Network


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

Hotspot


Dive into the research topics where Nils C. Gassen is active.

Publication


Featured researches published by Nils C. Gassen.


Biological Psychiatry | 2011

FK506 binding protein 5 shapes stress responsiveness: modulation of neuroendocrine reactivity and coping behavior.

Chadi Touma; Nils C. Gassen; Leonie Herrmann; Joyce Cheung-Flynn; Dominik R. Büll; Irina A. Ionescu; Jan-Michael Heinzmann; Alana Knapman; Anna Siebertz; Anna-Mareike Depping; Jakob Hartmann; Felix Hausch; Mathias V. Schmidt; Florian Holsboer; Marcus Ising; Marc B. Cox; Ulrike Schmidt; Theo Rein

BACKGROUND The Hsp90 cochaperone FK506 binding protein 5 (FKBP5) is an established regulator of the glucocorticoid receptor (GR), and numerous genetic studies have linked it to stress-related diseases such as major depression or posttraumatic stress disorder. However, translational studies including genetic animal models are lacking. METHODS Mice deficient of FKBP5 were generated and analyzed in comparison with wildtype littermates. They were subjected to several test paradigms characterizing their emotionality, stress reactivity, and coping behavior as well as hypothalamus-pituitary-adrenal axis function and regulation. Moreover, protein expression of GR and FKBP5 was determined in different brain structures 8 days after stress exposure. The combined dexamethasone/corticotropin-releasing hormone test was performed both in mice and healthy human subjects of different FKBP5 genotypes. The GR function was evaluated by reporter gene assays. RESULTS Under basal conditions, deletion of FKBP5 did not change exploratory drive, locomotor activity, anxiety-related behavior, stress-coping, or depression-like behavior. After exposure to different acute stressors of sufficient intensity, however, it led to a more active coping behavior. Moreover, loss of FKBP5 decreased hypothalamus-pituitary-adrenal axis reactivity and GR expression changes in response to stressors. In mice and humans, the FKBP5 genotype also determined the outcome of the dexamethasone/corticotropin-releasing hormone test. CONCLUSIONS This study in mice and humans presents FKBP5 as a decisive factor for the physiological stress response, shaping neuroendocrine reactivity as well as coping behavior. This lends strong support to the concept emerging from human studies of FKBP5 as important factor governing gene-environment interactions relevant for the etiology of affective disorders.


Neuropsychopharmacology | 2016

Gene–Stress–Epigenetic Regulation of FKBP5 : Clinical and Translational Implications

Anthony S. Zannas; Tobias Wiechmann; Nils C. Gassen; Elisabeth B. Binder

Stress responses and related outcomes vary markedly across individuals. Elucidating the molecular underpinnings of this variability is of great relevance for developing individualized prevention strategies and treatments for stress-related disorders. An important modulator of stress responses is the FK506-binding protein 51 (FKBP5/FKBP51). FKBP5 acts as a co-chaperone that modulates not only glucocorticoid receptor activity in response to stressors but also a multitude of other cellular processes in both the brain and periphery. Notably, the FKBP5 gene is regulated via complex interactions among environmental stressors, FKBP5 genetic variants, and epigenetic modifications of glucocorticoid-responsive genomic sites. These interactions can result in FKBP5 disinhibition that has been shown to contribute to a number of aberrant phenotypes in both rodents and humans. Consequently, FKBP5 blockade may hold promise as treatment intervention for stress-related disorders, and recently developed selective FKBP5 blockers show encouraging results in vitro and in rodent models. Although risk for stress-related disorders is conferred by multiple environmental and genetic factors, the findings related to FKBP5 illustrate how a deeper understanding of the molecular and systemic mechanisms underlying specific gene–environment interactions may provide insights into the pathogenesis of stress-related disorders.


The International Journal of Neuropsychopharmacology | 2015

Hypothalamic-pituitary-adrenal axis dysfunction and illness progression in bipolar disorder.

