Network


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

Hotspot


Dive into the research topics where Arndt Manzel is active.

Publication


Featured researches published by Arndt Manzel.


Nature | 2013

Sodium chloride drives autoimmune disease by the induction of pathogenic TH17 cells.

Markus Kleinewietfeld; Arndt Manzel; Jens Titze; Heda Kvakan; Nir Yosef; Ralf A. Linker; Dominik Müller; David A. Hafler

There has been a marked increase in the incidence of autoimmune diseases in the past half-century. Although the underlying genetic basis of this class of diseases has recently been elucidated, implicating predominantly immune-response genes, changes in environmental factors must ultimately be driving this increase. The newly identified population of interleukin (IL)-17-producing CD4+ helper T cells (TH17 cells) has a pivotal role in autoimmune diseases. Pathogenic IL-23-dependent TH17 cells have been shown to be critical for the development of experimental autoimmune encephalomyelitis (EAE), an animal model for multiple sclerosis, and genetic risk factors associated with multiple sclerosis are related to the IL-23–TH17 pathway. However, little is known about the environmental factors that directly influence TH17 cells. Here we show that increased salt (sodium chloride, NaCl) concentrations found locally under physiological conditions in vivo markedly boost the induction of murine and human TH17 cells. High-salt conditions activate the p38/MAPK pathway involving nuclear factor of activated T cells 5 (NFAT5; also called TONEBP) and serum/glucocorticoid-regulated kinase 1 (SGK1) during cytokine-induced TH17 polarization. Gene silencing or chemical inhibition of p38/MAPK, NFAT5 or SGK1 abrogates the high-salt-induced TH17 cell development. The TH17 cells generated under high-salt conditions display a highly pathogenic and stable phenotype characterized by the upregulation of the pro-inflammatory cytokines GM-CSF, TNF-α and IL-2. Moreover, mice fed with a high-salt diet develop a more severe form of EAE, in line with augmented central nervous system infiltrating and peripherally induced antigen-specific TH17 cells. Thus, increased dietary salt intake might represent an environmental risk factor for the development of autoimmune diseases through the induction of pathogenic TH17 cells.


Immunity | 2015

Dietary Fatty Acids Directly Impact Central Nervous System Autoimmunity via the Small Intestine

Aiden Haghikia; Stefanie Jörg; Alexander Duscha; Johannes Berg; Arndt Manzel; Anne Waschbisch; Anna Hammer; De-Hyung Lee; Caroline May; Nicola Wilck; András Balogh; Annika I. Ostermann; Nils Helge Schebb; Denis A. Akkad; Diana A. Grohme; Markus Kleinewietfeld; Stefan Kempa; Jan Thöne; Seray Demir; Dominik Müller; Ralf Gold; Ralf A. Linker

Growing empirical evidence suggests that nutrition and bacterial metabolites might impact the systemic immune response in the context of disease and autoimmunity. We report that long-chain fatty acids (LCFAs) enhanced differentiation and proliferation of T helper 1 (Th1) and/or Th17 cells and impaired their intestinal sequestration via p38-MAPK pathway. Alternatively, dietary short-chain FAs (SCFAs) expanded gut T regulatory (Treg) cells by suppression of the JNK1 and p38 pathway. We used experimental autoimmune encephalomyelitis (EAE) as a model of T cell-mediated autoimmunity to show that LCFAs consistently decreased SCFAs in the gut and exacerbated disease by expanding pathogenic Th1 and/or Th17 cell populations in the small intestine. Treatment with SCFAs ameliorated EAE and reduced axonal damage via long-lasting imprinting on lamina-propria-derived Treg cells. These data demonstrate a direct dietary impact on intestinal-specific, and subsequently central nervous system-specific, Th cell responses in autoimmunity, and thus might have therapeutic implications for autoimmune diseases such as multiple sclerosis.


Proceedings of the National Academy of Sciences of the United States of America | 2009

Role of the renin-angiotensin system in autoimmune inflammation of the central nervous system.

Johannes Stegbauer; De-Hyung Lee; Silvia Seubert; Gisa Ellrichmann; Arndt Manzel; Heda Kvakan; Dominik Müller; Stefanie Gaupp; Lars Christian Rump; Ralf Gold; Ralf A. Linker

Angiotensin II is the principle effector molecule of the renin angiotensin system (RAS). It exerts its various actions on the cardiovascular and renal system, mainly via interaction with the angiotensin II type-1 receptor (AT1R), which contributes to blood pressure regulation and development of hypertension but may also mediate effects on the immune system. Here we study the role of the RAS in myelin-oligodendrocyte glycoprotein-induced experimental autoimmune encephalomyelitis (MOG-EAE), a model mimicking many aspects of multiple sclerosis. Quantitative RT-PCR analyses showed an up-regulation of renin, angiotensin-converting enzyme, as well as AT1R in the inflamed spinal cord and the immune system, including antigen presenting cells (APC). Treatment with the renin inhibitor aliskiren, the angiotensin II converting-enzyme inhibitor enalapril, as well as preventive or therapeutic application of the AT1R antagonist losartan, resulted in a significantly ameliorated course of MOG-EAE. Blockade of AT1R did not directly impact on T-cell responses, but significantly reduced numbers of CD11b+ or CD11c+ APC in immune organs and in the inflamed spinal cord. Additionally, AT1R blockade impaired the expression of CCL2, CCL3, and CXCL10, and reduced CCL2-induced APC migration. Our findings suggest a pivotal role of the RAS in autoimmune inflammation of the central nervous system and identify RAS blockade as a potential new target for multiple sclerosis therapy.


Current Allergy and Asthma Reports | 2014

Role of "Western diet" in inflammatory autoimmune diseases.

Arndt Manzel; Dominik N. Müller; David A. Hafler; Susan E. Erdman; Ralf A. Linker; Markus Kleinewietfeld

Developed societies, although having successfully reduced the burden of infectious disease, constitute an environment where metabolic, cardiovascular, and autoimmune diseases thrive. Living in westernized countries has not fundamentally changed the genetic basis on which these diseases emerge, but has strong impact on lifestyle and pathogen exposure. In particular, nutritional patterns collectively termed the “Western diet”, including high-fat and cholesterol, high-protein, high-sugar, and excess salt intake, as well as frequent consumption of processed and ‘fast foods’, promote obesity, metabolic syndrome, and cardiovascular disease. These factors have also gained high interest as possible promoters of autoimmune diseases. Underlying metabolic and immunologic mechanisms are currently being intensively explored. This review discusses the current knowledge relative to the association of “Western diet” with autoimmunity, and highlights the role of T cells as central players linking dietary influences to autoimmune pathology.


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.


Experimental Neurology | 2016

High salt drives Th17 responses in experimental autoimmune encephalomyelitis without impacting myeloid dendritic cells

Stefanie Jörg; Jan Kissel; Arndt Manzel; Markus Kleinewietfeld; Aiden Haghikia; Ralf Gold; Dominik Müller; Ralf A. Linker

Recently, we have shown that high dietary salt intake aggravates T helper cell (Th) 17 responses and neuroinflammation. Here, we employed in vitro assays for myeloid dendritic cell (mDC) maturation, DC cytokine production, T cell activation and ex vivo analyses in murine experimental autoimmune encephalomyelitis (EAE) to investigate whether the salt effect on Th17 cells is further mediated through DCs in vivo. In cell culture, an excess of 40mM sodium chloride did neither affect the generation, maturation nor the function of DCs, but, in different assays, significantly increased Th17 differentiation. During the initiation phase of MOG35-55 EAE, we did not observe altered DC frequencies or co-stimulatory capacities in lymphoid organs, while IL-17A production and Th17 cells in the spleen were significantly increased. Complementary ex vivo analyses of the spinal cord during the effector phase of EAE showed increased frequencies of Th17 cells, but did not reveal differences in phenotypes of CNS invading DCs. Finally, adaption of transgenic mice harboring a MOG specific T cell receptor to a high-salt diet led to aggravated clinical disease only after active immunization. Wild-type mice adapted to a high-salt diet in the effector phase of EAE, bypassing the priming phase of T cells, only displayed mildly aggravated disease. In summary, our data argue for a direct effect of NaCl on Th17 cells in neuroinflammation rather than an effect primarily exerted via DCs. These data may further fuel our understanding on the dietary impact on different immune cell subsets in autoimmune diseases, such as multiple sclerosis.


Proceedings of the National Academy of Sciences of the United States of America | 2016

Role of the receptor Mas in macrophage-mediated inflammation in vivo

Anna Hammer; Guang Yang; Juliane Friedrich; Ágnes Melinda Kovács; De-Hyung Lee; Katharina Grave; Stefanie Jörg; Natalia Alenina; Janina Grosch; Jürgen Winkler; Ralf Gold; Michael Bader; Arndt Manzel; Lars Christian Rump; Dominik N. Müller; Ralf A. Linker; Johannes Stegbauer

Significance The alternative renin–angiotensin system pathway, the angiotensin (Ang)-(1–7)/Mas axis, may counterbalance Ang II-mediated proinflammatory effects. To investigate the role of the Ang-(1–7)/Mas axis in immune cell function and inflammatory diseases in vivo, we used two different chronic inflammatory animal models. Deletion of Mas affects macrophage function and phenotype independently of the underlying phagocyte stimulus and aggravates the clinical course of experimental autoimmune encephalomyelitis as well as atherosclerosis in mice by tipping the in vivo balance from M(IL-4+IL-13)- to M(LPS+IFNγ)-like macrophages. Thus, modulation of the Ang-(1–7)/Mas axis counteracts the proinflammatory role of Ang II by regulating the delicate equilibrium between M(LPS+IFNγ)- and M(IL-4+IL-13)-like macrophages, thereby representing a promising pharmacological target for chronic inflammatory diseases. Recently, an alternative renin–angiotensin system pathway has been described, which involves binding of angiotensin-(1–7) to its receptor Mas. The Mas axis may counterbalance angiotensin-II–mediated proinflammatory effects, likely by affecting macrophage function. Here we investigate the role of Mas in murine models of autoimmune neuroinflammation and atherosclerosis, which both involve macrophage-driven pathomechanisms. Mas signaling affected macrophage polarization, migration, and macrophage-mediated T-cell activation. Mas deficiency exacerbated the course of experimental autoimmune encephalomyelitis and increased macrophage infiltration as well as proinflammatory gene expression in the spleen and spinal cord. Furthermore, Mas deficiency promoted atherosclerosis by affecting macrophage infiltration and migration and led to increased oxidative stress as well as impaired endothelial function in ApoE-deficient mice. In summary, we identified the Mas axis as an important factor in macrophage function during inflammation of the central nervous and vascular system in vivo. Modulating the Mas axis may constitute an interesting therapeutic target in multiple sclerosis and/or atherosclerosis.


Experimental & Translational Stroke Medicine | 2011

Vascular pathology in multiple sclerosis: mind boosting or myth busting?

Anne Waschbisch; Arndt Manzel; Ralf A. Linker; De-Hyung Lee

The investigation of central nervous system vascular changes in the pathophysiology of multiple sclerosis (MS) is a time-honored concept. Yet, recent reports on changes in venous cerebrospinal outflow, the advent of new magnetic resonance imaging techniques and the investigation of immunomodulatory properties of several vascular mediators on the molecular level have added new excitement to hypotheses centering around vascular pathology as determining factor in the pathophysiology of MS. Here we critically review the concept of chronic cerebrospinal venous insufficiency in MS patients and describe new imaging techniques including perfusion weighted imaging, susceptibility weighted imaging and diffusion weighted imaging which reveal central nervous system hypoperfusion, perivascular iron deposition and diffuse structural changes in the MS brain. On a molecular basis, vascular mediators represent interesting targets connecting vascular pathology with immunomodulation. In summary, the relation of venous changes to the pathophysiology of MS may not be as simple as initially described and it certainly seems awkward to think of the complex disease MS solely as result of a simple venous outflow obstruction. Yet, the investigation of new vascular concepts as one variable in the pathophysiology of the autoimmune attack seems very worthwhile and may add to a better understanding of this devastating disorder.


Blood | 2015

New role for the (pro)renin receptor in T-cell development.

Sabrina Geisberger; Ulrike Maschke; Matthias Gebhardt; Markus Kleinewietfeld; Arndt Manzel; Ralf A. Linker; Ann Patricia Chidgey; Ralf Dechend; Genevieve Nguyen; Oliver Daumke; Dominik N. Müller; Mark D. Wright; Katrina J. Binger

The (pro)renin receptor (PRR) was originally thought to be important for regulating blood pressure via the renin-angiotensin system. However, it is now emerging that PRR has instead a generic role in cellular development. Here, we have specifically deleted PRR from T cells. T-cell-specific PRR-knockout mice had a significant decrease in thymic cellularity, corresponding with a 100-fold decrease in the number of CD4(+) and CD8(+) thymocytes, and a large increase in double-negative (DN) precursors. Gene expression analysis on sorted DN3 thymocytes indicated that PRR-deficient thymocytes have perturbations in key cellular pathways essential at the DN3 stage, including transcription and translation. Further characterization of DN T-cell progenitors leads us to propose that PRR deletion affects thymocyte survival and development at multiple stages; from DN3 through to DN4, double-positive, and single-positive CD4 and CD8. Our study thus identifies a new role for PRR in T-cell development.


Scientific Reports | 2016

Neprilysin is a Mediator of Alternative Renin-Angiotensin-System Activation in the Murine and Human Kidney

Oliver Domenig; Arndt Manzel; Nadja Grobe; Eva Königshausen; Christopher C. Kaltenecker; Johannes J. Kovarik; Johannes Stegbauer; Susan B. Gurley; Dunja van Oyen; Marlies Antlanger; Michael Bader; Daisy Motta-Santos; Robson A.S. Santos; Khalid M. Elased; Marcus D. Säemann; Ralf A. Linker; Marko Poglitsch

Cardiovascular and renal pathologies are frequently associated with an activated renin-angiotensin-system (RAS) and increased levels of its main effector and vasoconstrictor hormone angiotensin II (Ang II). Angiotensin-converting-enzyme-2 (ACE2) has been described as a crucial enzymatic player in shifting the RAS towards its so-called alternative vasodilative and reno-protective axis by enzymatically converting Ang II to angiotensin-(1-7) (Ang-(1-7)). Yet, the relative contribution of ACE2 to Ang-(1-7) formation in vivo has not been elucidated. Mass spectrometry based quantification of angiotensin metabolites in the kidney and plasma of ACE2 KO mice surprisingly revealed an increase in Ang-(1-7), suggesting additional pathways to be responsible for alternative RAS activation in vivo. Following assessment of angiotensin metabolism in kidney homogenates, we identified neprilysin (NEP) to be a major source of renal Ang-(1-7) in mice and humans. These findings were supported by MALDI imaging, showing NEP mediated Ang-(1-7) formation in whole kidney cryo-sections in mice. Finally, pharmacologic inhibition of NEP resulted in strongly decreased Ang-(1-7) levels in murine kidneys. This unexpected new role of NEP may have implications for the combination therapy with NEP-inhibitors and angiotensin-receptor-blockade, which has been shown being a promising therapeutic approach for heart failure therapy.

Collaboration


Dive into the Arndt Manzel's collaboration.

Top Co-Authors

Avatar

Ralf A. Linker

University of Erlangen-Nuremberg

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

De-Hyung Lee

University of Erlangen-Nuremberg

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dominik N. Müller

Max Delbrück Center for Molecular Medicine

View shared research outputs
Top Co-Authors

Avatar

Ralf Gold

Ruhr University Bochum

View shared research outputs
Top Co-Authors

Avatar

Marko Poglitsch

Medical University of Vienna

View shared research outputs
Researchain Logo
Decentralizing Knowledge