Network


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

Hotspot


Dive into the research topics where Milan Theurl is active.

Publication


Featured researches published by Milan Theurl.


Journal of Immunology | 2007

Visfatin, an Adipocytokine with Proinflammatory and Immunomodulating Properties

Alexander R. Moschen; Arthur Kaser; Barbara Enrich; Birgit A. Mosheimer; Milan Theurl; Harald Niederegger; Herbert Tilg

Adipocytokines are mainly adipocyte-derived cytokines regulating metabolism and as such are key regulators of insulin resistance. Some adipocytokines such as adiponectin and leptin affect immune and inflammatory functions. Visfatin (pre-B cell colony-enhancing factor) has recently been identified as a new adipocytokine affecting insulin resistance by binding to the insulin receptor. In this study, we show that recombinant visfatin activates human leukocytes and induces cytokine production. In CD14+ monocytes, visfatin induces the production of IL-1β, TNF-α, and especially IL-6. Moreover, it increases the surface expression of costimulatory molecules CD54, CD40, and CD80. Visfatin-stimulated monocytes show augmented FITC-dextran uptake and an enhanced capacity to induce alloproliferative responses in human lymphocytes. Visfatin-induced effects involve p38 as well as MEK1 pathways as determined by inhibition with MAPK inhibitors and we observed activation of NF-κB. In vivo, visfatin induces circulating IL-6 in BALB/c mice. In patients with inflammatory bowel disease, plasma levels of visfatin are elevated and its mRNA expression is significantly increased in colonic tissue of Crohn’s and ulcerative colitis patients compared with healthy controls. Macrophages, dendritic cells, and colonic epithelial cells might be additional sources of visfatin as determined by confocal microscopy. Visfatin can be considered a new proinflammatory adipocytokine.


Blood | 2009

Regulation of iron homeostasis in anemia of chronic disease and iron deficiency anemia: diagnostic and therapeutic implications.

Igor Theurl; Elmar Aigner; Milan Theurl; Manfred Nairz; Markus Seifert; Andrea Schroll; Thomas Sonnweber; Lukas Eberwein; Derrick Ryan Witcher; Anthony T. Murphy; Victor J. Wroblewski; Eva Wurz; Christian Datz; Guenter Weiss

The anemia of chronic disease (ACD) is characterized by macrophage iron retention induced by cytokines and the master regulator hepcidin. Hepcidin controls cellular iron efflux on binding to the iron export protein ferroportin. Many patients, however, present with both ACD and iron deficiency anemia (ACD/IDA), the latter resulting from chronic blood loss. We used a rat model of ACD resulting from chronic arthritis and mimicked ACD/IDA by additional phlebotomy to define differing iron-regulatory pathways. Iron retention during inflammation occurs in macrophages and the spleen, but not in the liver. In rats and humans with ACD, serum hepcidin concentrations are elevated, which is paralleled by reduced duodenal and macrophage expression of ferroportin. Individuals with ACD/IDA have significantly lower hepcidin levels than ACD subjects, and ACD/IDA persons, in contrast to ACD subjects, were able to absorb dietary iron from the gut and to mobilize iron from macrophages. Circulating hepcidin levels affect iron traffic in ACD and ACD/IDA and are more responsive to the erythropoietic demands for iron than to inflammation. Hepcidin determination may aid to differentiate between ACD and ACD/IDA and in selecting appropriate therapy for these patients.


Cellular Microbiology | 2007

The co-ordinated regulation of iron homeostasis in murine macrophages limits the availability of iron for intracellular Salmonella typhimurium

Manfred Nairz; Igor Theurl; Susanne Ludwiczek; Milan Theurl; Sabine Mair; Gernot Fritsche; Günter Weiss

In being both, a modifier of cellular immune effector pathways and an essential nutrient for microbes, iron is a critical determinant in host–pathogen interaction. Here, we investigated the metabolic changes of macrophage iron homeostasis and immune function following the infection of RAW264.7 murine macrophages with Salmonella typhimurium. We observed an enhanced expression of the principal iron export protein, ferroportin 1, and a subsequent increase of iron efflux in Salmonella‐infected phagocytes. In parallel, the expression of haem oxygenase 1 and of the siderophore‐binding peptide lipocalin 2 was markedly enhanced following pathogen entry. Collectively, these modulations reduced both the cytoplasmatic labile iron and the ferritin storage iron pool within macrophages, thus restricting the acquisition of iron by intramacrophage Salmonella. Correspondingly, limitation of macrophage iron decreased microbial survival, whereas iron supplementation impaired immune response pathways in Salmonella‐infected macrophages (nitric oxide formation and tumour necrosis factor‐α production) and promoted intracellular bacterial proliferation. Our findings suggest that the enhancement of ferroportin 1‐mediated iron efflux, the upregulation of the haem‐degrading enzyme haem oxygenase 1 and the induction of lipocalin 2 following infection concordantly aim at withholding iron from intracellular S. typhimurium and to increase antimicrobial immune effector pathways thus limiting pathogen proliferation.


Blood | 2011

Pharmacologic inhibition of hepcidin expression reverses anemia of chronic inflammation in rats.

Igor Theurl; Andrea Schroll; Thomas Sonnweber; Manfred Nairz; Milan Theurl; Wolfgang Willenbacher; Kathrin Eller; Dominik Wolf; Markus Seifert; Chia Chi Sun; Charles C. Hong; Tracey Menhall; Patrick Gearing; Herbert Y. Lin; Guenter Weiss

Anemia of chronic inflammation (ACI) is the most frequent anemia in hospitalized patients and is associated with significant morbidity. A major underlying mechanism of ACI is the retention of iron within cells of the reticuloendothelial system (RES), thus making the metal unavailable for efficient erythropoiesis. This reticuloendothelial iron sequestration is primarily mediated by excess levels of the iron regulatory peptide hepcidin down-regulating the functional expression of the only known cellular iron export protein ferroportin resulting in blockade of iron egress from these cells. Using a well-established rat model of ACI, we herein provide novel evidence for effective treatment of ACI by blocking endogenous hepcidin production using the small molecule dorsomorphin derivative LDN-193189 or the protein soluble hemojuvelin-Fc (HJV.Fc) to inhibit bone morphogenetic protein-Smad mediated signaling required for effective hepcidin transcription. Pharmacologic inhibition of hepcidin expression results in mobilization of iron from the RES, stimulation of erythropoiesis and correction of anemia. Thus, hepcidin lowering agents are a promising new class of pharmacologic drugs to effectively combat ACI.


Nature Medicine | 2007

Ca2+ channel blockers reverse iron overload by a new mechanism via divalent metal transporter-1

Susanne Ludwiczek; Igor Theurl; Martina U. Muckenthaler; Martin Jakab; Sabine Mair; Milan Theurl; Judit Kiss; Markus Paulmichl; Matthias W. Hentze; Markus Ritter; Guenter Weiss

Hereditary hemochromatosis and transfusional iron overload are frequent clinical conditions associated with progressive iron accumulation in parenchymal tissues, leading to eventual organ failure. We have discovered a new mechanism to reverse iron overload—pharmacological modulation of the divalent metal transporter-1 (DMT-1). DMT-1 mediates intracellular iron transport during the transferrin cycle and apical iron absorption in the duodenum. Its additional functions in iron handling in the kidney and liver are less well understood. We show that the L-type calcium channel blocker nifedipine increases DMT-1–mediated cellular iron transport 10- to 100-fold at concentrations between 1 and 100 μM. Mechanistically, nifedipine causes this effect by prolonging the iron-transporting activity of DMT-1. We show that nifedipine mobilizes iron from the liver of mice with primary and secondary iron overload and enhances urinary iron excretion. Modulation of DMT-1 function by L-type calcium channel blockers emerges as a new pharmacological therapy for the treatment of iron overload disorders.


Immunity | 2011

Erythropoietin Contrastingly Affects Bacterial Infection and Experimental Colitis by Inhibiting Nuclear Factor-κB-Inducible Immune Pathways

Manfred Nairz; Andrea Schroll; Alexander R. Moschen; Thomas Sonnweber; Milan Theurl; Igor Theurl; Nicole Taub; Christina Jamnig; Daniela Neurauter; Lukas A. Huber; Herbert Tilg; Patrizia Moser; Günter Weiss

Summary Erythropoietin (EPO) is the principal cytokine regulating erythropoiesis through its receptor, EPOR. Interestingly, EPORs are also found on immune cells with incompletely understood functions. Here, we show that EPO inhibits the induction of proinflammatory genes including tumor necrosis factor (TNF)-α and inducible nitric oxide (NO) synthase in activated macrophages, which is mechanistically attributable to blockage of nuclear factor (NF)-κB p65 activation by EPO. Accordingly, in systemic Salmonella infection, treatment of mice with EPO results in reduced survival and impaired pathogen clearance because of diminished formation of anti-microbial effector molecules such as TNF-α and NO. However, neutralization of endogenous EPO or genetic ablation of Epor promotes Salmonella elimination. In contrast, in chemically induced colitis, EPO-EPOR interaction decreases the production of NF-κB-inducible immune mediators, thus limiting tissue damage and ameliorating disease severity. These immune-modulatory effects of EPO may be of therapeutic relevance in infectious and inflammatory diseases.


Blood | 2009

Absence of functional Hfe protects mice from invasive Salmonella enterica serovar Typhimurium infection via induction of lipocalin-2.

Manfred Nairz; Igor Theurl; Andrea Schroll; Milan Theurl; Gernot Fritsche; Ewald Lindner; Markus Seifert; Marie Laure V Crouch; Klaus Hantke; Shizuo Akira; Ferric C. Fang; Günter Weiss

Mutations of HFE are associated with hereditary hemochromatosis, but their influence on host susceptibility to infection is incompletely understood. We report that mice lacking one or both Hfe alleles are protected from septicemia with Salmonella Typhimurium, displaying prolonged survival and improved control of bacterial replication. This increased resistance is paralleled by an enhanced production of the enterochelin-binding peptide lipocalin-2 (Lcn2), which reduces the availability of iron for Salmonella within Hfe-deficient macrophages. Accordingly, Hfe(-/-)Lcn2(-/-) macrophages are unable to efficiently control the infection or to withhold iron from intracellular Salmonella. Correspondingly, the protection conferred by the Hfe defect is abolished in Hfe(-/-) mice infected with enterochelin-deficient Salmonella as well as in Hfe(-/-)Lcn2(-/-) mice infected with wild-type bacteria. Thus, by induction of the iron-capturing peptide Lcn2, absence of functional Hfe confers host resistance to systemic infection with Salmonella, thereby providing an evolutionary advantage which may account for the high prevalence of genetic hemochromatosis.


Nature Medicine | 2016

On-demand erythrocyte disposal and iron recycling requires transient macrophages in the liver

Igor Theurl; Ingo Hilgendorf; Manfred Nairz; Piotr Tymoszuk; David Haschka; Malte Asshoff; Shun He; Louisa M.S. Gerhardt; Tobias A. W. Holderried; Markus Seifert; Sieghart Sopper; Ashley M. Fenn; Atsushi Anzai; Sara Rattik; Cameron S. McAlpine; Milan Theurl; Peter Wieghofer; Yoshiko Iwamoto; Georg F. Weber; Nina K Harder; Benjamin G. Chousterman; Tara Arvedson; Mary McKee; Fudi Wang; Oliver M D Lutz; Emanuele Rezoagli; Lorenzo Berra; Marco Prinz; Matthias Nahrendorf; Guenter Weiss

Iron is an essential component of the erythrocyte protein hemoglobin and is crucial to oxygen transport in vertebrates. In the steady state, erythrocyte production is in equilibrium with erythrocyte removal. In various pathophysiological conditions, however, erythrocyte life span is compromised severely, which threatens the organism with anemia and iron toxicity. Here we identify an on-demand mechanism that clears erythrocytes and recycles iron. We show that monocytes that express high levels of lymphocyte antigen 6 complex, locus C1 (LY6C1, also known as Ly-6C) ingest stressed and senescent erythrocytes, accumulate in the liver via coordinated chemotactic cues, and differentiate into ferroportin 1 (FPN1, encoded by SLC40A1)-expressing macrophages that can deliver iron to hepatocytes. Monocyte-derived FPN1+Tim-4neg macrophages are transient, reside alongside embryonically derived T cell immunoglobulin and mucin domain containing 4 (Timd4, also known as Tim-4)high Kupffer cells (KCs), and depend on the growth factor Csf1 and the transcription factor Nrf2 (encoded by Nfe2l2). The spleen, likewise, recruits iron-loaded Ly-6Chigh monocytes, but these do not differentiate into iron-recycling macrophages, owing to the suppressive action of Csf2. The accumulation of a transient macrophage population in the liver also occurs in mouse models of hemolytic anemia, anemia of inflammation, and sickle cell disease. Inhibition of monocyte recruitment to the liver during stressed erythrocyte delivery leads to kidney and liver damage. These observations identify the liver as the primary organ that supports rapid erythrocyte removal and iron recycling, and uncover a mechanism by which the body adapts to fluctuations in erythrocyte integrity.


Journal of Hepatology | 2010

Hepatic but not brain iron is rapidly chelated by deferasirox in aceruloplasminemia due to a novel gene mutation

Armin Finkenstedt; Elisabeth Wolf; Elmar Höfner; Bethina Isasi Gasser; Sylvia Bösch; Rania Bakry; Marc Creus; Christian Kremser; Michael Schocke; Milan Theurl; Patrizia Moser; Melanie Schranz; G. Bonn; Werner Poewe; Wolfgang Vogel; Andreas R. Janecke; Heinz Zoller

Background & Aims Aceruloplasminemia is a rare autosomal recessive neurodegenerative disease associated with brain and liver iron accumulation which typically presents with movement disorders, retinal degeneration, and diabetes mellitus. Ceruloplasmin is a multi-copper ferroxidase that is secreted into plasma and facilitates cellular iron export and iron binding to transferrin. Results A novel homozygous ceruloplasmin gene mutation, c.2554+1G>T, was identified as the cause of aceruloplasminemia in three affected siblings. Two siblings presented with movement disorders and diabetes. Complementary DNA sequencing showed that this mutation causes skipping of exon 14 and deletion of amino acids 809–852 while preserving the open reading frame. Western blotting of liver extracts and sera of affected patients showed retention of the abnormal protein in the liver. Aceruloplasminemia was associated with severe brain and liver iron overload, where hepatic mRNA expression of the iron hormone hepcidin was increased, corresponding to the degree of iron overload. Hepatic iron concentration normalized after 3 and 5 months of iron chelation therapy with deferasirox, which was also associated with reduced insulin demands. During short term treatment there was no clinical or imaging evidence for significant effects on brain iron overload. Conclusions Aceruloplasminemia can show an incomplete clinical penetrance but is invariably associated with iron accumulation in the liver and in the brain. Iron accumulation in aceruloplasminemia is a result of defective cellular iron export, where hepcidin regulation is appropriate for the degree of iron overload. Iron chelation with deferasirox was effective in mobilizing hepatic iron but has no effect on brain iron.


British Journal of Haematology | 2010

Growth differentiation factor 15 in anaemia of chronic disease, iron deficiency anaemia and mixed type anaemia.

Igor Theurl; Armin Finkenstedt; Andrea Schroll; Manfred Nairz; Thomas Sonnweber; Rosa Bellmann-Weiler; Milan Theurl; Markus Seifert; Victor J. Wroblewski; Anthony T. Murphy; Derrick R. Witcher; Heinz Zoller; Günter Weiss

Recently, the iron and erythropoiesis‐controlled growth differentiation factor 15 (GDF15) has been shown to inhibit the expression of hepcidin in β‐thalassaemia patients, thereby increasing iron absorption despite iron overload. To access the diagnostic and pathogenic impact of GDF15 in inflammatory anaemia the association of GDF15 expression with serum iron parameters and hepcidin was studied in patients suffering from iron deficiency anaemia (IDA), anaemia of chronic disease (ACD) and ACD subjects with true iron deficiency (ACD/IDA). GDF15 was significantly increased in both ACD and ACD/IDA, but not in IDA subjects as compared to controls. In contrast, hepcidin levels were significantly lower in IDA and ACD/IDA subjects than in ACD patients. IDA and ACD/IDA, but not ACD, showed an association between GDF15 and soluble transferrin receptor, an indicator of iron requirement for erythropoiesis. However, GDF15 did not correlate to hepcidin in either patient group. While GDF15 levels were linked to the needs for erythropoiesis and iron homeostasis in IDA, the immunity‐driven increase of GDF15 may not primarily affect iron homeostasis and hepcidin expression. This indicates that other ACD‐related factors may overcome the regulatory effects of GDF15 on hepcidin expression during inflammation.

Collaboration


Dive into the Milan Theurl's collaboration.

Top Co-Authors

Avatar

Igor Theurl

Innsbruck Medical University

View shared research outputs
Top Co-Authors

Avatar

Manfred Nairz

Innsbruck Medical University

View shared research outputs
Top Co-Authors

Avatar

Markus Seifert

Innsbruck Medical University

View shared research outputs
Top Co-Authors

Avatar

Andrea Schroll

Innsbruck Medical University

View shared research outputs
Top Co-Authors

Avatar

Guenter Weiss

Innsbruck Medical University

View shared research outputs
Top Co-Authors

Avatar

Thomas Sonnweber

Innsbruck Medical University

View shared research outputs
Top Co-Authors

Avatar

Günter Weiss

Innsbruck Medical University

View shared research outputs
Top Co-Authors

Avatar

Alexander R. Moschen

Innsbruck Medical University

View shared research outputs
Top Co-Authors

Avatar

David Haschka

Innsbruck Medical University

View shared research outputs
Top Co-Authors

Avatar

Rosa Bellmann-Weiler

Innsbruck Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge