Network


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

Hotspot


Dive into the research topics where Ulrike Träger is active.

Publication


Featured researches published by Ulrike Träger.


Brain | 2014

HTT-lowering reverses Huntington’s disease immune dysfunction caused by NFκB pathway dysregulation

Ulrike Träger; Ralph Andre; Nayana Lahiri; Anna Magnusson-Lind; Andreas Weiss; Stephan Grueninger; Chris McKinnon; Eva Sirinathsinghji; Shira Kahlon; Edith L. Pfister; Roger Moser; Holger Hummerich; Michael Antoniou; Gillian P. Bates; Ruth Luthi-Carter; Mark W. Lowdell; Maria Björkqvist; Gary R. Ostroff; Neil Aronin; Sarah J. Tabrizi

Huntingtons disease is an inherited neurodegenerative disorder caused by a CAG repeat expansion in the huntingtin gene. The peripheral innate immune system contributes to Huntingtons disease pathogenesis and has been targeted successfully to modulate disease progression, but mechanistic understanding relating this to mutant huntingtin expression in immune cells has been lacking. Here we demonstrate that human Huntingtons disease myeloid cells produce excessive inflammatory cytokines as a result of the cell-intrinsic effects of mutant huntingtin expression. A direct effect of mutant huntingtin on the NFκB pathway, whereby it interacts with IKKγ, leads to increased degradation of IκB and subsequent nuclear translocation of RelA. Transcriptional alterations in intracellular immune signalling pathways are also observed. Using a novel method of small interfering RNA delivery to lower huntingtin expression, we show reversal of disease-associated alterations in cellular function-the first time this has been demonstrated in primary human cells. Glucan-encapsulated small interfering RNA particles were used to lower huntingtin levels in human Huntingtons disease monocytes/macrophages, resulting in a reversal of huntingtin-induced elevated cytokine production and transcriptional changes. These findings improve our understanding of the role of innate immunity in neurodegeneration, introduce glucan-encapsulated small interfering RNA particles as tool for studying cellular pathogenesis ex vivo in human cells and raise the prospect of immune cell-directed HTT-lowering as a therapeutic in Huntingtons disease.


Journal of Clinical Investigation | 2012

Mutant huntingtin fragmentation in immune cells tracks Huntington’s disease progression

Andreas Weiss; Ulrike Träger; Edward J. Wild; Stephan Grueninger; Ruth Farmer; Christian Landles; Rachael I. Scahill; Nayana Lahiri; Salman Haider; Douglas Macdonald; Chris Frost; Gillian P. Bates; Graeme Bilbe; Rainer Kuhn; Ralph Andre; Sarah J. Tabrizi

Huntingtons disease (HD) is a fatal, inherited neurodegenerative disorder caused by an expanded CAG repeat in the gene encoding huntingtin (HTT). Therapeutic approaches to lower mutant HTT (mHTT) levels are expected to proceed to human trials, but noninvasive quantification of mHTT is not currently possible. The importance of the peripheral immune system in neurodegenerative disease is becoming increasingly recognized. Peripheral immune cells have been implicated in HD pathogenesis, but HTT levels in these cells have not been quantified before. A recently described time-resolved Förster resonance energy transfer (TR-FRET) immunoassay was used to quantify mutant and total HTT protein levels in leukocytes from patients with HD. Mean mHTT levels in monocytes, T cells, and B cells differed significantly between patients with HD and controls and between pre-manifest mutation carriers and those with clinical onset. Monocyte and T cell mHTT levels were significantly associated with disease burden scores and caudate atrophy rates in patients with HD. mHTT N-terminal fragments detected in HD PBMCs may explain the progressive increase in mHTT levels in these cells. These findings indicate that quantification of mHTT in peripheral immune cells by TR-FRET holds significant promise as a noninvasive disease biomarker.


Journal of Clinical Investigation | 2012

Mutant huntingtin impairs immune cell migration in Huntington disease

Wanda Kwan; Ulrike Träger; Dimitrios Davalos; Jill Bouchard; Ralph Andre; Aaron Miller; Andreas Weiss; Flaviano Giorgini; Christine S. Cheah; Thomas Möller; Nephi Stella; Katerina Akassoglou; Sarah J. Tabrizi; Paul J. Muchowski

In Huntington disease (HD), immune cells are activated before symptoms arise; however, it is unclear how the expression of mutant huntingtin (htt) compromises the normal functions of immune cells. Here we report that primary microglia from early postnatal HD mice were profoundly impaired in their migration to chemotactic stimuli, and expression of a mutant htt fragment in microglial cell lines was sufficient to reproduce these deficits. Microglia expressing mutant htt had a retarded response to a laser-induced brain injury in vivo. Leukocyte recruitment was defective upon induction of peritonitis in HD mice at early disease stages and was normalized upon genetic deletion of mutant htt in immune cells. Migration was also strongly impaired in peripheral immune cells from pre-manifest human HD patients. Defective actin remodeling in immune cells expressing mutant htt likely contributed to their migration deficit. Our results suggest that these functional changes may contribute to immune dysfunction and neurodegeneration in HD, and may have implications for other polyglutamine expansion diseases in which mutant proteins are ubiquitously expressed.


Neurobiology of Disease | 2015

Characterisation of immune cell function in fragment and full-length Huntington's disease mouse models.

Ulrike Träger; Ralph Andre; Anna Magnusson-Lind; James R. Miller; Colum Connolly; Andreas Weiss; Stephan Grueninger; Edina Silajdžić; Donna L. Smith; Blair R. Leavitt; Gillian P. Bates; Maria Björkqvist; Sarah J. Tabrizi

Inflammation is a growing area of research in neurodegeneration. In Huntingtons disease (HD), a fatal inherited neurodegenerative disease caused by a CAG-repeat expansion in the gene encoding huntingtin, patients have increased plasma levels of inflammatory cytokines and circulating monocytes that are hyper-responsive to immune stimuli. Several mouse models of HD also show elevated plasma levels of inflammatory cytokines. To further determine the degree to which these models recapitulate observations in HD patients, we evaluated various myeloid cell populations from different HD mouse models to determine whether they are similarly hyper-responsive, as well as measuring other aspects of myeloid cell function. Myeloid cells from each of the three mouse models studied, R6/2, HdhQ150 knock-in and YAC128, showed increased cytokine production when stimulated. However, bone marrow CD11b+ cells did not show the same hyper-responsive phenotype as spleen and blood cells. Furthermore, macrophages isolated from R6/2 mice show increased levels of phagocytosis, similar to findings in HD patients. Taken together, these results show significant promise for these mouse models to be used to study targeting innate immune pathways identified in human cells, thereby helping to understand the role the peripheral immune system plays in HD progression.


Journal of Molecular Medicine | 2013

Peripheral inflammation in neurodegeneration

Ulrike Träger; Sarah J. Tabrizi

Neuroinflammation is now a well-characterised feature of neurodegenerative diseases. Immune dysfunction outside the central nervous system is also increasingly recognised as part of the diseases. Peripheral inflammation has emerged as a modulator of disease progression and neuropathology in several neurodegenerative diseases, making it targetable in new therapeutic approaches. In addition, the easy accessibility of blood immune cells and markers makes them ideal candidates for use as possible biomarkers and a potential model of central immune cells.


Journal of Neurochemistry | 2016

Laquinimod dampens hyperactive cytokine production in Huntington's disease patient myeloid cells

Lucianne Dobson; Ulrike Träger; Ruth Farmer; Liat Hayardeny; Pippa Loupe; Michael R. Hayden; Sarah J. Tabrizi

Huntingtons disease (HD) is a neurodegenerative condition characterized by pathology in the brain and peripheral tissues. Hyperactivity of the innate immune system, due in part to NFκB pathway dysregulation, is an early and active component of HD. Evidence suggests targeting immune disruption may slow disease progression. Laquinimod is an orally active immunomodulator that down‐regulates proinflammatory cytokine production in peripheral blood mononuclear cells, and in the brain down‐regulates astrocytic and microglial activation by modulating NFκB signalling. Laquinimod had beneficial effects on inflammation, brain atrophy and disease progression in multiple sclerosis (MS) in two phase III clinical trials. This study investigated the effects of laquinimod on hyperactive proinflammatory cytokine release and NFκB signalling in HD patient myeloid cell cultures. Monocytes from manifest (manHD) and pre‐manifest (preHD) HD gene carriers and healthy volunteers (HV) were treated with laquinimod and stimulated with lipopolysaccharide. After 24 h pre‐treatment with 5 μM laquinimod, manHD monocytes released lower levels of IL‐1β, IL‐5, IL‐8, IL‐10, IL‐13 and TNFα in response to stimulation. PreHD monocytes released lower levels of IL‐8, IL‐10 and IL‐13, with no reduction observed in HV monocytes. The effects of laquinimod on dysfunctional NFκB signalling in HD was assessed by inhibitor of kappa B (IκB) degradation kinetics, nuclear translocation of NFκB and interactions between IκB kinase (IKK) and HTT, in HD myeloid cells. No differences were observed between laquinimod‐treated and untreated conditions. These results provide evidence that laquinimod dampens hyper‐reactive cytokine release from manHD and preHD monocytes, with a much reduced effect on HV monocytes.


Human Molecular Genetics | 2016

RNA-Seq of Huntington’s disease patient myeloid cells reveals innate transcriptional dysregulation associated with proinflammatory pathway activation

James R. Miller; Kitty Lo; Ralph Andre; Davina J. Hensman Moss; Ulrike Träger; Timothy Stone; Lesley Jones; Peter Holmans; Vincent Plagnol; Sarah J. Tabrizi

Innate immune activation beyond the central nervous system is emerging as a vital component of the pathogenesis of neurodegeneration. Huntington’s disease (HD) is a fatal neurodegenerative disorder caused by a CAG repeat expansion in the huntingtin gene. The systemic innate immune system is thought to act as a modifier of disease progression; however, the molecular mechanisms remain only partially understood. Here we use RNA-sequencing to perform whole transcriptome analysis of primary monocytes from thirty manifest HD patients and thirty-three control subjects, cultured with and without a proinflammatory stimulus. In contrast with previous studies that have required stimulation to elicit phenotypic abnormalities, we demonstrate significant transcriptional differences in HD monocytes in their basal, unstimulated state. This includes previously undetected increased resting expression of genes encoding numerous proinflammatory cytokines, such as IL6. Further pathway analysis revealed widespread resting enrichment of proinflammatory functional gene sets, while upstream regulator analysis coupled with Western blotting suggests that abnormal basal activation of the NFĸB pathway plays a key role in mediating these transcriptional changes. That HD myeloid cells have a proinflammatory phenotype in the absence of stimulation is consistent with a priming effect of mutant huntingtin, whereby basal dysfunction leads to an exaggerated inflammatory response once a stimulus is encountered. These data advance our understanding of mutant huntingtin pathogenesis, establish resting myeloid cells as a key source of HD immune dysfunction, and further demonstrate the importance of systemic immunity in the potential treatment of HD and the wider study of neurodegeneration.


PLOS Currents | 2013

JAK/STAT Signalling in Huntington's Disease Immune Cells.

Ulrike Träger; Anna Magnusson-Lind; Nayana Lahiri Swales; Edward J. Wild; Janet North; Mark W. Lowdell; Maria Björkqvist

Huntington’s disease (HD) is an inherited neurodegenerative disorder caused by a CAG repeat expansion in the huntingtin (HTT) gene. Both central and peripheral innate immune activation have been described as features of the disease. Isolated human HD monocytes have been shown to produce more cytokines upon LPS stimulation compared to control monocytes. Understanding alterations in the signalling cascades responsible and activated by this increase in pro-inflammatory cytokine production is crucial in understanding the molecular basis of this phenomenon. Here we investigated the signalling cascade most commonly activated by pro-inflammatory cytokines such as IL-6 – the JAK/STAT signalling cascade. Using flow cytometry, we show that one out of three key transcription factors activated by JAK/STAT signalling is altered in primary human HD innate immune cells, suggesting that this pathway may only play a minor, additive role in the immune cell dysfunction in HD.


Proceedings of the National Academy of Sciences of the United States of America | 2017

KEAP1-modifying small molecule reveals muted NRF2 signaling responses in neural stem cells from Huntington's disease patients

Luisa Quinti; Sharadha Dayalan Naidu; Ulrike Träger; Xiqun Chen; Kimberly B. Kegel-Gleason; David Llères; Colum Connolly; Vanita Chopra; Cho Low; Sébastien Moniot; Ellen Sapp; Adelaide Tousley; Petr Vodicka; Michael J. Van Kanegan; Linda S. Kaltenbach; Lisa A. Crawford; Matthew Fuszard; Maureen Higgins; James R. Miller; Ruth Farmer; Vijay Potluri; Susanta Samajdar; Lisa Meisel; Ningzhe Zhang; Andrew Snyder; Ross L. Stein; Steven M. Hersch; Eranthie Weerapana; Michael A. Schwarzschild; Clemens Steegborn

Significance Chronic neuroinflammation and oxidative stress are likely complicit in driving disease progression in Huntingtons disease (HD). Here, we describe the mechanism of action of a unique chemical scaffold that is highly selective for activation of NRF2, the master transcriptional regulator of cellular antiinflammatory and antioxidant defense genes. The use of this scaffold revealed that NRF2 activation responses were muted in HD patient-derived neural stem cells, suggesting increased susceptibility of this critical renewable cell population to oxidative stress in HD brain. However, pharmacological activation of NRF2 was able to repress inflammatory responses in mouse microglia and astrocytes, the principal cellular mediators of neuroinflammation, and in blood monocytes from HD patients. Our results suggest multiple protective benefits of NRF2 activation for HD patients. The activity of the transcription factor nuclear factor-erythroid 2 p45-derived factor 2 (NRF2) is orchestrated and amplified through enhanced transcription of antioxidant and antiinflammatory target genes. The present study has characterized a triazole-containing inducer of NRF2 and elucidated the mechanism by which this molecule activates NRF2 signaling. In a highly selective manner, the compound covalently modifies a critical stress-sensor cysteine (C151) of the E3 ligase substrate adaptor protein Kelch-like ECH-associated protein 1 (KEAP1), the primary negative regulator of NRF2. We further used this inducer to probe the functional consequences of selective activation of NRF2 signaling in Huntingtons disease (HD) mouse and human model systems. Surprisingly, we discovered a muted NRF2 activation response in human HD neural stem cells, which was restored by genetic correction of the disease-causing mutation. In contrast, selective activation of NRF2 signaling potently repressed the release of the proinflammatory cytokine IL-6 in primary mouse HD and WT microglia and astrocytes. Moreover, in primary monocytes from HD patients and healthy subjects, NRF2 induction repressed expression of the proinflammatory cytokines IL-1, IL-6, IL-8, and TNFα. Together, our results demonstrate a multifaceted protective potential of NRF2 signaling in key cell types relevant to HD pathology.


Scientific Reports | 2017

Allele-Selective Suppression of Mutant Huntingtin in Primary Human Blood Cells

James R. Miller; Edith L. Pfister; Wanzhao Liu; Ralph Andre; Ulrike Träger; Lori A. Kennington; Kimberly Lo; Sipke Dijkstra; Douglas A. MacDonald; Gary R. Ostroff; Neil Aronin; Sarah J. Tabrizi

Post-transcriptional gene silencing is a promising therapy for the monogenic, autosomal dominant, Huntington’s disease (HD). However, wild-type huntingtin (HTT) has important cellular functions, so the ideal strategy would selectively lower mutant HTT while sparing wild-type. HD patients were genotyped for heterozygosity at three SNP sites, before phasing each SNP allele to wild-type or mutant HTT. Primary ex vivo myeloid cells were isolated from heterozygous patients and transfected with SNP-targeted siRNA, using glucan particles taken up by phagocytosis. Highly selective mRNA knockdown was achieved when targeting each allele of rs362331 in exon 50 of the HTT transcript; this selectivity was also present on protein studies. However, similar selectivity was not observed when targeting rs362273 or rs362307. Furthermore, HD myeloid cells are hyper-reactive compared to control. Allele-selective suppression of either wild-type or mutant HTT produced a significant, equivalent reduction in the cytokine response of HD myeloid cells to LPS, suggesting that wild-type HTT has a novel immune function. We demonstrate a sequential therapeutic process comprising genotyping and mutant HTT-linkage of SNPs, followed by personalised allele-selective suppression in a small patient cohort. We further show that allele-selectivity in ex vivo patient cells is highly SNP-dependent, with implications for clinical trial target selection.

Collaboration


Dive into the Ulrike Träger's collaboration.

Top Co-Authors

Avatar

Sarah J. Tabrizi

UCL Institute of Neurology

View shared research outputs
Top Co-Authors

Avatar

Ralph Andre

UCL Institute of Neurology

View shared research outputs
Top Co-Authors

Avatar

James R. Miller

UCL Institute of Neurology

View shared research outputs
Top Co-Authors

Avatar

Gillian P. Bates

UCL Institute of Neurology

View shared research outputs
Top Co-Authors

Avatar

Nayana Lahiri

UCL Institute of Neurology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Edward J. Wild

University College London

View shared research outputs
Researchain Logo
Decentralizing Knowledge