Gabriel Rodrigo Fries; Mirela Paiva Vasconcelos-Moreno; Carolina Gubert; Barbara T. Santos; Juliana Sartori; Bárbara Eisele; Pamela Ferrari; Adam Fijtman; Joëlle Rüegg; Nils C. Gassen; Flávio Kapczinski; Theo Rein; Marcia Kauer-Sant’Anna

Background: Impaired stress resilience and a dysfunctional hypothalamic-pituitary-adrenal (HPA) axis are suggested to play key roles in the pathophysiology of illness progression in bipolar disorder (BD), but the mechanisms leading to this dysfunction have never been elucidated. This study aimed to examine HPA axis activity and underlying molecular mechanisms in patients with BD and unaffected siblings of BD patients. Methods: Twenty-four euthymic patients with BD, 18 siblings of BD patients, and 26 healthy controls were recruited for this study. All subjects underwent a dexamethasone suppression test followed by analyses associated with the HPA axis and the glucocorticoid receptor (GR). Results: Patients with BD, particularly those at a late stage of illness, presented increased salivary post-dexamethasone cortisol levels when compared to controls (p = 0.015). Accordingly, these patients presented reduced ex vivo GR responsiveness (p = 0.008) and increased basal protein levels of FK506-binding protein 51 (FKBP51, p = 0.012), a co-chaperone known to desensitize GR, in peripheral blood mononuclear cells. Moreover, BD patients presented increased methylation at the FK506-binding protein 5 (FKBP5) gene. BD siblings presented significantly lower FKBP51 protein levels than BD patients, even though no differences were found in FKBP5 basal mRNA levels. Conclusions: Our data suggest that the epigenetic modulation of the FKBP5 gene, along with increased FKBP51 levels, is associated with the GR hyporesponsiveness seen in BD patients. Our findings are consistent with the notion that unaffected first-degree relatives of BD patients share biological factors that influence the disorder, and that such changes are more pronounced in the late stages of the illness.


Science Signaling | 2015

Chaperoning epigenetics: FKBP51 decreases the activity of DNMT1 and mediates epigenetic effects of the antidepressant paroxetine

Nils C. Gassen; Gabriel Rodrigo Fries; Anthony S. Zannas; Jakob Hartmann; Jürgen Zschocke; Kathrin Hafner; Tania Carrillo-Roa; Jessica Steinbacher; S. Nicole Preißinger; Lianne Hoeijmakers; M. Knop; Frank Weber; Stefan Kloiber; Susanne Lucae; George P. Chrousos; Thomas Carell; Marcus Ising; Elisabeth B. Binder; Mathias V. Schmidt; Joëlle Rüegg; Theo Rein

Chaperone switching at the kinase CDK5 mediates epigenetic effects of antidepressants. Antidepressants chaperone DNA methylation Epigenetic changes are associated with depression. Some depressed patients have increased DNA methylation and decreased expression of the gene encoding BDNF, a secreted factor important for synaptic plasticity. Rein et al. found that some antidepressants inhibit epigenetic changes by causing a switch in chaperone binding to the DNA methyltransferase DNMT1. The chaperones FKBP51 and FKBP52 competed for binding to CDK5, a kinase that activates DNMT1. The authors found that cells from depressed patients or cultured mouse astrocytes exposed to the antidepressant paroxetine favored the FKBP51-CDK5 interaction, resulting in reduced activity of DNMT1 and DNA methylation, and increased the expression of BDNF. These effects of paroxetine on patient blood cells isolated before therapy correlated with a positive clinical response to antidepressants, suggesting that a simple blood test may aid in personalizing treatment for depression. Epigenetic processes, such as DNA methylation, and molecular chaperones, including FK506-binding protein 51 (FKBP51), are independently implicated in stress-related mental disorders and antidepressant drug action. FKBP51 associates with cyclin-dependent kinase 5 (CDK5), which is one of several kinases that phosphorylates and activates DNA methyltransferase 1 (DNMT1). We searched for a functional link between FKBP51 (encoded by FKBP5) and DNMT1 in cells from mice and humans, including those from depressed patients, and found that FKBP51 competed with its close homolog FKBP52 for association with CDK5. In human embryonic kidney (HEK) 293 cells, expression of FKBP51 displaced FKBP52 from CDK5, decreased the interaction of CDK5 with DNMT1, reduced the phosphorylation and enzymatic activity of DNMT1, and diminished global DNA methylation. In mouse embryonic fibroblasts and primary mouse astrocytes, FKBP51 mediated several effects of paroxetine, namely, decreased the protein-protein interactions of DNMT1 with CDK5 and FKBP52, reduced phosphorylation of DNMT1, and decreased the methylation and increased the expression of the gene encoding brain-derived neurotrophic factor (Bdnf). In human peripheral blood cells, FKBP5 expression inversely correlated with both global and BDNF methylation. Peripheral blood cells isolated from depressed patients that were then treated ex vivo with paroxetine revealed that the abundance of BDNF positively correlated and phosphorylated DNMT1 inversely correlated with that of FKBP51 in cells and with clinical treatment success in patients, supporting the relevance of this FKBP51-directed pathway that prevents epigenetic suppression of gene expression.


Autophagy | 2015

FKBP5/FKBP51 enhances autophagy to synergize with antidepressant action.

Nils C. Gassen; Jakob Hartmann; Mathias V. Schmidt; Theo Rein

Levels of autophagy markers rise upon treatment of cells with antidepressants. However, it was not known whether this phenomenon might be linked to other antidepressant pathways or to any physiological effect. In this punctum, we summarize and discuss our recent findings that provide evidence for a role of the cochaperone FKBP5/FKBP51 (FK506 binding protein 5) in autophagy as a prerequisite for antidepressant action in cells, mice, and humans. FKBP5 associates with BECN1, changes its phosphorylation and protein levels and enhances markers of autophagy and autophagic flux. The effects of antidepressants on autophagy as well as their physiological effects in mice and human depend on FKBP5.


Neuroscience & Biobehavioral Reviews | 2017

Life stress, glucocorticoid signaling, and the aging epigenome: Implications for aging-related diseases

Nils C. Gassen; George P. Chrousos; Elisabeth B. Binder; Anthony S. Zannas

HighlightsEpigenetic mechanisms may contribute to the known link between life stress and aging‐related phenotypes.Glucocorticoid signaling may mediate the molecular effects of life stress on the aging epigenome.The proposed model could have implications for the prevention and treatment of aging‐related diseases. ABSTRACT Life stress has been associated with accelerated cellular aging and increased risk for developing aging‐related diseases; however, the underlying molecular mechanisms remain elusive. A highly relevant process that may underlie this association is epigenetic regulation. In this review, we build upon existing evidence to propose a model whereby exposure to life stress, in part via its effects on the hypothalamic‐pituitary axis and the glucocorticoid signaling system, may alter the epigenetic landscape across the lifespan and, consequently, influence genomic regulation and function in ways that are conducive to the development of aging‐related diseases. This model is supported by recent studies showing that life stressors and stress‐related phenotypes can accelerate epigenetic aging, a measure that is based on DNA methylation prediction of chronological age and has been associated with several aging‐related disease phenotypes. We discuss the implications of this model for the prevention and treatment of aging‐related diseases, as well as the challenges and limitations of this line of research.


Journal of Sleep Research | 2014

Deficiency of FK506-binding protein (FKBP) 51 alters sleep architecture and recovery sleep responses to stress in mice

Stefana Albu; Christoph P.N. Romanowski; M. Letizia Curzi; Vladimira Jakubcakova; Cornelia Flachskamm; Nils C. Gassen; Jakob Hartmann; Mathias V. Schmidt; Ulrike Schmidt; Theo Rein; Florian Holsboer; Felix Hausch; Marcelo Paez-Pereda; Mayumi Kimura

FK506‐binding protein 51 (FKBP51) is a co‐chaperone of the glucocorticoid receptor, functionally linked to its activity via an ultra‐short negative feedback loop. Thus, FKBP51 plays an important regulatory role in the hypothalamic–pituitary–adrenocortical (HPA) axis necessary for stress adaptation and recovery. Previous investigations illustrated that HPA functionality is influenced by polymorphisms in the gene encoding FKBP51, which are associated with both increased protein levels and depressive episodes. Because FKBP51 is a key molecule in stress responses, we hypothesized that its deletion impacts sleep. To study FKBP51‐involved changes in sleep, polysomnograms of FKBP51 knockout (KO) mice and wild‐type (WT) littermates were compared at baseline and in the recovery phase after 6‐h sleep deprivation (SD) and 1‐h restraint stress (RS). Using another set of animals, the 24‐h profiles of hippocampal free corticosterone levels were also determined. The most dominant effect of FKBP51 deletion appeared as increased nocturnal wake, where the bout length was significantly extended while non‐rapid eye movement sleep (NREMS) and rapid eye movement sleep were rather suppressed. After both SD and RS, FKBP51KO mice exhibited less recovery or rebound sleep than WTs, although slow‐wave activity during NREMS was higher in KOs, particularly after SD. Sleep compositions of KOs were nearly opposite to sleep profiles observed in human depression. This might result from lower levels of free corticosterone in FKBP51KO mice, confirming reduced HPA reactivity. The results indicate that an FKBP51 deletion yields a pro‐resilience sleep phenotype. FKBP51 could therefore be a therapeutic target for stress‐induced mood and sleep disorders.


Neuropsychopharmacology | 2015

Homer1/mGluR5 Activity Moderates Vulnerability to Chronic Social Stress

Klaus V. Wagner; Jakob Hartmann; Christiana Labermaier; Alexander S. Häusl; Gengjing Zhao; Daniela Harbich; Bianca Schmid; Xiao-Dong Wang; Sara Santarelli; Christine Kohl; Nils C. Gassen; Natalie Matosin; Marcel Schieven; Christian Webhofer; Christoph W. Turck; Lothar Lindemann; Georg Jaschke; Joseph G. Wettstein; Theo Rein; Marianne B. Müller; Mathias V. Schmidt

Stress-induced psychiatric disorders, such as depression, have recently been linked to changes in glutamate transmission in the central nervous system. Glutamate signaling is mediated by a range of receptors, including metabotropic glutamate receptors (mGluRs). In particular, mGluR subtype 5 (mGluR5) is highly implicated in stress-induced psychopathology. The major scaffold protein Homer1 critically interacts with mGluR5 and has also been linked to several psychopathologies. Yet, the specific role of Homer1 in this context remains poorly understood. We used chronic social defeat stress as an established animal model of depression and investigated changes in transcription of Homer1a and Homer1b/c isoforms and functional coupling of Homer1 to mGluR5. Next, we investigated the consequences of Homer1 deletion, overexpression of Homer1a, and chronic administration of the mGluR5 inverse agonist CTEP (2-chloro-4-((2,5-dimethyl-1-(4-(trifluoromethoxy)phenyl)-1H-imidazol-4-yl)ethynyl)pyridine) on the effects of chronic stress. In mice exposed to chronic stress, Homer1b/c, but not Homer1a, mRNA was upregulated and, accordingly, Homer1/mGluR5 coupling was disrupted. We found a marked hyperactivity behavior as well as a dysregulated hypothalamic–pituitary–adrenal axis activity in chronically stressed Homer1 knockout (KO) mice. Chronic administration of the selective and orally bioavailable mGluR5 inverse agonist, CTEP, was able to recover behavioral alterations induced by chronic stress, whereas overexpression of Homer1a in the hippocampus led to an increased vulnerability to chronic stress, reflected in an increased physiological response to stress as well as enhanced depression-like behavior. Overall, our results implicate the glutamatergic system in the emergence of stress-induced psychiatric disorders, and support the Homer1/mGluR5 complex as a target for the development of novel antidepressant agents.


Molecular Psychiatry | 2016

FKBP51 inhibits GSK3β and augments the effects of distinct psychotropic medications

Nils C. Gassen; Jakob Hartmann; Anthony S. Zannas; A Kretzschmar; Jürgen Zschocke; G Maccarrone; Kathrin Hafner; Andreas Zellner; Lorenz K. Kollmannsberger; Klaus V. Wagner; Divya Mehta; Stefan Kloiber; Christoph W. Turck; Susanne Lucae; George P. Chrousos; Florian Holsboer; Elisabeth B. Binder; Marcus Ising; Mathias V. Schmidt; Theo Rein

Psychotropic medications target glycogen synthase kinase 3β (GSK3β), but the functional integration with other factors relevant for drug efficacy is poorly understood. We discovered that the suggested psychiatric risk factor FK506 binding protein 51 (FKBP51) increases phosphorylation of GSK3β at serine 9 (pGSK3βS9). FKBP51 associates with GSK3β mainly through its FK1 domain; furthermore, it also changes GSK3βs heterocomplex assembly by associating with the phosphatase PP2A and the kinase cyclin-dependent kinase 5. FKBP51 acts through GSK3β on the downstream targets Tau, β-catenin and T-cell factor/lymphoid enhancing factor (TCF/LEF). Lithium and the antidepressant (AD) paroxetine (PAR) functionally synergize with FKBP51, as revealed by reporter gene and protein association analyses. Deletion of FKBP51 blunted the PAR- or lithium-induced increase in pGSK3βS9 in cells and mice and attenuated the behavioral effects of lithium treatment. Clinical improvement in depressive patients was predicted by baseline GSK3β pathway activity and by pGSK3βS9 reactivity to ex vivo treatment of peripheral blood mononuclear lymphocytes with lithium or PAR. In sum, FKBP51-directed GSK3β activity contributes to the action of psychotropic medications. Components of the FKBP51–GSK3β pathway may be useful as biomarkers predicting AD response and as targets for the development of novel ADs.


npj Schizophrenia | 2016

Molecular evidence of synaptic pathology in the CA1 region in schizophrenia.

Natalie Matosin; Francesca Fernandez-Enright; Jeremy S. Lum; Martin Engel; Jessica L. Andrews; Nils C. Gassen; Klaus V. Wagner; Mathias V. Schmidt; Kelly A. Newell

Alterations of postsynaptic density (PSD)95-complex proteins in schizophrenia ostensibly induce deficits in synaptic plasticity, the molecular process underlying cognitive functions. Although some PSD95-complex proteins have been previously examined in the hippocampus in schizophrenia, the status of other equally important molecules is unclear. This is especially true in the cornu ammonis (CA)1 hippocampal subfield, a region that is critically involved in the pathophysiology of the illness. We thus performed a quantitative immunoblot experiment to examine PSD95 and several of its associated proteins in the CA1 region, using post mortem brain samples derived from schizophrenia subjects with age-, sex-, and post mortem interval-matched controls (n=20/group). Our results indicate a substantial reduction in PSD95 protein expression (−61.8%). Further analysis showed additional alterations to the scaffold protein Homer1 (Homer1a: +42.9%, Homer1b/c: −24.6%), with a twofold reduction in the ratio of Homer1b/c:Homer1a isoforms (P=0.011). Metabotropic glutamate receptor 1 (mGluR1) protein levels were significantly reduced (−32.7%), and Preso, a protein that supports interactions between Homer1 or PSD95 with mGluR1, was elevated (+83.3%). Significant reduction in synaptophysin (−27.8%) was also detected, which is a validated marker of synaptic density. These findings support the presence of extensive molecular abnormalities to PSD95 and several of its associated proteins in the CA1 region in schizophrenia, offering a small but significant step toward understanding how proteins in the PSD are altered in the schizophrenia brain, and their relevance to overall hippocampal and cognitive dysfunction in the illness.

Collaboration


Dive into the Nils C. Gassen'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