Salt Transiently Inhibits Mitochondrial Energetics in Mononuclear Phagocytes

Research output: Contribution to journalJournal articleResearchpeer-review

Standard

Salt Transiently Inhibits Mitochondrial Energetics in Mononuclear Phagocytes. / Geisberger, Sabrina; Bartolomaeus, Hendrik; Neubert, Patrick; Willebrand, Ralf; Zasada, Christin; Bartolomaeus, Thomas; McParland, Victoria; Swinnen, Dries; Geuzens, Anneleen; Maifeld, András; Krampert, Luka; Vogl, Marion; Mähler, Anja; Wilck, Nicola; Marko, Lajos; Tilic, Ekin; Forslund, Sofia K; Binger, Katrina J; Stegbauer, Johannes; Dechend, Ralf; Kleinewietfeld, Markus; Jantsch, Jonathan; Kempa, Stefan; Müller, Dominik N.

In: Circulation, Vol. 144, No. 2, 2021, p. 144–158.

Research output: Contribution to journalJournal articleResearchpeer-review

Harvard

Geisberger, S, Bartolomaeus, H, Neubert, P, Willebrand, R, Zasada, C, Bartolomaeus, T, McParland, V, Swinnen, D, Geuzens, A, Maifeld, A, Krampert, L, Vogl, M, Mähler, A, Wilck, N, Marko, L, Tilic, E, Forslund, SK, Binger, KJ, Stegbauer, J, Dechend, R, Kleinewietfeld, M, Jantsch, J, Kempa, S & Müller, DN 2021, 'Salt Transiently Inhibits Mitochondrial Energetics in Mononuclear Phagocytes', Circulation, vol. 144, no. 2, pp. 144–158. https://doi.org/10.1161/CIRCULATIONAHA.120.052788

APA

Geisberger, S., Bartolomaeus, H., Neubert, P., Willebrand, R., Zasada, C., Bartolomaeus, T., McParland, V., Swinnen, D., Geuzens, A., Maifeld, A., Krampert, L., Vogl, M., Mähler, A., Wilck, N., Marko, L., Tilic, E., Forslund, S. K., Binger, K. J., Stegbauer, J., ... Müller, D. N. (2021). Salt Transiently Inhibits Mitochondrial Energetics in Mononuclear Phagocytes. Circulation, 144(2), 144–158. https://doi.org/10.1161/CIRCULATIONAHA.120.052788

Vancouver

Geisberger S, Bartolomaeus H, Neubert P, Willebrand R, Zasada C, Bartolomaeus T et al. Salt Transiently Inhibits Mitochondrial Energetics in Mononuclear Phagocytes. Circulation. 2021;144(2):144–158. https://doi.org/10.1161/CIRCULATIONAHA.120.052788

Author

Geisberger, Sabrina ; Bartolomaeus, Hendrik ; Neubert, Patrick ; Willebrand, Ralf ; Zasada, Christin ; Bartolomaeus, Thomas ; McParland, Victoria ; Swinnen, Dries ; Geuzens, Anneleen ; Maifeld, András ; Krampert, Luka ; Vogl, Marion ; Mähler, Anja ; Wilck, Nicola ; Marko, Lajos ; Tilic, Ekin ; Forslund, Sofia K ; Binger, Katrina J ; Stegbauer, Johannes ; Dechend, Ralf ; Kleinewietfeld, Markus ; Jantsch, Jonathan ; Kempa, Stefan ; Müller, Dominik N. / Salt Transiently Inhibits Mitochondrial Energetics in Mononuclear Phagocytes. In: Circulation. 2021 ; Vol. 144, No. 2. pp. 144–158.

Bibtex

@article{1e585dc666824065a6bfeeb0dd0c1faa,
title = "Salt Transiently Inhibits Mitochondrial Energetics in Mononuclear Phagocytes",
abstract = "Background: Dietary high salt (HS) is a leading risk factor for mortality and morbidity. Serum sodium transiently increases postprandially, but can also accumulate at sites of inflammation affecting differentiation and function of innate and adaptive immune cells. Here, we focus on how changes in extracellular sodium, mimicking alterations in the circulation and tissues, affect the early metabolic, transcriptional and functional adaption of human and murine mononuclear phagocytes (MNP). Methods: Using Seahorse technology, pulsed stable isotope-resolved metabolomics and enzyme activity assays we characterize the central carbon metabolism and mitochondrial function of human and murine MNP under HS in vitro. HS as well as pharmacologic uncoupling of the electron transport chain (ETC) under normal salt (NS) is used to analyze mitochondrial function on immune cell activation and function (as determined by E.coli killing and CD4+ T cell migration capacity). In two independent clinical studies we analyze the impact of a HS diet over two weeks (NCT02509962) and short-term salt challenge by a single meal (NCT04175249) on mitochondrial function of human monocytes in vivo. Results: Extracellular sodium was taken up into the intracellular compartment followed by the inhibition of mitochondrial respiration in murine and human macrophages (MΦ). Mechanistically, HS reduces mitochondrial membrane potential, ETC complex II activity, oxygen consumption, and ATP production independently of the polarization status of MΦ. Subsequently, cell activation is altered with improved bactericidal function in HS-treated M1-like MΦ and diminished CD4+ T cell migration in HS-treated M2-like MΦ. Pharmacologic uncoupling of the ETC under NS phenocopies HS-induced transcriptional changes and bactericidal function of human and murine MNP. Clinically, also in vivo rise in plasma sodium concentration within the physiological range reversibly reduces mitochondrial function in human monocytes. In both, a 14-day and single meal HS challenge, healthy volunteers displayed a plasma sodium increase of{\~ }x = 2mM and{\~ }x = 2.3mM, respectively, that correlated with decreased monocytic mitochondrial oxygen consumption. Conclusions: Our data identify the disturbance of mitochondrial respiration as the initial step by which HS mechanistically influences immune cell function. While these functional changes might help to resolve bacterial infections, a shift towards pro-inflammation could accelerate inflammatory CVD.",
author = "Sabrina Geisberger and Hendrik Bartolomaeus and Patrick Neubert and Ralf Willebrand and Christin Zasada and Thomas Bartolomaeus and Victoria McParland and Dries Swinnen and Anneleen Geuzens and Andr{\'a}s Maifeld and Luka Krampert and Marion Vogl and Anja M{\"a}hler and Nicola Wilck and Lajos Marko and Ekin Tilic and Forslund, {Sofia K} and Binger, {Katrina J} and Johannes Stegbauer and Ralf Dechend and Markus Kleinewietfeld and Jonathan Jantsch and Stefan Kempa and M{\"u}ller, {Dominik N}",
year = "2021",
doi = "10.1161/CIRCULATIONAHA.120.052788",
language = "English",
volume = "144",
pages = "144–158",
journal = "Circulation",
issn = "0009-7322",
publisher = "Lippincott Williams & Wilkins",
number = "2",

}

RIS

TY - JOUR

T1 - Salt Transiently Inhibits Mitochondrial Energetics in Mononuclear Phagocytes

AU - Geisberger, Sabrina

AU - Bartolomaeus, Hendrik

AU - Neubert, Patrick

AU - Willebrand, Ralf

AU - Zasada, Christin

AU - Bartolomaeus, Thomas

AU - McParland, Victoria

AU - Swinnen, Dries

AU - Geuzens, Anneleen

AU - Maifeld, András

AU - Krampert, Luka

AU - Vogl, Marion

AU - Mähler, Anja

AU - Wilck, Nicola

AU - Marko, Lajos

AU - Tilic, Ekin

AU - Forslund, Sofia K

AU - Binger, Katrina J

AU - Stegbauer, Johannes

AU - Dechend, Ralf

AU - Kleinewietfeld, Markus

AU - Jantsch, Jonathan

AU - Kempa, Stefan

AU - Müller, Dominik N

PY - 2021

Y1 - 2021

N2 - Background: Dietary high salt (HS) is a leading risk factor for mortality and morbidity. Serum sodium transiently increases postprandially, but can also accumulate at sites of inflammation affecting differentiation and function of innate and adaptive immune cells. Here, we focus on how changes in extracellular sodium, mimicking alterations in the circulation and tissues, affect the early metabolic, transcriptional and functional adaption of human and murine mononuclear phagocytes (MNP). Methods: Using Seahorse technology, pulsed stable isotope-resolved metabolomics and enzyme activity assays we characterize the central carbon metabolism and mitochondrial function of human and murine MNP under HS in vitro. HS as well as pharmacologic uncoupling of the electron transport chain (ETC) under normal salt (NS) is used to analyze mitochondrial function on immune cell activation and function (as determined by E.coli killing and CD4+ T cell migration capacity). In two independent clinical studies we analyze the impact of a HS diet over two weeks (NCT02509962) and short-term salt challenge by a single meal (NCT04175249) on mitochondrial function of human monocytes in vivo. Results: Extracellular sodium was taken up into the intracellular compartment followed by the inhibition of mitochondrial respiration in murine and human macrophages (MΦ). Mechanistically, HS reduces mitochondrial membrane potential, ETC complex II activity, oxygen consumption, and ATP production independently of the polarization status of MΦ. Subsequently, cell activation is altered with improved bactericidal function in HS-treated M1-like MΦ and diminished CD4+ T cell migration in HS-treated M2-like MΦ. Pharmacologic uncoupling of the ETC under NS phenocopies HS-induced transcriptional changes and bactericidal function of human and murine MNP. Clinically, also in vivo rise in plasma sodium concentration within the physiological range reversibly reduces mitochondrial function in human monocytes. In both, a 14-day and single meal HS challenge, healthy volunteers displayed a plasma sodium increase of ̃x = 2mM and ̃x = 2.3mM, respectively, that correlated with decreased monocytic mitochondrial oxygen consumption. Conclusions: Our data identify the disturbance of mitochondrial respiration as the initial step by which HS mechanistically influences immune cell function. While these functional changes might help to resolve bacterial infections, a shift towards pro-inflammation could accelerate inflammatory CVD.

AB - Background: Dietary high salt (HS) is a leading risk factor for mortality and morbidity. Serum sodium transiently increases postprandially, but can also accumulate at sites of inflammation affecting differentiation and function of innate and adaptive immune cells. Here, we focus on how changes in extracellular sodium, mimicking alterations in the circulation and tissues, affect the early metabolic, transcriptional and functional adaption of human and murine mononuclear phagocytes (MNP). Methods: Using Seahorse technology, pulsed stable isotope-resolved metabolomics and enzyme activity assays we characterize the central carbon metabolism and mitochondrial function of human and murine MNP under HS in vitro. HS as well as pharmacologic uncoupling of the electron transport chain (ETC) under normal salt (NS) is used to analyze mitochondrial function on immune cell activation and function (as determined by E.coli killing and CD4+ T cell migration capacity). In two independent clinical studies we analyze the impact of a HS diet over two weeks (NCT02509962) and short-term salt challenge by a single meal (NCT04175249) on mitochondrial function of human monocytes in vivo. Results: Extracellular sodium was taken up into the intracellular compartment followed by the inhibition of mitochondrial respiration in murine and human macrophages (MΦ). Mechanistically, HS reduces mitochondrial membrane potential, ETC complex II activity, oxygen consumption, and ATP production independently of the polarization status of MΦ. Subsequently, cell activation is altered with improved bactericidal function in HS-treated M1-like MΦ and diminished CD4+ T cell migration in HS-treated M2-like MΦ. Pharmacologic uncoupling of the ETC under NS phenocopies HS-induced transcriptional changes and bactericidal function of human and murine MNP. Clinically, also in vivo rise in plasma sodium concentration within the physiological range reversibly reduces mitochondrial function in human monocytes. In both, a 14-day and single meal HS challenge, healthy volunteers displayed a plasma sodium increase of ̃x = 2mM and ̃x = 2.3mM, respectively, that correlated with decreased monocytic mitochondrial oxygen consumption. Conclusions: Our data identify the disturbance of mitochondrial respiration as the initial step by which HS mechanistically influences immune cell function. While these functional changes might help to resolve bacterial infections, a shift towards pro-inflammation could accelerate inflammatory CVD.

U2 - 10.1161/CIRCULATIONAHA.120.052788

DO - 10.1161/CIRCULATIONAHA.120.052788

M3 - Journal article

C2 - 33906377

VL - 144

SP - 144

EP - 158

JO - Circulation

JF - Circulation

SN - 0009-7322

IS - 2

ER -

ID: 260998304