Network


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

Hotspot


Dive into the research topics where Katharina Domschke is active.

Publication


Featured researches published by Katharina Domschke.


Molecular Psychiatry | 2011

TMEM132D, a new candidate for anxiety phenotypes: evidence from human and mouse studies

Ludwig Czibere; D. Roeske; Susanne Lucae; P. G. Unschuld; Stephan Ripke; Michael Specht; Martin A. Kohli; Stefan Kloiber; Marcus Ising; Angela Heck; Hildegard Pfister; P. Zimmermann; Roselind Lieb; Benno Pütz; Manfred Uhr; Peter Weber; Jan M. Deussing; Mariya Gonik; Mirjam Bunck; Melanie S. Kessler; Elisabeth Frank; Christa Hohoff; Katharina Domschke; Petra Krakowitzky; W. Maier; Borwin Bandelow; Christian Jacob; J. Deckert; Stefan Schreiber; Jana Strohmaier

The lifetime prevalence of panic disorder (PD) is up to 4% worldwide and there is substantial evidence that genetic factors contribute to the development of PD. Single-nucleotide polymorphisms (SNPs) in TMEM132D, identified in a whole-genome association study (GWAS), were found to be associated with PD in three independent samples, with a two-SNP haplotype associated in each of three samples in the same direction, and with a P-value of 1.2e−7 in the combined sample (909 cases and 915 controls). Independent SNPs in this gene were also associated with the severity of anxiety symptoms in patients affected by PD or panic attacks as well as in patients suffering from unipolar depression. Risk genotypes for PD were associated with higher TMEM132D mRNA expression levels in the frontal cortex. In parallel, using a mouse model of extremes in trait anxiety, we could further show that anxiety-related behavior was positively correlated with Tmem132d mRNA expression in the anterior cingulate cortex, central to the processing of anxiety/fear-related stimuli, and that in this animal model a Tmem132d SNP is associated with anxiety-related behavior in an F2 panel. TMEM132D may thus be an important new candidate gene for PD as well as more generally for anxiety-related behavior.


Experimental Brain Research | 2005

The methionine allele of the COMT polymorphism impairs prefrontal cognition in children and adolescents with ADHD.

Mark A. Bellgrove; Katharina Domschke; Ziarih Hawi; Aiveen Kirley; Celine Mullins; Ian H. Robertson; Michael Gill

ADHD is a highly heritable psychiatric disorder of childhood. A functional polymorphism (Val158Met) of the catechol-O-methyltransferase (COMT) gene has attracted interest as a candidate gene for ADHD. The high-activity valine variant of this polymorphism degrades prefrontal dopamine three to four times more quickly than the low-activity methionine variant and could therefore contribute to the proposed hypodopaminergic state in ADHD. Here we tested for association of this polymorphism with ADHD and examined its influence on prefrontal cognition in ADHD. We have previously reported no association of the Val158Met COMT gene polymorphism in 94 Irish ADHD families (Hawi et al. (2000) Am J Med Genet 96:282–284). Here we re-examined this finding with an extended sample of 179 ADHD cases using a family control design. We also examined the performance of children and adolescents with ADHD (n=61) on a standardised test of sustained attention. Analysis confirmed the absence of an association between the Val158Met COMT gene polymorphism and the clinical phenotype of ADHD. COMT genotype, however, affected prefrontal cognition in ADHD: ADHD children who were homozygous for the valine variant had significantly better sustained attention than those ADHD children possessing at least one copy of the methionine variant. Children possessing the methionine variant performed significantly below age-related norms on tests of sustained attention. Contrary to expectations, the methionine variant of the Val158Met COMT gene polymorphism impaired prefrontally-mediated cognition in ADHD. This effect may be understood by positing a hyper-functioning of prefrontal dopaminergic systems. Against this background, the slower clearance of dopamine associated with the methionine variant of the COMT gene polymorphism may be disadvantageous to cognition in ADHD.


American Journal of Medical Genetics | 2005

Association analysis of the monoamine oxidase A and B genes with attention deficit hyperactivity disorder (ADHD) in an Irish sample : Preferential transmission of the MAO-A 941G allele to affected children

Katharina Domschke; Karen Sheehan; Naomi Lowe; Aiveen Kirley; Celine Mullins; Roderick O'Sullivan; Christine M. Freitag; Tim Becker; Judith Conroy; Michael Fitzgerald; Michael Gill; Ziarih Hawi

Pharmacological and genetic studies suggest the importance of the dopaminergic, serotonergic, and noradrenergic systems in the pathogenesis of attention deficit hyperactivity disorder (ADHD). Monoamine oxidases A and B (MAO‐A and MAO‐B) degrade biogenic amines such as dopamine and serotonin and thereby control the levels of these neurotransmitters in the central nervous system. We examined four polymorphisms in the MAO‐A gene (30 bp promoter VNTR, CA microsatellite in intron 2, 941G/T SNP in exon 8, and A/G SNP in intron 12) as well as two markers in the MAO‐B gene (CA microsatellite in intron 2 and T/C SNP in intron 13) for association with ADHD in an Irish sample of 179 nuclear families. TDT analysis of the examined MAO‐A markers revealed a significant association of the more active MAO‐A 941G allele with the disorder (χ2 = 5.1, P = 0.03, OR = 1.7). In addition, haplotype analysis revealed a significantly increased transmission of a haplotype consisting of the shorter allele of the promoter VNTR (allele 1), the 6‐repeat allele of the CA microsatellite and the G‐allele of the 941G/T SNP (famhap global statistic 34.54, P = 0.01) to ADHD cases. No significant distortion in the number of transmitted alleles was observed between the two examined MAO‐B polymorphisms and ADHD. These findings suggest the importance of the 941G/T MAO‐A polymorphism in the development of ADHD at least in the Irish population.


Molecular Psychiatry | 2014

MAOA and mechanisms of panic disorder revisited: from bench to molecular psychotherapy

Andreas Reif; Jan Richter; Benjamin Straube; Michael Höfler; Ulrike Lueken; Andrew T. Gloster; Heike Weber; Katharina Domschke; Lydia Fehm; A. Ströhle; Andreas Jansen; Alexander L. Gerlach; Martin Pyka; Isabelle Reinhardt; Christoph Konrad; André Wittmann; Bettina Pfleiderer; Georg W. Alpers; Paul Pauli; Thomas Lang; Volker Arolt; Hans-Ulrich Wittchen; Alfons O. Hamm; Tilo Kircher; Jürgen Deckert

Panic disorder with agoraphobia (PD/AG) is a prevalent mental disorder featuring a substantial complex genetic component. At present, only a few established risk genes exist. Among these, the gene encoding monoamine oxidase A (MAOA) is noteworthy given that genetic variation has been demonstrated to influence gene expression and monoamine levels. Long alleles of the MAOA-uVNTR promoter polymorphism are associated with PD/AG and correspond with increased enzyme activity. Here, we have thus investigated the impact of MAOA-uVNTR on therapy response, behavioral avoidance and brain activity in fear conditioning in a large controlled and randomized multicenter study on cognitive behavioral therapy (CBT) in PD/AG. The study consisted of 369 PD/AG patients, and genetic information was available for 283 patients. Carriers of the risk allele had significantly worse outcome as measured by the Hamilton Anxiety scale (46% responders vs 67%, P=0.017). This was accompanied by elevated heart rate and increased fear during an anxiety-provoking situation, that is, the behavioral avoidance task. All but one panic attack that happened during this task occurred in risk allele carriers and, furthermore, risk allele carriers did not habituate to the situation during repetitive exposure. Finally, functional neuroimaging during a classical fear conditioning paradigm evidenced that the protective allele is associated with increased activation of the anterior cingulate cortex upon presentation of the CS+ during acquisition of fear. Further differentiation between high- and low-risk subjects after treatment was observed in the inferior parietal lobes, suggesting differential brain activation patterns upon CBT. Taken together, we established that a genetic risk factor for PD/AG is associated with worse response to CBT and identify potential underlying neural mechanisms. These findings might govern how psychotherapy can include genetic information to tailor individualized treatment approaches.


Translational Psychiatry | 2016

MAOA gene hypomethylation in panic disorder-reversibility of an epigenetic risk pattern by psychotherapy.

C. Ziegler; Jan Richter; M Mahr; A Gajewska; Miriam A. Schiele; A Gehrmann; Brigitte Schmidt; Klaus-Peter Lesch; Thomas Lang; Sylvia Helbig-Lang; Paul Pauli; Tilo Kircher; Andreas Reif; Winfried Rief; Anna N. Vossbeck-Elsebusch; Volker Arolt; Hans-Ulrich Wittchen; Alfons O. Hamm; J. Deckert; Katharina Domschke

Epigenetic signatures such as methylation of the monoamine oxidase A (MAOA) gene have been found to be altered in panic disorder (PD). Hypothesizing temporal plasticity of epigenetic processes as a mechanism of successful fear extinction, the present psychotherapy-epigenetic study for we believe the first time investigated MAOA methylation changes during the course of exposure-based cognitive behavioral therapy (CBT) in PD. MAOA methylation was compared between N=28 female Caucasian PD patients (discovery sample) and N=28 age- and sex-matched healthy controls via direct sequencing of sodium bisulfite-treated DNA extracted from blood cells. MAOA methylation was furthermore analyzed at baseline (T0) and after a 6-week CBT (T1) in the discovery sample parallelized by a waiting time in healthy controls, as well as in an independent sample of female PD patients (N=20). Patients exhibited lower MAOA methylation than healthy controls (P<0.001), and baseline PD severity correlated negatively with MAOA methylation (P=0.01). In the discovery sample, MAOA methylation increased up to the level of healthy controls along with CBT response (number of panic attacks; T0–T1: +3.37±2.17%), while non-responders further decreased in methylation (−2.00±1.28%; P=0.001). In the replication sample, increases in MAOA methylation correlated with agoraphobic symptom reduction after CBT (P=0.02–0.03). The present results support previous evidence for MAOA hypomethylation as a PD risk marker and suggest reversibility of MAOA hypomethylation as a potential epigenetic correlate of response to CBT. The emerging notion of epigenetic signatures as a mechanism of action of psychotherapeutic interventions may promote epigenetic patterns as biomarkers of lasting extinction effects.


Pharmacopsychiatry | 2017

Consensus Guidelines for Therapeutic Drug Monitoring in Neuropsychopharmacology: Update 2017

Christoph Hiemke; N. Bergemann; Hans Willi Clement; Andreas Conca; J. Deckert; Katharina Domschke; Gabriel Eckermann; Karin Egberts; Manfred Gerlach; C Greiner; Gerhard Gründer; Ekkehard Haen; Ursula Havemann-Reinecke; Gudrun Hefner; R. Helmer; Ger Janssen; E. Jaquenoud; Gerd Laux; Thomas Messer; Rainald Mössner; Matthias J. Müller; Michael Paulzen; Bruno Pfuhlmann; Peter Riederer; Alois Saria; B. Schoppek; Georgios Schoretsanitis; Markus J. Schwarz; M. Silva Gracia; Benedikt Stegmann

Authors C. Hiemke1, 2, N. Bergemann3, H. W. Clement4, A. Conca5, J. Deckert6, K. Domschke7, G. Eckermann8, K. Egberts9, M. Gerlach9, C. Greiner10, G. Gründer11, E. Haen12, U. Havemann-Reinecke13, G. Hefner14, R. Helmer15, G. Janssen16, E. Jaquenoud17, G. Laux18, T. Messer19, R. Mössner20, M. J. Müller21, M. Paulzen11, B. Pfuhlmann22, P. Riederer6, A. Saria23, B. Schoppek24, G. Schoretsanitis25, M. Schwarz26, M. Silva Gracia12, B. Stegmann12, W. Steimer27, J. C. Stingl10, M. Uhr28, S. Ulrich29, S. Unterecker6, R. Waschgler30, G. Zernig23, 31, G. Zurek32, P. Baumann33


Molecular Psychiatry | 2015

Multimodal imaging of a tescalcin (TESC)-regulating polymorphism (rs7294919)-specific effects on hippocampal gray matter structure

Udo Dannlowski; Hans J. Grabe; Katharina Wittfeld; J Klaus; Carsten Konrad; Dominik Grotegerd; Ronny Redlich; Thomas Suslow; Nils Opel; Patricia Ohrmann; Jürgen M. Bauer; Peter Zwanzger; I. Laeger; Christa Hohoff; Volker Arolt; Walter Heindel; M. Deppe; Katharina Domschke; Katrin Hegenscheid; Henry Völzke; David Stacey; H. E. Meyer zu Schwabedissen; Harald Kugel; Bernhard T. Baune

In two large genome-wide association studies, an intergenic single-nucleotide polymorphism (SNP; rs7294919) involved in TESC gene regulation has been associated with hippocampus volume. Further characterization of neurobiological effects of the TESC gene is warranted using multimodal brain-wide structural and functional imaging. Voxel-based morphometry (VBM8) was used in two large, well-characterized samples of healthy individuals of West-European ancestry (Münster sample, N=503; SHIP-TREND, N=721) to analyze associations between rs7294919 and local gray matter volume. In subsamples, white matter fiber structure was investigated using diffusion tensor imaging (DTI) and limbic responsiveness was measured by means of functional magnetic resonance imaging (fMRI) during facial emotion processing (N=220 and N=264, respectively). Furthermore, gene x environment (G × E) interaction and gene x gene interaction with SNPs from genes previously found to be associated with hippocampal size (FKBP5, Reelin, IL-6, TNF-α, BDNF and 5-HTTLPR/rs25531) were explored. We demonstrated highly significant effects of rs7294919 on hippocampal gray matter volumes in both samples. In whole-brain analyses, no other brain areas except the hippocampal formation and adjacent temporal structures were associated with rs7294919. There were no genotype effects on DTI and fMRI results, including functional connectivity measures. No G × E interaction with childhood maltreatment was found in both samples. However, an interaction between rs7294919 and rs2299403 in the Reelin gene was found that withstood correction for multiple comparisons. We conclude that rs7294919 exerts highly robust and regionally specific effects on hippocampal gray matter structures, but not on other neuropsychiatrically relevant imaging markers. The biological interaction between TESC and RELN pointing to a neurodevelopmental origin of the observed findings warrants further mechanistic investigations.


Molecular Psychiatry | 2016

Candidate genes in panic disorder: meta-analyses of 23 common variants in major anxiogenic pathways.

Aaron S. Howe; Henriette N. Buttenschøn; Ali Bani-Fatemi; Eduard Maron; Takeshi Otowa; Elisabeth B. Binder; Noomi Gregersen; Ole Mors; David P. D. Woldbye; Katharina Domschke; Andreas Reif; J Shlik; S Kõks; Yukiko Kawamura; Akinori Miyashita; Ryozo Kuwano; Katsushi Tokunaga; Hisashi Tanii; Jordan W. Smoller; Tsukasa Sasaki; Diana Koszycki; V. De Luca

The utilization of molecular genetics approaches in examination of panic disorder (PD) has implicated several variants as potential susceptibility factors for panicogenesis. However, the identification of robust PD susceptibility genes has been complicated by phenotypic diversity, underpowered association studies and ancestry-specific effects. In the present study, we performed a succinct review of case–control association studies published prior to April 2015. Meta-analyses were performed for candidate gene variants examined in at least three studies using the Cochrane Mantel–Haenszel fixed-effect model. Secondary analyses were also performed to assess the influences of sex, agoraphobia co-morbidity and ancestry-specific effects on panicogenesis. Meta-analyses were performed on 23 variants in 20 PD candidate genes. Significant associations after correction for multiple testing were observed for three variants, TMEM132D rs7370927 (T allele: odds ratio (OR)=1.27, 95% confidence interval (CI): 1.15–1.40, P=2.49 × 10−6), rs11060369 (CC genotype: OR=0.65, 95% CI: 0.53–0.79, P=1.81 × 10−5) and COMT rs4680 (Val (G) allele: OR=1.27, 95% CI: 1.14–1.42, P=2.49 × 10−5) in studies with samples of European ancestry. Nominal associations that did not survive correction for multiple testing were observed for NPSR1 rs324891 (T allele: OR=1.22, 95% CI: 1.07–1.38, P=0.002), TPH1 rs1800532 (AA genotype: OR=1.46, 95% CI: 1.14–1.89, P=0.003) and HTR2A rs6313 (T allele: OR=1.19, 95% CI: 1.07–1.33, P=0.002) in studies with samples of European ancestry and for MAOA-uVNTR in female PD (low-active alleles: OR=1.21, 95% CI: 1.07–1.38, P=0.004). No significant associations were observed in the secondary analyses considering sex, agoraphobia co-morbidity and studies with samples of Asian ancestry. Although these findings highlight a few associations, PD likely involves genetic variation in a multitude of biological pathways that is diverse among populations. Future studies must incorporate larger sample sizes and genome-wide approaches to further quantify the observed genetic variation among populations and subphenotypes of PD.


Translational Psychiatry | 2014

Prenatal stress-induced programming of genome-wide promoter DNA methylation in 5-HTT-deficient mice

Karla-Gerlinde Schraut; Sissi Jakob; M.T. Weidner; Angelika Schmitt; Claus-Jürgen Scholz; Tatyana Strekalova; N El Hajj; Lars Eijssen; Katharina Domschke; Andreas Reif; T. Haaf; Gabriela Ortega; Harry W.M. Steinbusch; Klaus-Peter Lesch; D.L.A. van den Hove

The serotonin transporter gene (5-HTT/SLC6A4)-linked polymorphic region has been suggested to have a modulatory role in mediating effects of early-life stress exposure on psychopathology rendering carriers of the low-expression short (s)-variant more vulnerable to environmental adversity in later life. The underlying molecular mechanisms of this gene-by-environment interaction are not well understood, but epigenetic regulation including differential DNA methylation has been postulated to have a critical role. Recently, we used a maternal restraint stress paradigm of prenatal stress (PS) in 5-HTT-deficient mice and showed that the effects on behavior and gene expression were particularly marked in the hippocampus of female 5-Htt+/− offspring. Here, we examined to which extent these effects are mediated by differential methylation of DNA. For this purpose, we performed a genome-wide hippocampal DNA methylation screening using methylated-DNA immunoprecipitation (MeDIP) on Affymetrix GeneChip Mouse Promoter 1.0 R arrays. Using hippocampal DNA from the same mice as assessed before enabled us to correlate gene-specific DNA methylation, mRNA expression and behavior. We found that 5-Htt genotype, PS and their interaction differentially affected the DNA methylation signature of numerous genes, a subset of which showed overlap with the expression profiles of the corresponding transcripts. For example, a differentially methylated region in the gene encoding myelin basic protein (Mbp) was associated with its expression in a 5-Htt-, PS- and 5-Htt × PS-dependent manner. Subsequent fine-mapping of this Mbp locus linked the methylation status of two specific CpG sites to Mbp expression and anxiety-related behavior. In conclusion, hippocampal DNA methylation patterns and expression profiles of female prenatally stressed 5-Htt+/− mice suggest that distinct molecular mechanisms, some of which are promoter methylation-dependent, contribute to the behavioral effects of the 5-Htt genotype, PS exposure and their interaction.


Molecular Psychiatry | 2017

GLRB allelic variation associated with agoraphobic cognitions, increased startle response and fear network activation : a potential neurogenetic pathway to panic disorder

Jürgen Deckert; Heike Weber; C Villmann; Tina B. Lonsdorf; Jan Richter; Marta Andreatta; Alejandro Arias-Vasquez; L Hommers; Lindsey Kent; C. Schartner; Sven Cichon; C Wolf; N Schaefer; C R von Collenberg; B Wachter; Robert Blum; Dirk Schümann; R Scharfenort; J Schumacher; Andreas J. Forstner; Christian Baumann; M A Schiele; Swantje Notzon; Peter Zwanzger; Joost Janzing; Tessel E. Galesloot; Lambertus A. Kiemeney; Agnieszka Gajewska; Evelyn Glotzbach-Schoon; Andreas Mühlberger

The molecular genetics of panic disorder (PD) with and without agoraphobia (AG) are still largely unknown and progress is hampered by small sample sizes. We therefore performed a genome-wide association study with a dimensional, PD/AG-related anxiety phenotype based on the Agoraphobia Cognition Questionnaire (ACQ) in a sample of 1370 healthy German volunteers of the CRC TRR58 MEGA study wave 1. A genome-wide significant association was found between ACQ and single non-coding nucleotide variants of the GLRB gene (rs78726293, P=3.3 × 10−8; rs191260602, P=3.9 × 10−8). We followed up on this finding in a larger dimensional ACQ sample (N=2547) and in independent samples with a dichotomous AG phenotype based on the Symptoms Checklist (SCL-90; N=3845) and a case–control sample with the categorical phenotype PD/AG (Ncombined =1012) obtaining highly significant P-values also for GLRB single-nucleotide variants rs17035816 (P=3.8 × 10−4) and rs7688285 (P=7.6 × 10−5). GLRB gene expression was found to be modulated by rs7688285 in brain tissue, as well as cell culture. Analyses of intermediate PD/AG phenotypes demonstrated increased startle reflex and increased fear network, as well as general sensory activation by GLRB risk gene variants rs78726293, rs191260602, rs17035816 and rs7688285. Partial Glrb knockout mice demonstrated an agoraphobic phenotype. In conjunction with the clinical observation that rare coding GLRB gene mutations are associated with the neurological disorder hyperekplexia characterized by a generalized startle reaction and agoraphobic behavior, our data provide evidence that non-coding, although functional GLRB gene polymorphisms may predispose to PD by increasing startle response and agoraphobic cognitions.

Collaboration


Dive into the Katharina Domschke'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

Heike Weber

University of Würzburg

View shared research outputs
Top Co-Authors

Avatar

Paul Pauli

University of Würzburg

View shared research outputs
Top Co-Authors

Avatar

Jan Richter

University of Greifswald

View shared research outputs
Top Co-Authors

Avatar

Alfons O. Hamm

University of Greifswald

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge