Network


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

Hotspot


Dive into the research topics where Tommy Regen is active.

Publication


Featured researches published by Tommy Regen.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Locus ceruleus controls Alzheimer's disease pathology by modulating microglial functions through norepinephrine

Michael T. Heneka; Fabian Nadrigny; Tommy Regen; Ana Martinez-Hernandez; Lucia Dumitrescu-Ozimek; Dick Terwel; Daniel Jardanhazi-Kurutz; Jochen Walter; Frank Kirchhoff; Uwe-Karsten Hanisch; Markus P. Kummer

Locus ceruleus (LC)-supplied norepinephrine (NE) suppresses neuroinflammation in the brain. To elucidate the effect of LC degeneration and subsequent NE deficiency on Alzheimers disease pathology, we evaluated NE effects on microglial key functions. NE stimulation of mouse microglia suppressed Aβ-induced cytokine and chemokine production and increased microglial migration and phagocytosis of Aβ. Induced degeneration of the locus ceruleus increased expression of inflammatory mediators in APP-transgenic mice and resulted in elevated Aβ deposition. In vivo laser microscopy confirmed a reduced recruitment of microglia to Aβ plaque sites and impaired microglial Aβ phagocytosis in NE-depleted APP-transgenic mice. Supplying the mice the norepinephrine precursor L-threo-DOPS restored microglial functions in NE-depleted mice. This indicates that decrease of NE in locus ceruleus projection areas facilitates the inflammatory reaction of microglial cells in AD and impairs microglial migration and phagocytosis, thereby contributing to reduced Aβ clearance. Consequently, therapies targeting microglial phagocytosis should be tested under NE depletion.


Infection and Immunity | 2010

Toll-Like Receptor Stimulation Enhances Phagocytosis and Intracellular Killing of Nonencapsulated and Encapsulated Streptococcus pneumoniae by Murine Microglia

Sandra Ribes; Sandra Ebert; Tommy Regen; Amit Agarwal; Simone C. Tauber; Dirk Czesnik; Annette Spreer; Stephanie Bunkowski; Helmut Eiffert; Uwe Karsten Hanisch; Sven Hammerschmidt; Roland Nau

ABSTRACT Toll-like receptors (TLRs) are crucial pattern recognition receptors in innate immunity that are expressed in microglia, the resident macrophages of the brain. TLR2, -4, and -9 are important in the responses against Streptococcus pneumoniae, the most common agent causing bacterial meningitis beyond the neonatal period. Murine microglial cultures were stimulated with agonists for TLR1/2 (Pam3CSK4), TLR4 (lipopolysaccharide), and TLR9 (CpG oligodeoxynucleotide) for 24 h and then exposed to either the encapsulated D39 (serotype 2) or the nonencapsulated R6 strain of S. pneumoniae. After stimulation, the levels of interleukin-6 and CCL5 (RANTES [regulated upon activation normal T-cell expressed and secreted]) were increased, confirming microglial activation. The TLR1/2, -4, and -9 agonist-stimulated microglia ingested significantly more bacteria than unstimulated cells (P < 0.05). The presence of cytochalasin D, an inhibitor of actin polymerizaton, blocked >90% of phagocytosis. Along with an increased phagocytic activity, the intracellular bacterial killing was also increased in TLR-stimulated cells compared to unstimulated cells. Together, our data suggest that microglial stimulation by these TLRs may increase the resistance of the brain against pneumococcal infections.


Glia | 2010

Microglia promote colonization of brain tissue by breast cancer cells in a Wnt-dependent way.

Tobias Pukrop; Faramarz Dehghani; Han-Ning Chuang; Raphaela Lohaus; Kathrin Bayanga; Stephan Heermann; Tommy Regen; Denise van Rossum; Florian Klemm; Matthias Schulz; Laila Siam; Anja Hoffmann; Lorenz Trümper; Christine Stadelmann; Ingo Bechmann; Uwe-Karsten Hanisch; Claudia Binder

Although there is increasing evidence that blood‐derived macrophages support tumor progression, it is still unclear whether specialized resident macrophages, such as brain microglia, also play a prominent role in metastasis formation. Here, we show that microglia enhance invasion and colonization of brain tissue by breast cancer cells, serving both as active transporters and guiding rails. This is antagonized by inactivation of microglia as well as by the Wnt inhibitor Dickkopf‐2. Proinvasive microglia demonstrate altered morphology, but neither upregulation of M2‐like cytokines nor differential gene expression. Bacterial lipopolysacharide shifts tumor‐educated microglia into a classical M1 phenotype, reduces their proinvasive function, and unmasks inflammatory and Wnt signaling as the most strongly regulated pathways. Histological findings in human brain metastases underline the significance of these results. In conclusion, microglia are critical for the successful colonization of the brain by epithelial cancer cells, suggesting inhibition of proinvasive microglia as a promising antimetastatic strategy.


Glia | 2012

Toll-like receptor activation reveals developmental reorganization and unmasks responder subsets of microglia.

Jörg Scheffel; Tommy Regen; Denise van Rossum; Stefanie Seifert; Sandra Ribes; Roland Nau; Roham Parsa; Robert A. Harris; Hendrikus W.G.M. Boddeke; Han-Ning Chuang; Tobias Pukrop; Johannes T. Wessels; Tanja Jürgens; Doron Merkler; Wolfgang Brück; Mareike Schnaars; Mikael Simons; Helmut Kettenmann; Uwe-Karsten Hanisch

The sentinel and immune functions of microglia require rapid and appropriate reactions to infection and damage. Their Toll‐like receptors (TLRs) sense both as threats. However, whether activated microglia mount uniform responses or whether subsets conduct selective tasks is unknown. We demonstrate that murine microglia reorganize their responses to TLR activations postnatally and that this process comes with a maturation of TLR4‐organized functions. Although induction of MHCI for antigen presentation remains as a pan‐populational feature, synthesis of TNFα becomes restricted to a subset, even within adult central nervous system regions. Response heterogeneity is evident ex vivo, in situ, and in vivo, but is not limited to TNFα production or to TLR‐triggered functions. Also, clearance activities for myelin under physiological and pathophysiological conditions, IFNγ‐enforced upregulation of MHCII, or challenged inductions of other proinflammatory factors reveal dissimilar microglial contributions. Notably, response heterogeneity is also confirmed in human brain tissue. Our findings suggest that microglia divide by constitutive and inducible capacities. Privileged production of inflammatory mediators assigns a master control to subsets. Sequestration of clearance of endogenous material versus antigen presentation in exclusive compartments can separate potentially interfering functions. Finally, subsets rather than a uniform population of microglia may assemble the reactive phenotypes in responses during infection, injury, and rebuilding, warranting consideration in experimental manipulation and therapeutic strategies.


PLOS Pathogens | 2010

T Cell-Dependence of Lassa Fever Pathogenesis

Lukas Flatz; Toni Rieger; Doron Merkler; Andreas Bergthaler; Tommy Regen; Mariann Schedensack; Lukas Bestmann; Admar Verschoor; Mario Kreutzfeldt; Wolfgang Brück; Uwe-Karsten Hanisch; Stephan Günther; Daniel D. Pinschewer

Lassa virus (LASV), the causative agent of Lassa fever (LF), is endemic in West Africa, accounting for substantial morbidity and mortality. In spite of ongoing research efforts, LF pathogenesis and mechanisms of LASV immune control remain poorly understood. While normal laboratory mice are resistant to LASV, we report that mice expressing humanized instead of murine MHC class I (MHC-I) failed to control LASV infection and develop severe LF. Infection of MHC-I knockout mice confirmed a key role for MHC-I-restricted T cell responses in controlling LASV. Intriguingly we found that T cell depletion in LASV-infected HHD mice prevented disease, irrespective of high-level viremia. Widespread activation of monocyte/macrophage lineage cells, manifest through inducible NO synthase expression, and elevated IL-12p40 serum levels indicated a systemic inflammatory condition. The absence of extensive monocyte/macrophage activation in T cell-depleted mice suggested that T cell responses contribute to deleterious innate inflammatory reactions and LF pathogenesis. Our observations in mice indicate a dual role for T cells, not only protecting from LASV, but also enhancing LF pathogenesis. The possibility of T cell-driven enhancement and immunopathogenesis should be given consideration in future LF vaccine development.


Journal of Neuroimmunology | 2010

Inflammatory cytokine release of astrocytes in vitro is reduced by all-trans retinoic acid

Sabien van Neerven; Andrei Nemes; Philipp Imholz; Tommy Regen; Bernd Denecke; Sonja Johann; Cordian Beyer; Uwe-Karsten Hanisch; Jörg Mey

In the central nervous system inflammation is mediated by microglia and astrocytes. To investigate its regulation, murine astrocyte cultures were treated with bacterial lipopolysaccharides (LPS) and analyzed with Affymetrix gene array, qRT-PCR and ELISA. Cells responded to LPS with a strong upregulation of pro-inflammatory cytokines and chemokines. Treatment with the transcriptional activator retinoic acid (RA) suppressed mRNA expression and protein release of several important cytokines (IL-1β 4%, IL-6 21%, TNFα 30%, IL-12p40 42%, and IL-12p35/p40 27%; p<0.01). The data are consistent with the hypothesis that all-trans RA takes part in endogenous anti-inflammatory feedback loops in the CNS.


Infection and Immunity | 2009

Toll-Like Receptor Prestimulation Increases Phagocytosis of Escherichia coli DH5α and Escherichia coli K1 Strains by Murine Microglial Cells

Sandra Ribes; Sandra Ebert; Dirk Czesnik; Tommy Regen; Andre Zeug; Stephanie Bukowski; Alexander Mildner; Helmut Eiffert; Uwe-Karsten Hanisch; Sven Hammerschmidt; Roland Nau

ABSTRACT Meningitis and meningoencephalitis caused by Escherichia coli are associated with high rates of mortality. When an infection occurs, Toll-like receptors (TLRs) expressed by microglial cells can recognize pathogen-associated molecular patterns and activate multiple steps in the inflammatory response that coordinate the brains local defense, such as phagocytosis of invading pathogens. An upregulation of the phagocytic ability of reactive microglia could improve the host defense in immunocompromised patients against pathogens such as E. coli. Here, murine microglial cultures were stimulated with the TLR agonists Pam3CSK4 (TLR1/TLR2), lipopolysaccharide (TLR4), and CpG oligodeoxynucleotide (TLR9) for 24 h. Upon stimulation, levels of tumor necrosis factor alpha and the neutrophil chemoattractant CXCL1 were increased, indicating microglial activation. Phagocytic activity was studied after adding either E. coli DH5α or E. coli K1 strains. After 60 and 90 min of bacterial exposure, the number of ingested bacteria was significantly higher in cells prestimulated with TLR agonists than in unstimulated controls (P < 0.01). Addition of cytochalasin D, an inhibitor of actin polymerization, blocked >90% of phagocytosis. We also analyzed the ability of microglia to kill the ingested E. coli strains. Intracellularly surviving bacteria were quantified at different time points (90, 150, 240, and 360 min) after 90 min of phagocytosis. The number of bacteria killed intracellularly after 6 h was higher in cells primed with the different TLR agonists than in unstimulated microglia. Our data suggest that microglial stimulation by the TLR system can increase bacterial phagocytosis and killing. This approach could improve central nervous system resistance to infections in immunocompromised patients.


Brain Behavior and Immunity | 2011

CD14 and TRIF govern distinct responsiveness and responses in mouse microglial TLR4 challenges by structural variants of LPS.

Tommy Regen; Denise van Rossum; Jörg Scheffel; Maria-Eleni Kastriti; Natalia H. Revelo; Marco Prinz; Wolfgang Brück; Uwe-Karsten Hanisch

Toll-like receptor (TLR) 4 responds to a range of agonists in infection and injury, but is best known for the recognition of bacterial lipopolysaccharides (LPS). Assembly in heterologous receptor complexes as well as signaling through both MyD88 and TRIF adaptor proteins, as unmatched by other TLRs, could underlie its versatile response options, probably also in a cell type-dependent manner. We show that microglia, the CNS macrophages, react to diverse LPS variants, including smooth (S) and rough (R) LPS chemotypes, with cytokine/chemokine induction, MHC I expression and suppression of myelin phagocytosis. The TLR4 co-receptor CD14 was shown in peritoneal macrophages to be essential for S-LPS effects and the link of both S- and R-LPS to TRIF signaling. In contrast, cd14(-/-) microglia readily respond to S- and R-LPS, suggesting an a priori high(er) sensitivity to both chemotypes, while CD14 confers increased S- and R-LPS potencies and compensates for their differences. Importantly, CD14 controls the magnitude and shapes the profile of cyto/chemokine production, this influence being itself regulated by critical LPS concentrations. Comparing reactive phenotypes of microglia with deficiencies in CD14, MyD88 and TRIF (cd14(-/-), myd88(-/-), and trif(lps2)), we found that distinct signaling routes organize for individual functions in either concerted or non-redundant fashion and that CD14 has contributions beyond the link to TRIF. Modulation of response profiles by key cytokines finally reveals that the microglial TLR4 can differentiate between the class of LPS structures and a self-derived agonist, fibronectin. It thus proves as a sophisticated decision maker in infectious and non-infectious CNS challenges.


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

TLR4-activated microglia require IFN-γ to induce severe neuronal dysfunction and death in situ

Ismini E. Papageorgiou; Andrea Lewen; Lukas V. Galow; Tiziana Cesetti; Jörg Scheffel; Tommy Regen; Uwe-Karsten Hanisch; Oliver Kann

Significance Microglia (brain macrophages) become rapidly activated in most neuropsychiatric disorders. A popular concept is that a single pathogenic stimulus, such as bacterial lipopolysaccharide (LPS) through Toll-like receptor 4 (TLR4), is sufficient to induce a reactive proinflammatory phenotype in microglia that exerts neurotoxicity. This concept is biologically risky, however. Here we provide evidence that chronic activation with either LPS or the leukocyte cytokine IFN-γ induces different reactive phenotypes in microglia of postnatal hippocampal tissue. Notably, these phenotypes only moderately alter diverse neuronal functions. In contrast, coactivation of TLR4 and IFN-γ receptors results in massive neural dysfunction and death. Thus, activation of TLR4 in microglia in situ requires concomitant IFN-γ signaling from other host immune cells to induce neurodegeneration. Microglia (tissue-resident macrophages) represent the main cell type of the innate immune system in the CNS; however, the mechanisms that control the activation of microglia are widely unknown. We systematically explored microglial activation and functional microglia–neuron interactions in organotypic hippocampal slice cultures, i.e., postnatal cortical tissue that lacks adaptive immunity. We applied electrophysiological recordings of local field potential and extracellular K+ concentration, immunohistochemistry, design-based stereology, morphometry, Sholl analysis, and biochemical analyses. We show that chronic activation with either bacterial lipopolysaccharide through Toll-like receptor 4 (TLR4) or leukocyte cytokine IFN-γ induces reactive phenotypes in microglia associated with morphological changes, population expansion, CD11b and CD68 up-regulation, and proinflammatory cytokine (IL-1β, TNF-α, IL-6) and nitric oxide (NO) release. Notably, these reactive phenotypes only moderately alter intrinsic neuronal excitability and gamma oscillations (30–100 Hz), which emerge from precise synaptic communication of glutamatergic pyramidal cells and fast-spiking, parvalbumin-positive GABAergic interneurons, in local hippocampal networks. Short-term synaptic plasticity and extracellular potassium homeostasis during neural excitation, also reflecting astrocyte function, are unaffected. In contrast, the coactivation of TLR4 and IFN-γ receptors results in neuronal dysfunction and death, caused mainly by enhanced microglial inducible nitric oxide synthase (iNOS) expression and NO release, because iNOS inhibition is neuroprotective. Thus, activation of TLR4 in microglia in situ requires concomitant IFN-γ receptor signaling from peripheral immune cells, such as T helper type 1 and natural killer cells, to unleash neurotoxicity and inflammation-induced neurodegeneration. Our findings provide crucial mechanistic insight into the complex process of microglia activation, with relevance to several neurologic and psychiatric disorders.


Journal of Neurochemistry | 2010

Inflammatory chemokine release of astrocytes in vitro is reduced by all-trans retinoic acid.

Sabien van Neerven; Tommy Regen; Dhana Wolf; Andrei Nemes; Sonja Johann; Cordian Beyer; Uwe-Karsten Hanisch; Jörg Mey

J. Neurochem. (2010) 114, 1511–1526.

Collaboration


Dive into the Tommy Regen's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sandra Ribes

University of Göttingen

View shared research outputs
Top Co-Authors

Avatar

Roland Nau

University of Göttingen

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jörg Scheffel

University of Göttingen

View shared research outputs
Top Co-Authors

Avatar

Hana Janova

University of Göttingen

View shared research outputs
Top Co-Authors

Avatar

Marco Prinz

University of Freiburg

View shared research outputs
Top Co-Authors

Avatar

Sandra Ebert

University of Göttingen

View shared research outputs
Top Co-Authors

Avatar

Tobias Pukrop

University of Göttingen

View shared research outputs
Researchain Logo
Decentralizing Knowledge