogated IL18 release despite increased NLRP3 gene expression. This unexpected attenuated rapid inflammasome activation was accompanied by a significant increase of TNFAIP3 and IRAKM expression. Moreover, TNFAIP3 protein levels of circulating monocytes showed positive correlation with high sensitive troponin T (hsTnT), implying an association between TNFAIP3 upregulation and the severity of tissue injury. We suggest this monocyte attenuation to be a protective phenotype aftermath following a very early inflammatory wave in the ischemic area. Damage-associated molecular patterns (DAMPs) or other signals trigger a transitory negative feedback loop within newly recruited circulating monocytes as a mechanism to reduce post-injury tissue damage.

AB - Inflammasomes are crucial gatekeepers of the immune response, but their maladaptive activation associates with inflammatory pathologies. Besides canonical activation, monocytes can trigger non-transcriptional or rapid inflammasome activation that has not been well defined in the context of acute myocardial infarction (AMI). Rapid transcription-independent inflammasome activation induced by simultaneous TLR priming and triggering stimulus was measured by caspase-1 (CASP1) activity and interleukin release. Both classical and intermediate monocytes from healthy donors exhibited robust CASP1 activation, but only classical monocytes produced high mature interleukin-18 (IL18) release. We also recruited a limited number of coronary artery disease (CAD, n=31) and AMI (n=29) patients to evaluate their inflammasome function and expression profiles. Surprisingly, monocyte subpopulations isolated from blood collected during percutaneous coronary intervention (PCI) from AMI patients presented diminished CASP1 activity and abrogated IL18 release despite increased NLRP3 gene expression. This unexpected attenuated rapid inflammasome activation was accompanied by a significant increase of TNFAIP3 and IRAKM expression. Moreover, TNFAIP3 protein levels of circulating monocytes showed positive correlation with high sensitive troponin T (hsTnT), implying an association between TNFAIP3 upregulation and the severity of tissue injury. We suggest this monocyte attenuation to be a protective phenotype aftermath following a very early inflammatory wave in the ischemic area. Damage-associated molecular patterns (DAMPs) or other signals trigger a transitory negative feedback loop within newly recruited circulating monocytes as a mechanism to reduce post-injury tissue damage.

KW - Humans

KW - Inflammasomes/metabolism

KW - Interleukin-18/metabolism

KW - Interleukin-1beta/metabolism

KW - Monocytes

KW - Myocardial Infarction/pathology

KW - NLR Family, Pyrin Domain-Containing 3 Protein/metabolism

KW - Percutaneous Coronary Intervention

U2 - 10.3389/fimmu.2022.857455

DO - 10.3389/fimmu.2022.857455

M3 - SCORING: Journal article

C2 - 35558073

VL - 13

JO - FRONT IMMUNOL

JF - FRONT IMMUNOL

SN - 1664-3224

M1 - 857455

ER -