Network


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

Hotspot


Dive into the research topics where Karl Frei is active.

Publication


Featured researches published by Karl Frei.


Neuroscience Letters | 1991

Murine brain macrophages induce NMDA receptor mediated neurotoxicity in vitro by secreting glutamate

Daniela Piani; Karl Frei; Kim Q. Do; Michel Cuenod; Adriano Fontana

Supernatants (SN) of brain macrophages in culture induce death of cerebellar granule cells in vitro, while those of astrocytes and endothelial cells do not. This toxicity can be prevented by N-methyl-D-aspartate (NMDA) receptor antagonists. Macrophage SN contain high concentrations of glutamate. Reducing the glutamate level of macrophage SN, either by exposure to astrocytes or by enzymatic degradation abolished the toxic effect. Thus, macrophage neurotoxicity is mediated by glutamate acting on NMDA receptors, and might play a role in vivo in traumatic and cerebrovascular brain lesions.


Journal of Clinical Investigation | 1994

Anti-Fas/APO-1 antibody-mediated apoptosis of cultured human glioma cells. Induction and modulation of sensitivity by cytokines.

Michael Weller; Karl Frei; Peter Groscurth; Peter H. Krammer; Yasuhiro Yonekawa; Adriano Fontana

Fas/APO-1 is a transmembrane protein of the nerve growth factor/TNF alpha receptor family which signals apoptotic cell death in susceptible target cells. We have investigated the susceptibility of seven human malignant glioma cell lines to Fas/APO-1-dependent apoptosis. Sensitivity to Fas/APO-1 antibody-mediated cell killing correlated with cell surface expression of Fas/APO-1. Expression of Fas/APO-1 as well as Fas/APO-1-dependent cytotoxicity were augmented by preexposure of human malignant glioma cells to IFN gamma and TNF alpha. Further, pretreatment with TGF beta 2, IL1 and IL8 enhanced Fas/APO-1 antibody-induced glioma cell apoptosis whereas other cytokines including TNF beta, IL6, macrophage colony-stimulating factor, IL10 and IL13 had no such effect. None of the human malignant glioma cell lines was susceptible to TNF alpha-induced cytotoxicity. Fas/APO-1 antibody-sensitive glioma cell lines (n = 5), but not Fas/APO-1 antibody-resistant glioma cell lines (n = 2), became sensitive to TNF alpha when co-treated with inhibitors of RNA and protein synthesis. Resistance of human glioma cells to Fas/APO-1 antibody-mediated apoptosis was mainly related to low level expression of Fas/APO-1 and appeared not to be linked to overexpression of the anti-apoptotic protooncogene, bcl-2. Given the resistance of human malignant glioma to surgery, irradiation, chemotherapy and immunotherapy, we propose that Fas/APO-1 may be a promising target for a novel locoregionary approach to human malignant glioma. This strategy gains support from the demonstration of Fas/APO-1 expression in ex vivo human malignant glioma specimens and from the absence of Fas/APO-1 in normal human brain parenchyma.


The EMBO Journal | 1987

T cell suppressor factor from human glioblastoma cells is a 12.5-kd protein closely related to transforming growth factor-beta.

Michael Wrann; Stefan Bodmer; R de Martin; Christine Siepl; Renate Hofer-Warbinek; Karl Frei; Erhard Hofer; Adriano Fontana

T cell suppressor factor produced by human glioblastoma cells inhibits T cell proliferation in vitro and more specifically interferes with interleukin‐2 (IL‐2)‐dependent T cell growth. Here we report the purification of this factor from conditioned medium of the human glioblastoma cell line 308. Amino‐terminal sequence analysis of the 12.5‐kd protein demonstrates that eight out of the first 20 amino acids are identical to human transforming growth factor‐beta. Purified glioblastoma‐derived T cell suppressor factor and transforming growth factor‐beta from porcine platelets inhibit both IL‐2‐induced proliferation of ovalbumin‐specific T helper cells and lectin‐induced thymocyte proliferation with similar specific activities. If released by glioblastoma cells in vivo, the factor may contribute to impaired immunosurveillance and to the cellular immunodeficiency state detected in the patients.


The Journal of Infectious Diseases | 2002

Toll-like receptor 2-deficient mice are highly susceptible to Streptococcus pneumoniae meningitis because of reduced bacterial clearing and enhanced inflammation.

Hakim Echchannaoui; Karl Frei; Christian Schnell; Stephen L. Leib; Werner Zimmerli; Regine Landmann

Toll-like receptor-2 (TLR2) mediates host responses to gram-positive bacterial wall components. TLR2 function was investigated in a murine Streptococcus pneumoniae meningitis model in wild-type (wt) and TLR2-deficient (TLR2(-/-)) mice. TLR2(-/-) mice showed earlier time of death than wt mice (P<.02). Plasma interleukin-6 levels and bacterial numbers in blood and peripheral organs were similar for both strains. With ceftriaxone therapy, none of the wt but 27% of the TLR2(-/-) mice died (P<.04). Beyond 3 hours after infection, TLR2(-/-) mice had higher bacterial loads in brain than did wt mice, as assessed with luciferase-tagged S. pneumoniae by means of a Xenogen-CCD (charge-coupled device) camera. After 24 h, tumor necrosis factor activity was higher in cerebrospinal fluid of TLR2(-/-) than wt mice (P<.05) and was related to increased blood-brain barrier permeability (Evans blue staining, P<.02). In conclusion, the lack of TLR2 was associated with earlier death from meningitis, which was not due to sepsis but to reduced brain bacterial clearing, followed by increased intrathecal inflammation.


European Journal of Immunology | 1998

IL-6-deficient mice resist myelin oligodendrocyte glycoprotein-induced autoimmune encephalomyelitis.

Hans-Pietro Eugster; Karl Frei; Manfred Kopf; Hans Lassmann; Adriano Fontana

Experimental autoimmune encephalomyelitis (EAE) is induced by immunization with myelin components including myelin oligodendrocyte glycoprotein (MOG). Myelin‐specific Th1 cells enter the central nervous system (CNS) via binding of very late antigen 4 (VLA‐4) to the endothelial vascular cell adhesion molecule 1 (VCAM‐1). In the present study, mice with a homologous disruption of the gene encoding IL‐6 are found to be resistant to MOG‐induced EAE as evidenced by absence of clinical symptoms, minimal infiltration of CD3+ T cells and monocytes into the CNS and lack of demyelination. The failure to induce EAE in IL‐6− / − mice is not due to the absence of priming, since lymphocytes of immunized IL‐6− / − mice proliferate in response to MOG and produce pro‐inflammatory cytokines including IL‐2 and IFN‐γ. However, in MOG‐immunized IL‐6− / − mice, serum anti‐MOG antibody titers were found to be drastically reduced. This observation is unlikely to be responsible for resistance to EAE, because B cell‐deficient (μMT) mice proved to be fully susceptible to the disease. A striking difference between MOG‐immunized wild‐type (wt) and IL‐6− / − mice was the expression of endothelial VCAM‐1 and ICAM‐1, which were dramatically up‐regulated in the CNS in wt but not in IL‐6− / − mice. Taking into account recent studies on the role of VCAM‐1 in the entry of Th1 cells into the CNS, the absence of VCAM‐1 on endothelial cells in IL‐6− / − mice may explain their resistance to EAE.


Journal of Neuroimmunology | 1989

Human immunodeficiency virus type 1 (HIV-1) infection of the central nervous system; an evaluation of cytokines in cerebrospinal fluid

Paolo Gallo; Karl Frei; Christiane Rordorf; Janis Lazdins; B. Tavolato; Adriano Fontana

Cytokines play an important role not only for initiation of immune reactivity but also for development of tissue injury. Of 38 patients infected with human immunodeficiency virus type 1 (HIV-1) interleukin-1 beta (IL-1 beta) and interleukin-6 (IL-6) were identified in cerebrospinal fluid (CSF) of 22 (58%) and 16 (42%) patients, respectively. Among the IL-1 beta- and IL-6-positive CSF were eight of 15 HIV-1 patients with no clinical signs of central nervous system involvement and four of five patients with acquired immunodeficiency syndrome (AIDS) dementia complex. The presence of IL-6 was often associated with IL-1 beta and soluble interleukin-2 receptor in CSF as well as with intrathecal IgG synthesis. In none of the CSF samples tumor necrosis factor-alpha or interleukin-2 was detected.


Brain Research Reviews | 2001

Molecular and cellular permeability control at the blood-brain barrier

Sergio M. Gloor; Marco Wachtel; Marc F Bolliger; Hideyuki Ishihara; Regine Landmann; Karl Frei

The blood-brain barrier (BBB) is formed by brain capillary endothelial cells. These cells have at least three properties which distinguish them from their peripheral counterparts: (1) tight junctions (TJs) of extremely low permeability; (2) low rates of fluid-phase endocytosis; (3) specific transport and carrier molecules. In combination, these features restrict the nonspecific flux of ions, proteins, and other substances into the central nervous system (CNS) environment. The restriction protects neurons from harmful compositional fluctuations occurring in the blood and allows uptake of essential molecules. Breakdown of the BBB is associated with a variety of CNS disorders and results in aggravation of the condition. Restoration of the BBB is thus one strategy during therapy of CNS diseases. Its success depends on a precise knowledge of the structural and functional principles underlying BBB functionality. In this review we have tried to summarise the current knowledge of TJs, including information gained from non-neuronal systems, and describe selected mechanisms involved in permeability regulation.


European Journal of Immunology | 1999

Severity of symptoms and demyelination in MOG‐induced EAE depends on TNFR1

Hans-Pietro Eugster; Karl Frei; Rosilla Bachmann; Horst Bluethmann; Hans Lassmann; Adriano Fontana

The individual role of tumor necrosis factor receptor 1 (TNFR1) and TNFR2 signaling in experimental autoimmune encephalomeylitis (EAE) was investigated using mice lacking TNFR1 (TNFR1− / −), TNFR2 (TNFR2− / −) as well as double receptor (TNFR1 / 2− / −) and double ligand (TNF / LTα− / −) knockout mice. In wild‐type (wt) mice immunized with myelin oligodendrocyte glycoprotein (MOG) peptide 35 – 55 the clinical course is characterized by an acute disease onset with peak disease scores and a consecutive chronic phase lasting up to 60 days. Compared to control mice, TNF / LTα‐deficient mice showed a significant delay in disease onset and a remarkable reduction in demyelination which was, however, associated with increased inflammation. In TNFR1− / − and TNFR1 / 2− / − mice, the disease course was comparable to TNF / LTα‐deficient mice but rather monophasic and less severe at late time points. Likewise only minimal spinal cord demyelination became apparent. In contrast, the course of EAE in TNFR2− / − mice was severe and associated with remarkable demyelination. Taken together these findings define TNFR1 as crucial mediator in MOG‐induced EAE and suggest a protective role for TNFR2 signaling in the clinical course of EAE.


European Journal of Immunology | 1998

Mice with an inactivation of the inducible nitric oxide synthase gene are susceptible to experimental autoimmune encephalomyelitis.

Ulrike Sahrbacher; Franziska Lechner; Hans-Pietro Eugster; Karl Frei; Hans Lassmann; Adriano Fontana

Nitric oxide (NO) generated by the inducible nitric oxide synthase (iNOS) has been implicated in the pathogenesis of experimental autoimmune encephalomyelitis (EAE). In this study mice genetically deficient for iNOS are shown to be susceptible to EAE induced by immunization with myelin oligodendrocyte glycoprotein (MOG). In iNOS (–/–) mice the course of disease was earlier in onset and more aggressive compared to control animals. A disease‐relevant compensatory up‐regulation of neuronal (n)NOS and endothelial (e)NOS with increased production of NO in iNOS (–/–) mice is excluded by 1) the failure to detect increased nNOS and eNOS mRNA, 2) the absence of detection of nitrosylated tyrosine residues in EAE tissue indicating absence of NO‐derived peroxynitrite, and 3) the lack of disease‐preventing effects of NG‐nitro‐L‐arginine methylester. In conclusion, these results do not support the hypothesis that NO is crucial for the development of EAE.


The Journal of Neuroscience | 2011

Brain Infiltration of Leukocytes Contributes to the Pathophysiology of Temporal Lobe Epilepsy

Michela Zattoni; Maria Luisa Mura; Francine Deprez; Reto A. Schwendener; Britta Engelhardt; Karl Frei; Jean-Marc Fritschy

Clinical and experimental evidence indicates that inflammatory processes contribute to the pathophysiology of epilepsy, but underlying mechanisms remain mostly unknown. Using immunohistochemistry for CD45 (common leukocyte antigen) and CD3 (T-lymphocytes), we show here microglial activation and infiltration of leukocytes in sclerotic tissue from patients with mesial temporal lobe epilepsy (TLE), as well as in a model of TLE (intrahippocampal kainic acid injection), characterized by spontaneous, nonconvulsive focal seizures. Using specific markers of lymphocytes, microglia, macrophages, and neutrophils in kainate-treated mice, we investigated with pharmacological and genetic approaches the contribution of innate and adaptive immunity to kainate-induced inflammation and neurodegeneration. Furthermore, we used EEG analysis in mutant mice lacking specific subsets of lymphocytes to explore the significance of inflammatory processes for epileptogenesis. Blood–brain barrier disruption and neurodegeneration in the kainate-lesioned hippocampus were accompanied by sustained ICAM-1 upregulation, microglial cell activation, and infiltration of CD3+ T-cells. Moreover, macrophage infiltration was observed, selectively in the dentate gyrus where prominent granule cell dispersion was evident. Unexpectedly, depletion of peripheral macrophages by systemic clodronate liposome administration affected granule cell survival. Neurodegeneration was aggravated in kainate-lesioned mice lacking T- and B-cells (RAG1-knock-out), because of delayed invasion by Gr-1+ neutrophils. Most strikingly, these mutant mice exhibited early onset of spontaneous recurrent seizures, suggesting a strong impact of immune-mediated responses on network excitability. Together, the concerted action of adaptive and innate immunity triggered locally by intrahippocampal kainate injection contributes seizure-suppressant and neuroprotective effects, shedding new light on neuroimmune interactions in temporal lobe epilepsy.

Collaboration


Dive into the Karl Frei's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
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