Network


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

Hotspot


Dive into the research topics where Jürgen Bernhagen is active.

Publication


Featured researches published by Jürgen Bernhagen.


Frontiers in Immunology | 2013

Arrest functions of the MIF ligand/receptor axes in atherogenesis

Sabine Tillmann; Jürgen Bernhagen; Heidi Noels

Macrophage migration inhibitory factor (MIF) has been defined as an important chemokine-like function (CLF) chemokine with an essential role in monocyte recruitment and arrest. Adhesion of monocytes to the vessel wall and their transendothelial migration are critical in atherogenesis and many other inflammatory diseases. Chemokines carefully control all steps of the monocyte recruitment process. Those chemokines specialized in controlling arrest are typically immobilized on the endothelial surface, mediating the arrest of rolling monocytes by chemokine receptor-triggered pathways. The chemokine receptor CXCR2 functions as an important arrest receptor on monocytes. An arrest function has been revealed for the bona fide CXCR2 ligands CXCL1 and CXCL8, but genetic studies also suggested that additional arrest chemokines are likely to be involved in atherogenic leukocyte recruitment. While CXCR2 is known to interact with numerous CXC chemokine ligands, the CLF chemokine MIF, which structurally does not belong to the CXC chemokine sub-family, was surprisingly identified as a non-cognate ligand of CXCR2, responsible for critical arrest functions during the atherogenic process. MIF was originally identified as macrophage migration inhibitory factor (this function being eponymous), but is now known as a potent inflammatory cytokine with CLFs including chemotaxis and leukocyte arrest. This review will cover the mechanisms underlying these functions, including MIF’s effects on LFA1 integrin activity and signal transduction, and will discuss the structural similarities between MIF and the bona fide CXCR2 ligand CXCL8 while emphasizing the structural differences. As MIF also interacts with CXCR4, a chemokine receptor implicated in CXCL12-elicited lymphocyte arrest, the arrest potential of the MIF/CXCR4 axis will also be scrutinized as well as the recently identified role of pericyte MIF in attracting leukocytes exiting through venules as part of the pericyte “motility instruction program.”


Thrombosis and Haemostasis | 2013

Platelets are a previously unrecognised source of MIF

T. Strüßmann; Sabine Tillmann; T. Wirtz; R. Bucala; P. von Hundelshausen; Jürgen Bernhagen

Macrophage migration inhibitory factor (MIF) is an inflammatory cytokine with chemokine-like functions and a role in atherogenesis. MIF is secreted by various cells including endothelial cells and macrophages. Platelets are another prominent cell type with a role in atherogenesis and are a rich source of atherogenic chemokines. We asked whether platelets express and secrete MIF. In comparison, CXCL12 release was determined. We examined the subcellular localisation of MIF in platelets/megakaryocytes, studied its co-localisation with other platelet-derived mediators and asked whether platelets contain MIF mRNA. Moreover, we probed the functional role of platelet-derived MIF in inflammatory cell recruitment. Using Western blot and ELISA, we demonstrated and quantitated MIF protein in human and mouse platelets. Applying confocal-microscopy, MIF was found to localise in granular-like structures, but did not co-localise with known platelet cytokines. qPCR indicated that platelets contain low levels of MIF mRNA. ELISA measurements from human platelet supernatants showed that, whereas thrombin and collagen triggered the release of MIF and CXCL12, ADP and oxidised LDL promoted CXCL12 but not MIF secretion. Using Transwell assays, we demonstrated that platelet supernatants promoted monocyte chemotaxis and that this was blocked by neutralising MIF antibodies.This is the first report demonstrating MIF secretion from activated platelets, suggesting that platelets are a previously unrecognised source of MIF in inflammatory processes. There are distinct activating stimuli for MIF and CXCL12 secretion. A substantial portion of the chemotactic capacity of stimulated platelet supernatants is contributed by MIF, suggesting a role for platelet-derived MIF in atherogenic cell recruitment.


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

High expression levels of macrophage migration inhibitory factor sustain the innate immune responses of neonates

Thierry Roger; Anina Schneider; Manuela Weier; Fred C.G.J. Sweep; Didier Le Roy; Jürgen Bernhagen; Thierry Calandra; Eric Giannoni

Significance During pregnancy, high circulating levels of adenosine and prostaglandins reduce the ability of fetal immune cells to mount powerful proinflammatory responses. In contrast, newborns express 10-fold higher levels of the proinflammatory immune regulator migration inhibitory factor (MIF) compared with adults. MIF sustains cell activation and cytokine production and counterregulates adenosine and prostaglandin E2-mediated immunosuppression in newborn monocytes. Yet excessive MIF expression during an established infection worsens the outcome of newborn mice. Thus, we identify a unique role for MIF in regulating neonatal innate immune responses and propose that MIF has a protective role to reduce susceptibility to infection during the neonatal period but may favor uncontrolled inflammation during sepsis, leading to adverse outcomes. The vulnerability to infection of newborns is associated with a limited ability to mount efficient immune responses. High concentrations of adenosine and prostaglandins in the fetal and neonatal circulation hamper the antimicrobial responses of newborn immune cells. However, the existence of mechanisms counterbalancing neonatal immunosuppression has not been investigated. Remarkably, circulating levels of macrophage migration inhibitory factor (MIF), a proinflammatory immunoregulatory cytokine expressed constitutively, were 10-fold higher in newborns than in children and adults. Newborn monocytes expressed high levels of MIF and released MIF upon stimulation with Escherichia coli and group B Streptococcus, the leading pathogens of early-onset neonatal sepsis. Inhibition of MIF activity or MIF expression reduced microbial product-induced phosphorylation of p38 and ERK1/2 mitogen-activated protein kinases and secretion of cytokines. Recombinant MIF used at newborn, but not adult, concentrations counterregulated adenosine and prostaglandin E2-mediated inhibition of ERK1/2 activation and TNF production in newborn monocytes exposed to E. coli. In agreement with the concept that once infection is established high levels of MIF are detrimental to the host, treatment with a small molecule inhibitor of MIF reduced systemic inflammatory response, bacterial proliferation, and mortality of septic newborn mice. Altogether, these data provide a mechanistic explanation for how newborns may cope with an immunosuppressive environment to maintain a certain threshold of innate defenses. However, the same defense mechanisms may be at the expense of the host in conditions of severe infection, suggesting that MIF could represent a potential attractive target for immune-modulating adjunctive therapies for neonatal sepsis.


Basic Research in Cardiology | 2016

From basic mechanisms to clinical applications in heart protection, new players in cardiovascular diseases and cardiac theranostics: meeting report from the third international symposium on "New frontiers in cardiovascular research"

Hector A. Cabrera-Fuentes; Julián Aragonés; Jürgen Bernhagen; Andreas Boening; William A. Boisvert; Hans Erik Bøtker; Heerajnarain Bulluck; Stuart A. Cook; Fabio Di Lisa; Felix B. Engel; Bernd Engelmann; Fulvia Ferrazzi; Péter Ferdinandy; Alan Fong; Ingrid Fleming; Erich Gnaiger; Sauri Hernández-Reséndiz; Siavash Beikoghli Kalkhoran; Moo Hyun Kim; Sandrine Lecour; Elisa A. Liehn; Michael Marber; Manuel Mayr; Tetsuji Miura; Sang-Bing Ong; Karlheinz Peter; Daniel Sedding; Manvendra K. Singh; M.Saadeh Suleiman; Hans Schnittler

In this meeting report, particularly addressing the topic of protection of the cardiovascular system from ischemia/reperfusion injury, highlights are presented that relate to conditioning strategies of the heart with respect to molecular mechanisms and outcome in patients’ cohorts, the influence of co-morbidities and medications, as well as the contribution of innate immune reactions in cardioprotection. Moreover, developmental or systems biology approaches bear great potential in systematically uncovering unexpected components involved in ischemia–reperfusion injury or heart regeneration. Based on the characterization of particular platelet integrins, mitochondrial redox-linked proteins, or lipid-diol compounds in cardiovascular diseases, their targeting by newly developed theranostics and technologies opens new avenues for diagnosis and therapy of myocardial infarction to improve the patients’ outcome.


Antioxidants & Redox Signaling | 2015

Interaction of MIF Family Proteins in Myocardial Ischemia/Reperfusion Damage and Their Influence on Clinical Outcome of Cardiac Surgery Patients.

Christian Stoppe; Steffen Rex; Andreas Goetzenich; Sandra Kraemer; Christoph Emontzpohl; Josefin Soppert; Luisa Averdunk; Yu Sun; Rolf Rossaint; Hongqi Lue; Caleb Huang; Yan Song; Georgios Pantouris; Elias Lolis; Lin Leng; Wibke Schulte; Richard Bucala; Christian Weber; Jürgen Bernhagen

Abstract Aims: Cardiac surgery involves myocardial ischemia/reperfusion (I/R) with potentially deleterious consequences. Macrophage migration inhibitory factor (MIF) is a stress-regulating chemokine-like cytokine that protects against I/R damage, but functional links with its homolog, d-dopachrome tautomerase (MIF-2), and the circulating soluble receptor CD74 (sCD74) are unknown. In this study, we investigate the role of MIF, MIF-2, sCD74, and MIF genotypes in patients scheduled for elective single or complex surgical procedures such as coronary artery bypass grafting or valve replacement. Results: MIF and MIF-2 levels significantly increased intraoperatively, whereas measured sCD74 decreased correspondingly. Circulating sCD74/MIF complexes were detectable in 50% of patients and enhanced MIF antioxidant activity. Intraoperative MIF levels were independently associated with a reduced risk for the development of atrial fibrillation (AF) (odds ratio 0.99 [0.98–1.00]; p=0.007). Circulating levels of MIF-2, but not MIF, were associated with an increased frequency of organ dysfunction and predicted the occurrence of AF (area under the curve [AUC]=0.663; p=0.041) and pneumonia (AUC=0.708; p=0.040). Patients with a high-expression MIF genotype exhibited a reduced incidence of organ dysfunction compared with patients with low-expression MIF genotypes (3 vs. 25; p=0.042). Innovation: The current study comprehensively highlights the kinetics and clinical relevance of MIF family proteins and the MIF genotype in cardiac surgery patients. Conclusion: Our findings suggest that increased MIF levels during cardiac surgery feature organ-protective properties during myocardial I/R, while the soluble MIF receptor, sCD74, may enhance MIF antioxidant activity. In contrast, high MIF-2 levels are predictive of the development of organ dysfunction. Importantly, we provide first evidence for a gene–phenotype relationship between variant MIF alleles and clinical outcome in cardiac surgery patients. Antioxid. Redox Signal. 00, 000–000.


Atherosclerosis | 2015

Platelet-derived MIF: A novel platelet chemokine with distinct recruitment properties

T. Wirtz; Sabine Tillmann; Tim Strüßmann; Sandra Kraemer; Johan W. M. Heemskerk; Oliver Grottke; Meinrad Gawaz; Philipp von Hundelshausen; Jürgen Bernhagen

OBJECTIVEnMacrophage migration inhibitory factor (MIF) is an inflammatory cytokine with chemokine-like functions that plays a role in several inflammatory diseases including atherosclerosis. We recently demonstrated that in addition to macrophages and endothelial cells, platelets are a source of MIF. However, the functional relevance of platelet-derived MIF and differences to other platelet chemokines are unclear. Here, we sought to define the secretion pattern of platelet MIF and to characterize its functional profile in comparison with known atherogenic platelet chemokines.nnnMETHODS AND RESULTSnApplying ELISA, we show that MIF is released from thrombin-stimulated platelets after 2xa0h, whereas CXCL12 and CXCL4 are secreted within minutes. Applied to platelets, MIF, unlike CXCL12, did not enhance platelet activation as analyzed by platelet aggregation, CD62P exposure and chemokine secretion studies. In contrast, both MIF and CXCL12 attenuated ADP-induced calcium transients in platelets. Transmigration and monocyte flow adhesion assays toward conditioned platelet supernatants together with MIF antibody blockade or supernatants from Mif(-/-) mice suggested that platelet-derived MIF has a stronger chemotactic activity than CXCL12 at its respective optimal secretion interval, and showed that platelet MIF substantially contributes to monocyte adhesion on endothelial layers. Moreover, MIF was found to delay clot retraction.nnnCONCLUSIONSnWe demonstrate that MIF differs from other platelet-derived chemokines by delayed secretion kinetics and by a distinct autocrine/paracrine modulation potential. Importantly, MIF was found to be a major platelet-derived chemotactic recruitment factor with clot-modulating properties and therefore might be relevant in inflammatory diseases such as atherosclerosis.


Current Medicinal Chemistry | 2013

Hetero-Oligomerization of Chemokine Receptors: Diversity and Relevance for Function

Sandra Kraemer; S. Alampour Rajabi; O. El Bounkari; Jürgen Bernhagen

The G protein-coupled receptor (GPCR) family of membrane receptors encompasses over 1000 members, representing the largest known receptor family, with a variety of structurally different ligands. GPCRs are favorite targets for drug development in numerous diseases. Chemokine receptors are an important GPCR sub-class and are known to play a crucial role in the regulation of multiple physiological and various pathophysiological processes, including inflammation, atherosclerosis, cancer, and viral infections. Chemokine receptor activation is controlled by some 50 chemokine ligands which often act in a redundant and overlapping manner, enabling for a complex regulatory system together controlling and fine-tuning the specificity and spatio-temporal properties of the response. Recent findings have indicated that additionally the organization of chemokine receptors on the cell surface could be critical for driving their biological effects. In fact, chemokine receptors have increasingly been found to organize into homo- or hetero-oligomeric complexes, in part in a ligand-inducible manner, resulting in complex networks and crosstalk with other orthogonal signaling complexes. There has even been evidence for heterologous complex formation between chemokine receptors and non-chemokine receptor G protein-coupled receptors (GPCRs), and even non-GPCRs. However, the functional consequences of this kind of oligomerization have remained poorly understood, even for the chemokine receptor homo-oligomers. Yet, there is growing evidence that targeting homo- and/or hetero-oligomerization of chemokine receptors might be beneficial for the development of novel and specific therapeutics. In the present article, we highlight the multi-faceted complexity of chemokine receptor structures with a focus on their hetero-oligomerization properties.


Journal of Biological Chemistry | 2016

Macrophage migration inhibitory factor-CXCR4 receptor interactions: Evidence for partial allosteric agonism in comparison with CXCL12 chemokine

Deepa Rajasekaran; Sabine Gröning; Corinna Schmitz; Swen Zierow; Natalie Drucker; Maria Bakou; Kristian Kohl; André Mertens; Hongqi Lue; Christian Weber; Annie Xiao; Gary D. Luker; Aphrodite Kapurniotu; Elias Lolis; Jürgen Bernhagen

An emerging number of non-chemokine mediators are found to bind to classical chemokine receptors and to elicit critical biological responses. Macrophage migration inhibitory factor (MIF) is an inflammatory cytokine that exhibits chemokine-like activities through non-cognate interactions with the chemokine receptors CXCR2 and CXCR4, in addition to activating the type II receptor CD74. Activation of the MIF-CXCR2 and -CXCR4 axes promotes leukocyte recruitment, mediating the exacerbating role of MIF in atherosclerosis and contributing to the wealth of other MIF biological activities. Although the structural basis of the MIF-CXCR2 interaction has been well studied and was found to engage a pseudo-ELR and an N-like loop motif, nothing is known about the regions of CXCR4 and MIF that are involved in binding to each other. Using a genetic strain of Saccharomyces cerevisiae that expresses a functional CXCR4 receptor, site-specific mutagenesis, hybrid CXCR3/CXCR4 receptors, pharmacological reagents, peptide array analysis, chemotaxis, fluorescence spectroscopy, and circular dichroism, we provide novel molecular information about the structural elements that govern the interaction between MIF and CXCR4. The data identify similarities with classical chemokine-receptor interactions but also provide evidence for a partial allosteric agonist compared with CXCL12 that is possible due to the two binding sites of CXCR4.


ACS Chemical Biology | 2014

Development of Accessible Peptidic Tool Compounds To Study the Phosphatase PTP1B in Intact Cells

Christoph Meyer; Birgit Hoeger; K Temmerman; Marianna Tatarek-Nossol; Pogenberg; Jürgen Bernhagen; M Wilmanns; Aphrodite Kapurniotu; Maja Köhn

Protein tyrosine phosphatases (PTPs) play crucial roles in health and disease. Chemical modulators of their activity are vital tools to study their function. An important aspect is the accessibility of these tools, which is usually limited or not existent due to the required, often complex synthesis of the molecules. We describe here a strategy for the development of cellular active inhibitors and in-cell detection tools for PTP1B as a model PTP, which plays important roles in diabetes, obesity, and cancer. The tool compounds are based on a peptide sequence from PTP1Bs substrate Src, and the resulting compounds are commercially accessible through standard peptide synthesis. The peptide inhibitor is remarkably selective against a panel of PTPs. We provide the co-crystal structure of PTP1B with the sequence from Src and the optimized peptide inhibitor, showing the molecular basis of the interaction of PTP1B with part of its natural substrate and explaining the crucial interactions to enhance binding affinity, which are made possible by simple optimization of the sequence. Our approach enables the broad accessibility of PTP1B tools to researchers and has the potential for the systematic development of accessible PTP modulators to enable the study of PTPs.


Journal of Tissue Engineering and Regenerative Medicine | 2018

The effect of mechanical stress on the proliferation, adipogenic differentiation and gene expression of human adipose-derived stem cells.

Nora E. Paul; Bernd Denecke; Bong-Sung Kim; Alice Dreser; Jürgen Bernhagen; Norbert Pallua

To allow for a better implementation of external volume expansion to clinical applications for soft tissue regeneration, it is necessary to comprehensively understand the underlying mechanisms. As human adipose‐derived stem cells (hASCs) play a crucial role in soft tissue enlargement, we investigated the impact of cyclic stretch on gene expression, proliferation rate and adipogenic differentiation of these cells. After cyclic stretching, RNA was extracted and subjected to DNA microarray analysis and reverse transcription‐quantitative polymerase chain reaction (RT‐qPCR). Also, the expression of FABP4 mRNA was analysed by RT‐qPCR to test whether mechanical stretch affected adipogenic differentiation of hASCs. The proliferation rate was assessed using the alamarBlue assay and Ki‐67 staining. A cell cycle analysis was performed with flow cytometry and Western blot. We found that cyclic stretch significantly induced the expression of CYP1B1 mRNA. Furthermore, the adipogenic differentiation of hASCs was impaired, as was the proliferation. This was partly due to a decrease in extracellular signal‐regulated kinase (ERK) 1/2 and histone H3 phosphorylation, suggesting a growth arrest in the G2/M phase of the cell cycle. Enrichment analyses demonstrated that stretch‐regulated genes were over‐represented in pathways and biological processes involved in extracellular matrix organization, vascular remodelling and responses to cell stress. Taken together, mechanical stress impaired both proliferation and adipogenic differentiation, but led to a tissue‐remodelling phenotype of hASCs. These data suggest that extracellular matrix remodelling and neoangiogenesis may play a more important role in external volume expansion than proliferation and adipogenesis of hASCs. Copyright

Collaboration


Dive into the Jürgen Bernhagen's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Heidi Noels

RWTH Aachen University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge