Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Jonathan Jantsch is active.

Publication


Featured researches published by Jonathan Jantsch.


Nature Medicine | 2009

Macrophages regulate salt-dependent volume and blood pressure by a vascular endothelial growth factor-C–dependent buffering mechanism

Agnes Machnik; Wolfgang Neuhofer; Jonathan Jantsch; Anke Dahlmann; Tuomas Tammela; Katharina Machura; Joon-Keun Park; Franz-Xaver Beck; Dominik N. Müller; Wolfgang Derer; Jennifer Goss; Agata Ziomber; Peter Dietsch; Hubertus Wagner; Nico van Rooijen; Armin Kurtz; Karl F. Hilgers; Kari Alitalo; Kai-Uwe Eckardt; Friedrich C. Luft; Dontscho Kerjaschki; Jens Titze

In salt-sensitive hypertension, the accumulation of Na+ in tissue has been presumed to be accompanied by a commensurate retention of water to maintain the isotonicity of body fluids. We show here that a high-salt diet (HSD) in rats leads to interstitial hypertonic Na+ accumulation in skin, resulting in increased density and hyperplasia of the lymphcapillary network. The mechanisms underlying these effects on lymphatics involve activation of tonicity-responsive enhancer binding protein (TonEBP) in mononuclear phagocyte system (MPS) cells infiltrating the interstitium of the skin. TonEBP binds the promoter of the gene encoding vascular endothelial growth factor-C (VEGF-C, encoded by Vegfc) and causes VEGF-C secretion by macrophages. MPS cell depletion or VEGF-C trapping by soluble VEGF receptor-3 blocks VEGF-C signaling, augments interstitial hypertonic volume retention, decreases endothelial nitric oxide synthase expression and elevates blood pressure in response to HSD. Our data show that TonEBP–VEGF-C signaling in MPS cells is a major determinant of extracellular volume and blood pressure homeostasis and identify VEGFC as an osmosensitive, hypertonicity-driven gene intimately involved in salt-induced hypertension.


Journal of Immunology | 2008

Hypoxia and hypoxia-inducible factor-1 alpha modulate lipopolysaccharide-induced dendritic cell activation and function.

Jonathan Jantsch; Dipshikha Chakravortty; Nadine Turza; Alexander T. Prechtel; Björn Buchholz; Roman G. Gerlach; Melanie Volke; Joachim Gläsner; Christina Warnecke; Michael S. Wiesener; Kai-Uwe Eckardt; Alexander Steinkasserer; Michael Hensel; Carsten Willam

Dendritic cells (DC) play a key role in linking innate and adaptive immunity. In inflamed tissues, where DC become activated, oxygen tensions are usually low. Although hypoxia is increasingly recognized as an important determinant of cellular functions, the consequences of hypoxia and the role of one of the key players in hypoxic gene regulation, the transcription factor hypoxia inducible factor 1α (HIF-1α), are largely unknown. Thus, we investigated the effects of hypoxia and HIF-1α on murine DC activation and function in the presence or absence of an exogenous inflammatory stimulus. Hypoxia alone did not activate murine DC, but hypoxia combined with LPS led to marked increases in expression of costimulatory molecules, proinflammatory cytokine synthesis, and induction of allogeneic lymphocyte proliferation compared with LPS alone. This DC activation was accompanied by accumulation of HIF-1α protein levels, induction of glycolytic HIF target genes, and enhanced glycolytic activity. Using RNA interference techniques, knockdown of HIF-1α significantly reduced glucose use in DC, inhibited maturation, and led to an impaired capability to stimulate allogeneic T cells. Alltogether, our data indicate that HIF-1α and hypoxia play a crucial role for DC activation in inflammatory states, which is highly dependent on glycolysis even in the presence of oxygen.


Journal of Clinical Investigation | 2013

Immune cells control skin lymphatic electrolyte homeostasis and blood pressure

Helge Wiig; Agnes Schröder; Wolfgang Neuhofer; Jonathan Jantsch; Christoph W. Kopp; Tine V. Karlsen; Michael Boschmann; Jennifer Goss; Maija Bry; Natalia Rakova; Anke Dahlmann; Sven Brenner; Olav Tenstad; Harri Nurmi; Eero Mervaala; Hubertus Wagner; Franz-Xaver Beck; Dominik Müller; Dontscho Kerjaschki; Friedrich C. Luft; David G. Harrison; Kari Alitalo; Jens Titze

The skin interstitium sequesters excess Na+ and Cl- in salt-sensitive hypertension. Mononuclear phagocyte system (MPS) cells are recruited to the skin, sense the hypertonic electrolyte accumulation in skin, and activate the tonicity-responsive enhancer-binding protein (TONEBP, also known as NFAT5) to initiate expression and secretion of VEGFC, which enhances electrolyte clearance via cutaneous lymph vessels and increases eNOS expression in blood vessels. It is unclear whether this local MPS response to osmotic stress is important to systemic blood pressure control. Herein, we show that deletion of TonEBP in mouse MPS cells prevents the VEGFC response to a high-salt diet (HSD) and increases blood pressure. Additionally, an antibody that blocks the lymph-endothelial VEGFC receptor, VEGFR3, selectively inhibited MPS-driven increases in cutaneous lymphatic capillary density, led to skin Cl- accumulation, and induced salt-sensitive hypertension. Mice overexpressing soluble VEGFR3 in epidermal keratinocytes exhibited hypoplastic cutaneous lymph capillaries and increased Na+, Cl-, and water retention in skin and salt-sensitive hypertension. Further, we found that HSD elevated skin osmolality above plasma levels. These results suggest that the skin contains a hypertonic interstitial fluid compartment in which MPS cells exert homeostatic and blood pressure-regulatory control by local organization of interstitial electrolyte clearance via TONEBP and VEGFC/VEGFR3-mediated modification of cutaneous lymphatic capillary function.


Hypertension | 2013

(23)Na magnetic resonance imaging-determined tissue sodium in healthy subjects and hypertensive patients

Christoph W. Kopp; Peter Linz; Anke Dahlmann; Matthias Hammon; Jonathan Jantsch; Dominik Müller; Roland E. Schmieder; Alexander Cavallaro; Kai-Uwe Eckardt; Michael Uder; Friedrich C. Luft; Jens Titze

High dietary salt intake is associated with hypertension; the prevalence of salt-sensitive hypertension increases with age. We hypothesized that tissue Na+ might accumulate in hypertensive patients and that aging might be accompanied by Na+ deposition in tissue. We implemented 23Na magnetic resonance imaging to measure Na+ content of soft tissues in vivo earlier, but had not studied essential hypertension. We report on a cohort of 56 healthy control men and women, and 57 men and women with essential hypertension. The ages ranged from 22 to 90 years. 23Na magnetic resonance imaging measurements were made at the level of the calf. We observed age-dependent increases in Na+ content in muscle in men, whereas muscle Na+ content did not change with age in women. We estimated water content with conventional MRI and found no age-related increases in muscle water in men, despite remarkable Na+ accumulation, indicating water-free Na+ storage in muscle. With increasing age, there was Na+ deposition in the skin in both women and men; however, skin Na+ content remained lower in women. Similarly, this sex difference was found in skin water content, which was lower in women than in men. In contrast to muscle, increasing Na+ content was paralleled with increasing skin water content. When controlled for age, we found that patients with refractory hypertension had increased tissue Na+ content, compared with normotensive controls. These observations suggest that 23Na magnetic resonance imaging could have utility in assessing the role of tissue Na+ storage for cardiovascular morbidity and mortality in longitudinal studies.


Molecular Systems Biology | 2010

The phosphoproteome of toll‐like receptor‐activated macrophages

Gabriele Weintz; J. Olsen; Katja Frühauf; Ido Amit; Jonathan Jantsch; Jörg Mages; Cornelie Frech; Lars Dölken; Matthias Mann; Roland Lang

Recognition of microbial danger signals by toll‐like receptors (TLR) causes re‐programming of macrophages. To investigate kinase cascades triggered by the TLR4 ligand lipopolysaccharide (LPS) on systems level, we performed a global, quantitative and kinetic analysis of the phosphoproteome of primary macrophages using stable isotope labelling with amino acids in cell culture, phosphopeptide enrichment and high‐resolution mass spectrometry. In parallel, nascent RNA was profiled to link transcription factor (TF) phosphorylation to TLR4‐induced transcriptional activation. We reproducibly identified 1850 phosphoproteins with 6956 phosphorylation sites, two thirds of which were not reported earlier. LPS caused major dynamic changes in the phosphoproteome (24% up‐regulation and 9% down‐regulation). Functional bioinformatic analyses confirmed canonical players of the TLR pathway and highlighted other signalling modules (e.g. mTOR, ATM/ATR kinases) and the cytoskeleton as hotspots of LPS‐regulated phosphorylation. Finally, weaving together phosphoproteome and nascent transcriptome data by in silico promoter analysis, we implicated several phosphorylated TFs in primary LPS‐controlled gene expression.


Cell Metabolism | 2015

Cutaneous Na+ Storage Strengthens the Antimicrobial Barrier Function of the Skin and Boosts Macrophage-Driven Host Defense

Jonathan Jantsch; Valentin Schatz; Diana Friedrich; Agnes Schröder; Christoph W. Kopp; Isabel Siegert; Andreas Maronna; David Wendelborn; Peter Linz; Katrina J. Binger; Matthias Gebhardt; Matthias Heinig; Patrick Neubert; Fabian Fischer; Stefan Teufel; Jean-Pierre David; Clemens Neufert; Alexander Cavallaro; Natalia Rakova; Christoph Küper; Franz-Xaver Beck; Wolfgang Neuhofer; Dominik N. Müller; Gerold Schuler; Michael Uder; Christian Bogdan; Friedrich C. Luft; Jens Titze

Immune cells regulate a hypertonic microenvironment in the skin; however, the biological advantage of increased skin Na(+) concentrations is unknown. We found that Na(+) accumulated at the site of bacterial skin infections in humans and in mice. We used the protozoan parasite Leishmania major as a model of skin-prone macrophage infection to test the hypothesis that skin-Na(+) storage facilitates antimicrobial host defense. Activation of macrophages in the presence of high NaCl concentrations modified epigenetic markers and enhanced p38 mitogen-activated protein kinase (p38/MAPK)-dependent nuclear factor of activated T cells 5 (NFAT5) activation. This high-salt response resulted in elevated type-2 nitric oxide synthase (Nos2)-dependent NO production and improved Leishmania major control. Finally, we found that increasing Na(+) content in the skin by a high-salt diet boosted activation of macrophages in a Nfat5-dependent manner and promoted cutaneous antimicrobial defense. We suggest that the hypertonic microenvironment could serve as a barrier to infection.


Journal of Clinical Investigation | 2015

High salt reduces the activation of IL-4- and IL-13-stimulated macrophages

Katrina J. Binger; Matthias Gebhardt; Matthias Heinig; Carola Rintisch; Agnes Schroeder; Wolfgang Neuhofer; Karl F. Hilgers; Arndt Manzel; Christian Schwartz; Markus Kleinewietfeld; Jakob Voelkl; Valentin Schatz; Ralf A. Linker; Florian Lang; David Voehringer; Mark D. Wright; Norbert Hubner; Ralf Dechend; Jonathan Jantsch; Jens Titze; Dominik N. Müller

A high intake of dietary salt (NaCl) has been implicated in the development of hypertension, chronic inflammation, and autoimmune diseases. We have recently shown that salt has a proinflammatory effect and boosts the activation of Th17 cells and the activation of classical, LPS-induced macrophages (M1). Here, we examined how the activation of alternative (M2) macrophages is affected by salt. In stark contrast to Th17 cells and M1 macrophages, high salt blunted the alternative activation of BM-derived mouse macrophages stimulated with IL-4 and IL-13, M(IL-4+IL-13) macrophages. Salt-induced reduction of M(IL-4+IL-13) activation was not associated with increased polarization toward a proinflammatory M1 phenotype. In vitro, high salt decreased the ability of M(IL-4+IL-13) macrophages to suppress effector T cell proliferation. Moreover, mice fed a high salt diet exhibited reduced M2 activation following chitin injection and delayed wound healing compared with control animals. We further identified a high salt-induced reduction in glycolysis and mitochondrial metabolic output, coupled with blunted AKT and mTOR signaling, which indicates a mechanism by which NaCl inhibits full M2 macrophage activation. Collectively, this study provides evidence that high salt reduces noninflammatory innate immune cell activation and may thus lead to an overall imbalance in immune homeostasis.


Cellular Microbiology | 2003

Intracellular activities of Salmonella enterica in murine dendritic cells

Jonathan Jantsch; Cédric Cheminay; Dipshikha Chakravortty; Tobias M. Lindig; Joachim Hein; Michael Hensel

Dendritic cells (DC) efficiently phagocytose invading bacteria, but fail to kill intracellular pathogens such as Salmonella enterica serovar Typhimurium (S. Typhimurium). We analysed the intracellular fate of Salmonella in murine bone marrow‐derived DC (BM‐DC). The intracellular proliferation and subcellular localization were investigated for wild‐type S. Typhimurium and mutants deficient in Salmonella pathogenicity island 2 (SPI2), a complex virulence factor that is essential for systemic infections in the murine model and intracellular survival and replication in macrophages. Using a segregative plasmid to monitor intracellular cell division, we observed that, in BM‐DC, S. Typhimurium represents a static, non‐dividing population. In BM‐DC, S. Typhimurium resides in a membrane‐bound compartment that has acquired late endosomal markers. However, these bacteria respond to intracellular stimuli, because induction of SPI2 genes was observed. S. Typhimurium within DC are also able to translocate a virulence protein into their host cells. SPI2 function was not required for intracellular survival in DC, but we observed that the maturation of the Salmonella‐containing vesicle is different in DC infected with wild‐type bacteria and a strain deficient in SPI2. Our observations indicate that S. Typhimurium in DC are able to modify normal processes of their host cells.


Infection and Immunity | 2008

Functional analysis of the Salmonella pathogenicity island 2-mediated inhibition of antigen presentation in dendritic cells.

Serkan Halici; Sebastian F. Zenk; Jonathan Jantsch; Michael Hensel

ABSTRACT Salmonella enterica is a facultative intracellular pathogen that is able to modify host cell functions by means of effector proteins translocated by the type III secretion system (T3SS) encoded by Salmonella Pathogenicity Island 2 (SPI2). The SPI2-T3SS is also active in Salmonella after uptake by murine bone marrow-derived dendritic cells (BM-DC). We have previously shown that intracellular Salmonella interfere with the ability of BM-DC to stimulate antigen-dependent T-cell proliferation in an SPI2-T3SS-dependent manner. We observed that Salmonella-mediated inhibition of antigen presentation could be restored by external addition of peptides on major histocompatibility complex class II (MHC-II). The processing of antigens in Salmonella-infected cells was not altered; however, the intracellular loading of peptides on MHC-II was reduced as a function of the SPI2-T3SS. We set out to identify the effector proteins of the SPI2-T3SS involved in inhibition of antigen presentation and demonstrated that effector proteins SifA, SspH2, SlrP, PipB2, and SopD2 were equally important for the interference with antigen presentation, whereas SseF and SseG contributed to a lesser extent to this phenotype. These observations indicate the presence of a host cell-specific virulence function of a novel subset of SPI2-effector proteins.


Nature | 2017

Salt-responsive gut commensal modulates TH17 axis and disease

Nicola Wilck; Mariana Matus; Sean M. Kearney; Scott W. Olesen; Kristoffer Forslund; Hendrik Bartolomaeus; Stefanie Haase; Anja Mähler; András Balogh; Lajos Markó; Olga Vvedenskaya; Friedrich H. Kleiner; Dmitry Tsvetkov; Lars Klug; Paul Igor Costea; Shinichi Sunagawa; Lisa M. Maier; Natalia Rakova; Valentin Schatz; Patrick Neubert; Christian Frätzer; Alexander Krannich; Maik Gollasch; Diana A. Grohme; Beatriz F. Côrte-Real; Roman G. Gerlach; Marijana Basic; Athanasios Typas; Chuan Wu; Jens Titze

A Western lifestyle with high salt consumption can lead to hypertension and cardiovascular disease. High salt may additionally drive autoimmunity by inducing T helper 17 (TH17) cells, which can also contribute to hypertension. Induction of TH17 cells depends on gut microbiota; however, the effect of salt on the gut microbiome is unknown. Here we show that high salt intake affects the gut microbiome in mice, particularly by depleting Lactobacillus murinus. Consequently, treatment of mice with L. murinus prevented salt-induced aggravation of actively induced experimental autoimmune encephalomyelitis and salt-sensitive hypertension by modulating TH17 cells. In line with these findings, a moderate high-salt challenge in a pilot study in humans reduced intestinal survival of Lactobacillus spp., increased TH17 cells and increased blood pressure. Our results connect high salt intake to the gut–immune axis and highlight the gut microbiome as a potential therapeutic target to counteract salt-sensitive conditions.

Collaboration


Dive into the Jonathan Jantsch's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Christian Bogdan

University of Erlangen-Nuremberg

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kai-Uwe Eckardt

University of Erlangen-Nuremberg

View shared research outputs
Top Co-Authors

Avatar

Agnes Schröder

University of Erlangen-Nuremberg

View shared research outputs
Top Co-Authors

Avatar

Friedrich C. Luft

Max Delbrück Center for Molecular Medicine

View shared research outputs
Top Co-Authors

Avatar

Michael Hensel

University of Osnabrück

View shared research outputs
Top Co-Authors

Avatar

Carsten Willam

University of Erlangen-Nuremberg

View shared research outputs
Top Co-Authors

Avatar

Dominik N. Müller

Max Delbrück Center for Molecular Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge