Network


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

Hotspot


Dive into the research topics where Friederike Langhauser is active.

Publication


Featured researches published by Friederike Langhauser.


Blood | 2013

Regulatory T cells are strong promoters of acute ischemic stroke in mice by inducing dysfunction of the cerebral microvasculature

Christoph Kleinschnitz; Peter Kraft; Angela Dreykluft; Ina Hagedorn; Kerstin Göbel; Michael K. Schuhmann; Friederike Langhauser; Xavier Helluy; Tobias Schwarz; Stefan Bittner; Christian T. Mayer; Marc Brede; Csanad Varallyay; Mirko Pham; Martin Bendszus; Peter M. Jakob; Tim Magnus; Sven G. Meuth; Yoichiro Iwakura; Alma Zernecke; Tim Sparwasser; Bernhard Nieswandt; Guido Stoll; Heinz Wiendl

We have recently identified T cells as important mediators of ischemic brain damage, but the contribution of the different T-cell subsets is unclear. Forkhead box P3 (FoxP3)-positive regulatory T cells (Tregs) are generally regarded as prototypic anti-inflammatory cells that maintain immune tolerance and counteract tissue damage in a variety of immune-mediated disorders. In the present study, we examined the role of Tregs after experimental brain ischemia/reperfusion injury. Selective depletion of Tregs in the DEREG mouse model dramatically reduced infarct size and improved neurologic function 24 hours after stroke and this protective effect was preserved at later stages of infarct development. The specificity of this detrimental Treg effect was confirmed by adoptive transfer experiments in wild-type mice and in Rag1(-/-) mice lacking lymphocytes. Mechanistically, Tregs induced microvascular dysfunction in vivo by increased interaction with the ischemic brain endothelium via the LFA-1/ICAM-1 pathway and platelets and these findings were confirmed in vitro. Ablation of Tregs reduced microvascular thrombus formation and improved cerebral reperfusion on stroke, as revealed by ultra-high-field magnetic resonance imaging at 17.6 Tesla. In contrast, established immunoregulatory characteristics of Tregs had no functional relevance. We define herein a novel and unexpected role of Tregs in a primary nonimmunologic disease state.


Nature Medicine | 2013

Endothelial TWIK-related potassium channel-1 (TREK1) regulates immune-cell trafficking into the CNS

Stefan Bittner; Tobias Ruck; Michael K. Schuhmann; Alexander M. Herrmann; Hamid Moha ou Maati; Nicole Bobak; Kerstin Göbel; Friederike Langhauser; David Stegner; Petra Ehling; Marc Borsotto; Hans-Christian Pape; Bernhard Nieswandt; Christoph Kleinschnitz; Catherine Heurteaux; Hans-Joachim Galla; Thomas Budde; Heinz Wiendl; Sven G. Meuth

The blood-brain barrier (BBB) is an integral part of the neurovascular unit (NVU). The NVU is comprised of endothelial cells that are interconnected by tight junctions resting on a parenchymal basement membrane ensheathed by pericytes, smooth muscle cells and a layer of astrocyte end feet. Circulating blood cells, such as leukocytes, complete the NVU. BBB disruption is common in several neurological diseases, but the molecular mechanisms involved remain largely unknown. We analyzed the role of TWIK-related potassium channel-1 (TREK1, encoded by KCNK2) in human and mouse endothelial cells and the BBB. TREK1 was downregulated in endothelial cells by treatment with interferon-γ (IFN-γ) and tumor necrosis factor-α (TNF-α). Blocking TREK1 increased leukocyte transmigration, whereas TREK1 activation had the opposite effect. We identified altered mitogen-activated protein (MAP) kinase signaling, actin remodeling and upregulation of cellular adhesion molecules as potential mechanisms of increased migration in TREK1-deficient (Kcnk2−/−) cells. In Kcnk2−/− mice, brain endothelial cells showed an upregulation of the cellular adhesion molecules ICAM1, VCAM1 and PECAM1 and facilitated leukocyte trafficking into the CNS. Following the induction of experimental autoimmune encephalomyelitis (EAE) by immunization with a myelin oligodendrocyte protein (MOG)35–55 peptide, Kcnk2−/− mice showed higher EAE severity scores that were accompanied by increased cellular infiltrates in the central nervous system (CNS). The severity of EAE was attenuated in mice given the amyotrophic lateral sclerosis drug riluzole or fed a diet enriched with linseed oil (which contains the TREK-1 activating omega-3 fatty acid α-linolenic acid). These beneficial effects were reduced in Kcnk2−/− mice, suggesting TREK-1 activating compounds may be used therapeutically to treat diseases related to BBB dysfunction.


Blood | 2012

Kininogen deficiency protects from ischemic neurodegeneration in mice by reducing thrombosis, blood-brain barrier damage, and inflammation

Friederike Langhauser; Eva Göb; Peter Kraft; Christian Geis; Joachim P. Schmitt; Marc Brede; Kerstin Göbel; Xavier Helluy; Mirko Pham; Martin Bendszus; Peter M. Jakob; Guido Stoll; Sven G. Meuth; Bernhard Nieswandt; Keith R. McCrae; Christoph Kleinschnitz

Thrombosis and inflammation are hallmarks of ischemic stroke still unamenable to therapeutic interventions. High-molecular-weight kininogen (KNG) is a central constituent of the contact-kinin system which represents an interface between thrombotic and inflammatory circuits and is critically involved in stroke development. Kng(-/-) mice are protected from thrombosis after artificial vessel wall injury and lack the proinflammatory mediator bradykinin. We investigated the consequences of KNG deficiency in models of ischemic stroke. Kng(-/-) mice of either sex subjected to transient middle cerebral artery occlusion developed dramatically smaller brain infarctions and less severe neurologic deficits without an increase in infarct-associated hemorrhage. This protective effect was preserved at later stages of infarction as well as in elderly mice. Targeting KNG reduced thrombus formation in ischemic vessels and improved cerebral blood flow, and reconstitution of KNG-deficient mice with human KNG or bradykinin restored clot deposition and infarct susceptibility. Moreover, mice deficient in KNG showed less severe blood-brain barrier damage and edema formation, and the local inflammatory response was reduced compared with controls. Because KNG appears to be instrumental in pathologic thrombus formation and inflammation but dispensable for hemostasis, KNG inhibition may offer a selective and safe strategy for combating stroke and other thromboembolic diseases.


Antioxidants & Redox Signaling | 2013

Neuroprotection after stroke by targeting NOX4 as a source of oxidative stress.

Kim A. Radermacher; Kirstin Wingler; Friederike Langhauser; Sebastian Altenhöfer; Pamela W. M. Kleikers; J. J. Rob Hermans; Martin Hrabě de Angelis; Christoph Kleinschnitz; Harald Schmidt

SIGNIFICANCE Stroke, a leading cause of death and disability, poses a substantial burden for patients, relatives, and our healthcare systems. Only one drug is approved for treating stroke, and more than 30 contraindications exclude its use in 90% of all patients. Thus, new treatments are urgently needed. In this review, we discuss oxidative stress as a pathomechanism of poststroke neurodegeneration and the inhibition of its source, type 4 nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX4), as a conceptual breakthrough in stroke therapy. RECENT ADVANCES Among potential sources of reactive oxygen species (ROS), the NOXes stand out as the only enzyme family that is solely dedicated to forming ROS. In rodents, three cerebrovascular NOXes exist: the superoxide-forming NOX1 and 2 and the hydrogen peroxide-forming NOX4. Studies using NOX1 knockout mice gave conflicting results, which overall do not point to a role for this isoform. Several reports find NOX2 to be relevant in stroke, albeit to variable and moderate degrees. In our hands, NOX4 is, by far, the major source of oxidative stress and neurodegeneration on ischemic stroke. CRITICAL ISSUES We critically discuss the tools that have been used to validate the roles of NOX in stroke. We also highlight the relevance of different animal models and the need for advanced quality control in preclinical stroke research. FUTURE DIRECTIONS The development of isoform-specific NOX inhibitors presents a precious tool for further clarifying the role and drugability of NOX homologues. This could pave the avenue for the first clinically effective neuroprotectant applied poststroke, and even beyond this, stroke could provide a proof of principle for antioxidative stress therapy.


Stroke | 2014

Blocking of α4 Integrin Does Not Protect From Acute Ischemic Stroke in Mice

Friederike Langhauser; Peter Kraft; Eva Göb; Jonas Leinweber; Michael K. Schuhmann; Kristina Lorenz; Mathias Gelderblom; Stefan Bittner; Sven G. Meuth; Heinz Wiendl; Tim Magnus; Christoph Kleinschnitz

Background and Purpose— T lymphocytes have recently been identified as key mediators of tissue damage in ischemic stroke. The interaction between very late antigen-4 (VLA-4) and vascular adhesion molecule-1 is crucial for the transvascular egress of T lymphocytes, and inhibition of this interaction by specific antibodies is a powerful strategy to combat autoimmune neuroinflammation. However, whether pharmacological blocking of T-lymphocyte trafficking is also protective during brain ischemia is still unclear. We investigated the efficacy of a monoclonal antibody directed against VLA-4 in mouse models of ischemic stroke. Methods— Transient and permanent middle cerebral artery occlusion was induced in male C57Bl/6 mice. Animals treated with a monoclonal anti-CD49d antibody (300 &mgr;g) 24 hours before or 3 hours after the onset of cerebral ischemia and stroke outcome, including infarct size, functional status, and mortality, were assessed between day 1 and day 7. The numbers of immune cells invading the ischemic brain were determined by immunocytochemistry and flow cytometry. Results— Blocking of VLA-4 significantly reduced the invasion of T lymphocytes and neutrophils on day 5 after middle cerebral artery occlusion and inhibited the upregulation of vascular adhesion molecule-1. However, the anti-CD49d antibody failed to influence stroke outcome positively irrespective of the model or the time point investigated. Conclusions— Pharmacological inhibition of the VLA-4/vascular adhesion molecule-1 axis in experimental stroke was ineffective in our hands. Our results cast doubt on the effectiveness of anti-CD49d as a stroke treatment. Further translational studies should be performed before testing anti–VLA-4 antibodies in patients with stroke.


Stroke | 2016

Thromboinflammation in Stroke Brain Damage

Simon F. De Meyer; Frederik Denorme; Friederike Langhauser; Eva Geuss; Felix Fluri; Christoph Kleinschnitz

The main goal of ischemic stroke treatment is rapid recanalization of the occluded blood vessel to limit brain injury and to salvage threatened cerebral tissue. To achieve early vessel recanalization, thrombolysis using recombinant tissue-type plasminogen activator is currently the only approved pharmacological intervention. Only recently, endovascular therapy has made its way into the clinic extending the therapeutic time-window and increasing reperfusion rates. However, despite fast restoration of blood vessel patency, progressive stroke still develops in many patients, which has led to the concept of reperfusion injury. During the past decades, many studies have been addressing the mechanisms underlying ischemic stroke damage and cerebral reperfusion injury, but the picture remains far from complete.1 It has become clear that both thrombotic and inflammatory pathways are important pathophysiologic contributors to ischemic brain damage. At ischemic vascular lesions, blood platelets adhere and become activated, increasing the risk of secondary thrombotic events.2 At the same time, cerebral ischemia elicits a strong inflammatory response involving upregulation of cell adhesion molecules and cytokines as well as adhesion, activation, and transmigration of several subsets of leukocytes.3 Interestingly, emerging insights indicate an important link between these thrombotic and inflammatory pathways in stroke, which led to the concept of thromboinflammation in stroke pathology. In this review, we focus on recently discovered thromboinflammatory pathways of ischemic stroke and discuss the clinical potential of targeting thromboinflammation as a novel treatment strategy in stroke management. An overview of the key components is given in Tables I to III in the online-only Data Supplement. Collagen, von Willebrand factor (vWF), and platelet glycoprotein (GP) Ib together form an important axis that is crucial for initial platelet adhesion at sites of vascular injury.4 On exposure of the subendothelial matrix, platelets are able to adhere to exposed collagen via their collagen receptors GP …


Stroke | 2012

C1-Inhibitor Protects From Brain Ischemia-Reperfusion Injury by Combined Antiinflammatory and Antithrombotic Mechanisms

Nadine Heydenreich; Marc W. Nolte; Eva Göb; Friederike Langhauser; Marion Hofmeister; Peter Kraft; Christiane Albert-Weissenberger; Marc Brede; Csanad Varallyay; Kerstin Göbel; Sven G. Meuth; Bernhard Nieswandt; Gerhard Dickneite; Guido Stoll; Christoph Kleinschnitz

Background and Purpose— Inflammation and thrombosis are pathophysiological hallmarks of ischemic stroke still unamenable to therapeutic interventions. The contact-kinin system represents an interface between inflammatory and thrombotic circuits and is involved in stroke development. C1-inhibitor counteracts activation of the contact-kinin system at multiple levels. We investigated the therapeutic potential of C1-inhibitor in models of ischemic stroke. Methods— Male and female C57Bl/6 mice and rats of different ages were subjected to middle cerebral artery occlusion and treated with C1-inhibitor after 1 hour or 6 hours. Infarct volumes and functional outcomes were assessed between day 1 and day 7, and findings were validated by magnetic resonance imaging. Blood–brain barrier damage, thrombus formation, and the local inflammatory response were determined poststroke. Results— Treatment with 15.0 U C1-inhibitor, but not 7.5 U, 1 hour after stroke reduced infarct volumes by ≈60% and improved clinical scores in mice of either sex on day 1. This protective effect was preserved at later stages of infarction as well as in elderly mice and in another species, ie, rats. Delayed C1-inhibitor treatment still improved clinical outcome. Blood–brain barrier damage, edema formation, and inflammation were significantly lower compared with controls. Moreover, C1-inhibitor showed strong antithrombotic effects. Conclusions— C1-inhibitor is a multifaceted antiinflammatory and antithrombotic compound that protects from ischemic neurodegeneration in clinically meaningful settings.


Nature Communications | 2016

Blood coagulation factor XII drives adaptive immunity during neuroinflammation via CD87-mediated modulation of dendritic cells

Kerstin Göbel; Susann Pankratz; Chloi-Magdalini Asaridou; Alexander M. Herrmann; Stefan Bittner; Monika Merker; Tobias Ruck; Sarah Glumm; Friederike Langhauser; Peter Kraft; Thorsten F. Krug; Johanna Breuer; Martin Herold; Catharina C. Gross; Denise Beckmann; Adelheid Korb-Pap; Michael K. Schuhmann; Stefanie Kuerten; Ioannis Mitroulis; Clemens Ruppert; Marc W. Nolte; Con Panousis; Luisa Klotz; Beate E. Kehrel; Thomas Korn; Harald Langer; Thomas Pap; Bernhard Nieswandt; Heinz Wiendl; Triantafyllos Chavakis

Aberrant immune responses represent the underlying cause of central nervous system (CNS) autoimmunity, including multiple sclerosis (MS). Recent evidence implicated the crosstalk between coagulation and immunity in CNS autoimmunity. Here we identify coagulation factor XII (FXII), the initiator of the intrinsic coagulation cascade and the kallikrein–kinin system, as a specific immune cell modulator. High levels of FXII activity are present in the plasma of MS patients during relapse. Deficiency or pharmacologic blockade of FXII renders mice less susceptible to experimental autoimmune encephalomyelitis (a model of MS) and is accompanied by reduced numbers of interleukin-17A-producing T cells. Immune activation by FXII is mediated by dendritic cells in a CD87-dependent manner and involves alterations in intracellular cyclic AMP formation. Our study demonstrates that a member of the plasmatic coagulation cascade is a key mediator of autoimmunity. FXII inhibition may provide a strategy to combat MS and other immune-related disorders.


Annals of Neurology | 2015

Blocking of plasma kallikrein ameliorates stroke by reducing thromboinflammation

Eva Göb; Stephan Reymann; Friederike Langhauser; Michael K. Schuhmann; Peter Kraft; Ina Thielmann; Kerstin Göbel; Marc Brede; György A. Homola; Laszlo Solymosi; Guido Stoll; Christian Geis; Sven G. Meuth; Bernhard Nieswandt; Christoph Kleinschnitz

Recent evidence suggests that ischemic stroke is a thromboinflammatory disease. Plasma kallikrein (PK) cleaves high–molecular‐weight kininogen to release bradykinin (BK) and is a key constituent of the proinflammatory contact‐kinin system. In addition, PK can activate coagulation factor XII, the origin of the intrinsic coagulation cascade. Thus, PK triggers 2 important pathological pathways of stroke formation, thrombosis and inflammation.


Blood | 2016

ADAMTS13-mediated thrombolysis of t-PA–resistant occlusions in ischemic stroke in mice

Frederik Denorme; Friederike Langhauser; Linda Desender; Aline Vandenbulcke; Hanspeter Rottensteiner; Barbara Plaimauer; Olivier François; Tommy Andersson; Hans Deckmyn; Friedrich Scheiflinger; Christoph Kleinschnitz; Karen Vanhoorelbeke; Simon F. De Meyer

Rapid vascular recanalization forms the basis for successful treatment of cerebral ischemia. Currently, tissue plasminogen activator (t-PA) is the only approved thrombolytic drug for ischemic stroke. However, t-PA does not always result in efficient thrombus dissolution and subsequent blood vessel recanalization. To better understand thrombus composition, we analyzed thrombi retrieved from ischemic stroke patients and found a distinct presence of von Willebrand factor (VWF) in various samples. Thrombi contained on average 20.3% ± 10.1% VWF, and this was inversely correlated with thrombus red blood cell content. We hypothesized that ADAMTS13 can exert a thrombolytic effect in VWF-containing thrombi in the setting of stroke. To test this, we generated occlusive VWF-rich thrombi in the middle cerebral artery (MCA) of mice. Infusion of t-PA did not dissolve these MCA occlusions. Interestingly, administration of ADAMTS13 5 minutes after occlusion dose-dependently dissolved these t-PA-resistant thrombi resulting in fast restoration of MCA patency and consequently reduced cerebral infarct sizes (P < .005). Delayed ADAMTS13 administration 60 minutes after occlusion was still effective but to a lesser extent (P < .05). These data show for the first time a potent thrombolytic activity of ADAMTS13 in the setting of stroke, which might become useful in treatment of acute ischemic stroke.

Collaboration


Dive into the Friederike Langhauser's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Eva Göb

University of Würzburg

View shared research outputs
Top Co-Authors

Avatar

Marc Brede

University of Würzburg

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Peter Kraft

University of Würzburg

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge