Network


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

Hotspot


Dive into the research topics where Ueli Nachbur is active.

Publication


Featured researches published by Ueli Nachbur.


Nature | 2011

Linear ubiquitination prevents inflammation and regulates immune signalling

Björn Gerlach; Stefanie M. Cordier; Anna C. Schmukle; Christoph H. Emmerich; Eva Rieser; Tobias Haas; Andrew I. Webb; James A Rickard; Holly Anderton; W. Wei-Lynn Wong; Ueli Nachbur; Lahiru Gangoda; Uwe Warnken; Anthony W. Purcell; John Silke; Henning Walczak

Members of the tumour necrosis factor (TNF) receptor superfamily have important functions in immunity and inflammation. Recently linear ubiquitin chains assembled by a complex containing HOIL-1 and HOIP (also known as RBCK1 and RNF31, respectively) were implicated in TNF signalling, yet their relevance in vivo remained uncertain. Here we identify SHARPIN as a third component of the linear ubiquitin chain assembly complex, recruited to the CD40 and TNF receptor signalling complexes together with its other constituents, HOIL-1 and HOIP. Mass spectrometry of TNF signalling complexes revealed RIP1 (also known as RIPK1) and NEMO (also known as IKKγ or IKBKG) to be linearly ubiquitinated. Mutation of the Sharpin gene (Sharpincpdm/cpdm) causes chronic proliferative dermatitis (cpdm) characterized by inflammatory skin lesions and defective lymphoid organogenesis. Gene induction by TNF, CD40 ligand and interleukin-1β was attenuated in cpdm-derived cells which were rendered sensitive to TNF-induced death. Importantly, Tnf gene deficiency prevented skin lesions in cpdm mice. We conclude that by enabling linear ubiquitination in the TNF receptor signalling complex, SHARPIN interferes with TNF-induced cell death and, thereby, prevents inflammation. Our results provide evidence for the relevance of linear ubiquitination in vivo in preventing inflammation and regulating immune signalling.


Molecular Cell | 2012

The Ubiquitin Ligase XIAP Recruits LUBAC for NOD2 Signaling in Inflammation and Innate Immunity

Rune Busk Damgaard; Ueli Nachbur; Monica Yabal; W. Wei-Lynn Wong; Berthe Katrine Fiil; Mischa Kastirr; Eva Rieser; James A Rickard; Aleksandra Bankovacki; Christian Peschel; Juergen Ruland; Simon Bekker-Jensen; Niels Mailand; Thomas Kaufmann; Andreas Strasser; Henning Walczak; John Silke; Philipp J. Jost; Mads Gyrd-Hansen

Nucleotide-binding and oligomerization domain (NOD)-like receptors constitute a first line of defense against invading bacteria. X-linked Inhibitor of Apoptosis (XIAP) is implicated in the control of bacterial infections, and mutations in XIAP are causally linked to immunodeficiency in X-linked lymphoproliferative syndrome type-2 (XLP-2). Here, we demonstrate that the RING domain of XIAP is essential for NOD2 signaling and that XIAP contributes to exacerbation of inflammation-induced hepatitis in experimental mice. We find that XIAP ubiquitylates RIPK2 and recruits the linear ubiquitin chain assembly complex (LUBAC) to NOD2. We further show that LUBAC activity is required for efficient NF-κB activation and secretion of proinflammatory cytokines after NOD2 stimulation. Remarkably, XLP-2-derived XIAP variants have impaired ubiquitin ligase activity, fail to ubiquitylate RIPK2, and cannot facilitate NOD2 signaling. We conclude that XIAP and LUBAC constitute essential ubiquitin ligases in NOD2-mediated inflammatory signaling and propose that deregulation of NOD2 signaling contributes to XLP-2 pathogenesis.


Nature | 2013

A type III effector antagonizes death receptor signalling during bacterial gut infection

Jaclyn S. Pearson; Sze Ong; Catherine L. Kennedy; Michelle Kelly; Keith S. Robinson; Tania Lung; Ashley Mansell; Patrice Riedmaier; Claire Oates; Ali Zaid; Sabrina Mühlen; Valerie F. Crepin; Oliver Marchès; Ching-Seng Ang; Nicholas A. Williamson; Lorraine A. O'Reilly; Aleksandra Bankovacki; Ueli Nachbur; Giuseppe Infusini; Andrew I. Webb; John Silke; Andreas Strasser; Gad Frankel; Elizabeth L. Hartland

Successful infection by enteric bacterial pathogens depends on the ability of the bacteria to colonize the gut, replicate in host tissues and disseminate to other hosts. Pathogens such as Salmonella, Shigella and enteropathogenic and enterohaemorrhagic (EPEC and EHEC, respectively) Escherichia coli use a type III secretion system (T3SS) to deliver virulence effector proteins into host cells during infection that promote colonization and interfere with antimicrobial host responses. Here we report that the T3SS effector NleB1 from EPEC binds to host cell death-domain-containing proteins and thereby inhibits death receptor signalling. Protein interaction studies identified FADD, TRADD and RIPK1 as binding partners of NleB1. NleB1 expressed ectopically or injected by the bacterial T3SS prevented Fas ligand or TNF-induced formation of the canonical death-inducing signalling complex (DISC) and proteolytic activation of caspase-8, an essential step in death-receptor-induced apoptosis. This inhibition depended on the N-acetylglucosamine transferase activity of NleB1, which specifically modified Arg 117 in the death domain of FADD. The importance of the death receptor apoptotic pathway to host defence was demonstrated using mice deficient in the FAS signalling pathway, which showed delayed clearance of the EPEC-like mouse pathogen Citrobacter rodentium and reversion to virulence of an nleB mutant. The activity of NleB suggests that EPEC and other attaching and effacing pathogens antagonize death-receptor-induced apoptosis of infected cells, thereby blocking a major antimicrobial host response.


eLife | 2014

TNFR1-dependent cell death drives inflammation in Sharpin-deficient mice

James A Rickard; Holly Anderton; Nima Etemadi; Ueli Nachbur; Maurice Darding; Nieves Peltzer; Najoua Lalaoui; Kate E. Lawlor; Hannah K. Vanyai; Cathrine Hall; Aleks Bankovacki; Lahiru Gangoda; W. Wei-Lynn Wong; Jason Corbin; Chunzi Huang; Edward S. Mocarski; James M. Murphy; Warren S. Alexander; Anne K. Voss; David L. Vaux; William J. Kaiser; Henning Walczak; John Silke

SHARPIN regulates immune signaling and contributes to full transcriptional activity and prevention of cell death in response to TNF in vitro. The inactivating mouse Sharpin cpdm mutation causes TNF-dependent multi-organ inflammation, characterized by dermatitis, liver inflammation, splenomegaly, and loss of Peyers patches. TNF-dependent cell death has been proposed to cause the inflammatory phenotype and consistent with this we show Tnfr1, but not Tnfr2, deficiency suppresses the phenotype (and it does so more efficiently than Il1r1 loss). TNFR1-induced apoptosis can proceed through caspase-8 and BID, but reduction in or loss of these players generally did not suppress inflammation, although Casp8 heterozygosity significantly delayed dermatitis. Ripk3 or Mlkl deficiency partially ameliorated the multi-organ phenotype, and combined Ripk3 deletion and Casp8 heterozygosity almost completely suppressed it, even restoring Peyers patches. Unexpectedly, Sharpin, Ripk3 and Casp8 triple deficiency caused perinatal lethality. These results provide unexpected insights into the developmental importance of SHARPIN. DOI: http://dx.doi.org/10.7554/eLife.03464.001


Proceedings of the National Academy of Sciences of the United States of America | 2015

Eliminating hepatitis B by antagonizing cellular inhibitors of apoptosis

Gregor Ebert; Cody Allison; Simon Preston; James Cooney; Jesse G. Toe; Michael D. Stutz; Samar Ojaimi; Nikola Baschuk; Ueli Nachbur; Joseph Torresi; John Silke; C. Glenn Begley; Marc Pellegrini

Significance Current antiviral treatments for chronic hepatitis B virus (HBV) infection are effective in suppressing production of virus, but they have poor efficacy in promoting the elimination of infection. Hence, most patients with chronic HBV infection are maintained on antiviral therapies indefinitely. There is much interest in identifying treatments that promote the clearance of infected hepatocytes, thus purging the HBV DNA reservoir in the liver. Here, we show that the clinical-stage drug birinapant, which antagonizes host cell inhibitor of apoptosis proteins (cIAPs), preferentially promotes the killing of HBV-infected hepatocytes in a mouse model of HBV. Therefore, birinapant and other antagonists of cIAPs may be efficacious in the treatment of chronic HBV infection and may promote elimination of virus. We have shown that cellular inhibitor of apoptosis proteins (cIAPs) impair clearance of hepatitis B virus (HBV) infection by preventing TNF-mediated killing/death of infected cells. A key question, with profound therapeutic implications, is whether this finding can be translated to the development of drugs that promote elimination of infected cells. Drug inhibitors of cIAPs were developed as cancer therapeutics to promote TNF-mediated tumor killing. These drugs are also known as Smac mimetics, because they mimic the action of the endogenous protein Smac/Diablo that antagonizes cIAP function. Here, we show using an immunocompetent mouse model of chronic HBV infection that birinapant and other Smac mimetics are able to rapidly reduce serum HBV DNA and serum HBV surface antigen, and they promote the elimination of hepatocytes containing HBV core antigen. The efficacy of Smac mimetics in treating HBV infection is dependent on their chemistry, host CD4+ T cells, and TNF. Birinapant enhances the ability of entecavir, an antiviral nucleoside analog, to reduce viral DNA production in HBV-infected animals. These results indicate that birinapant and other Smac mimetics may have efficacy in treating HBV infection and perhaps, other intracellular infections.


Proceedings of the National Academy of Sciences of the United States of America | 2015

Cellular inhibitor of apoptosis proteins prevent clearance of hepatitis B virus

Gregor Ebert; Simon Preston; Cody Allison; James Cooney; Jesse G. Toe; Michael D. Stutz; Samar Ojaimi; Hamish W. Scott; Nikola Baschuk; Ueli Nachbur; Joseph Torresi; Ruth Chin; Danielle Colledge; Xin Li; Nadia Warner; Peter Revill; Scott Bowden; John Silke; C. Glenn Begley; Marc Pellegrini

Significance Hepatitis B virus (HBV) causes substantial morbidity and mortality. A large proportion of infected individuals controls infection but does not completely eradicate HBV DNA from the liver, and flares in hepatitis can be precipitated by immunosuppression. A proportion of individuals never controls infection, and these people are at substantial risk of developing liver failure and liver cancer. Current therapies are not effective at eliminating virus, and there is a major interest in developing functional cures for HBV infection. We identified host cell signaling molecules that can restrict the ability to eradicate infected cells. These molecules can be therapeutically targeted, and drugs that interfere with the function of these host cell proteins may be useful therapies to promote clearance of HBV infection. Hepatitis B virus (HBV) infection can result in a spectrum of outcomes from immune-mediated control to disease progression, cirrhosis, and liver cancer. The host molecular pathways that influence and contribute to these outcomes need to be defined. Using an immunocompetent mouse model of chronic HBV infection, we identified some of the host cellular and molecular factors that impact on infection outcomes. Here, we show that cellular inhibitor of apoptosis proteins (cIAPs) attenuate TNF signaling during hepatitis B infection, and they restrict the death of infected hepatocytes, thus allowing viral persistence. Animals with a liver-specific cIAP1 and total cIAP2 deficiency efficiently control HBV infection compared with WT mice. This phenotype was partly recapitulated in mice that were deficient in cIAP2 alone. These results indicate that antagonizing the function of cIAPs may promote the clearance of HBV infection.


Nature Communications | 2015

A RIPK2 inhibitor delays NOD signalling events yet prevents inflammatory cytokine production

Ueli Nachbur; Che A. Stafford; Aleksandra Bankovacki; Yifan Zhan; Lisa Lindqvist; Berthe Katrine Fiil; Yelena Khakham; Hyun Ja Ko; Jarrod J. Sandow; Hendrik Falk; Jessica K. Holien; Diep Chau; Joanne M. Hildebrand; James E. Vince; Phillip P Sharp; Andrew I. Webb; Katherine A. Jackman; Sabrina Mühlen; Catherine L. Kennedy; Kym N. Lowes; James M. Murphy; Mads Gyrd-Hansen; Michael W. Parker; Elizabeth L. Hartland; Andrew M. Lew; David C. S. Huang; Guillaume Lessene; John Silke

Intracellular nucleotide binding and oligomerization domain (NOD) receptors recognize antigens including bacterial peptidoglycans and initiate immune responses by triggering the production of pro-inflammatory cytokines through activating NF-κB and MAP kinases. Receptor interacting protein kinase 2 (RIPK2) is critical for NOD-mediated NF-κB activation and cytokine production. Here we develop and characterize a selective RIPK2 kinase inhibitor, WEHI-345, which delays RIPK2 ubiquitylation and NF-κB activation downstream of NOD engagement. Despite only delaying NF-κB activation on NOD stimulation, WEHI-345 prevents cytokine production in vitro and in vivo and ameliorates experimental autoimmune encephalomyelitis in mice. Our study highlights the importance of the kinase activity of RIPK2 for proper immune responses and demonstrates the therapeutic potential of inhibiting RIPK2 in NOD-driven inflammatory diseases.


eLife | 2015

TRAF2 regulates TNF and NF-κB signalling to suppress apoptosis and skin inflammation independently of Sphingosine kinase 1.

Nima Etemadi; Michaël Chopin; Holly Anderton; Maria C. Tanzer; James A Rickard; Waruni Abeysekera; Cathrine Hall; Sukhdeep Kaur Spall; Bing Wang; Yuquan Xiong; Timothy Hla; Stuart M. Pitson; Claudine S. Bonder; W. Wei-Lynn Wong; Matthias Ernst; Gordon K. Smyth; David L. Vaux; Stephen L. Nutt; Ueli Nachbur; John Silke

TRAF2 is a component of TNF superfamily signalling complexes and plays an essential role in the regulation and homeostasis of immune cells. TRAF2 deficient mice die around birth, therefore its role in adult tissues is not well-explored. Furthermore, the role of the TRAF2 RING is controversial. It has been claimed that the atypical TRAF2 RING cannot function as a ubiquitin E3 ligase but counterclaimed that TRAF2 RING requires a co-factor, sphingosine-1-phosphate, that is generated by the enzyme sphingosine kinase 1, to function as an E3 ligase. Keratinocyte-specific deletion of Traf2, but not Sphk1 deficiency, disrupted TNF mediated NF-κB and MAP kinase signalling and caused epidermal hyperplasia and psoriatic skin inflammation. This inflammation was driven by TNF, cell death, non-canonical NF-κB and the adaptive immune system, and might therefore represent a clinically relevant model of psoriasis. TRAF2 therefore has essential tissue specific functions that do not overlap with those of Sphk1. DOI: http://dx.doi.org/10.7554/eLife.10592.001


Nature microbiology | 2017

EspL is a bacterial cysteine protease effector that cleaves RHIM proteins to block necroptosis and inflammation.

Jaclyn S. Pearson; Sabrina Mühlen; Ueli Nachbur; Chi L. L. Pham; Ying Zhang; Joanne M. Hildebrand; Clare V. Oates; Tania Wong Fok Lung; Danielle J. Ingle; Laura F. Dagley; Aleksandra Bankovacki; Emma J. Petrie; Gunnar N. Schroeder; Valerie F. Crepin; Gad Frankel; Seth L. Masters; James E. Vince; James M. Murphy; Margaret Sunde; Andrew I. Webb; John Silke; Elizabeth L. Hartland

Cell death signalling pathways contribute to tissue homeostasis and provide innate protection from infection. Adaptor proteins such as receptor-interacting serine/threonine-protein kinase 1 (RIPK1), receptor-interacting serine/threonine-protein kinase 3 (RIPK3), TIR-domain-containing adapter-inducing interferon-β (TRIF) and Z-DNA-binding protein 1 (ZBP1)/DNA-dependent activator of IFN-regulatory factors (DAI) that contain receptor-interacting protein (RIP) homotypic interaction motifs (RHIM) play a key role in cell death and inflammatory signalling1–3. RHIM-dependent interactions help drive a caspase-independent form of cell death termed necroptosis4,5. Here, we report that the bacterial pathogen enteropathogenic Escherichia coli (EPEC) uses the type III secretion system (T3SS) effector EspL to degrade the RHIM-containing proteins RIPK1, RIPK3, TRIF and ZBP1/DAI during infection. This requires a previously unrecognized tripartite cysteine protease motif in EspL (Cys47, His131, Asp153) that cleaves within the RHIM of these proteins. Bacterial infection and/or ectopic expression of EspL leads to rapid inactivation of RIPK1, RIPK3, TRIF and ZBP1/DAI and inhibition of tumour necrosis factor (TNF), lipopolysaccharide or polyinosinic:polycytidylic acid (poly(I:C))-induced necroptosis and inflammatory signalling. Furthermore, EPEC infection inhibits TNF-induced phosphorylation and plasma membrane localization of mixed lineage kinase domain-like pseudokinase (MLKL). In vivo, EspL cysteine protease activity contributes to persistent colonization of mice by the EPEC-like mouse pathogen Citrobacter rodentium. The activity of EspL defines a family of T3SS cysteine protease effectors found in a range of bacteria and reveals a mechanism by which gastrointestinal pathogens directly target RHIM-dependent inflammatory and necroptotic signalling pathways.


FEBS Journal | 2013

Lymphotoxin α induces apoptosis, necroptosis and inflammatory signals with the same potency as tumour necrosis factor.

Nima Etemadi; Jessica K. Holien; Diep Chau; Grant Dewson; James M. Murphy; Warren S. Alexander; Michael W. Parker; John Silke; Ueli Nachbur

Both of the TNF superfamily ligands, TNF and LTα, can bind and signal through TNFR1 and TNFR2, yet mice mutant for each have different phenotypes. Part of this difference is because LTα but not TNF can activate Herpes Virus Entry Mediator and also heterotrimerise with LTβ to activate LTβR, which is consistent with the similar phenotypes of the LTα and LTβR deficient mice. However, it has also been reported that the LTα3 homotrimer signals differently than TNF through TNFR1, and has unique roles in initiation and exacerbation of some inflammatory diseases. Our modeling of the TNF/TNFR1 interface compared to the LTα3/TNFR1 structure revealed some differences that could affect signalling by the two ligands. To determine whether there were any functional differences in the ability of TNF and LTα3 to induce TNFR1‐dependent apoptosis or necroptosis, and if there were different requirements for cIAPs and Sharpin to transmit the TNFR1 signal, we compared the ability of cells to respond to TNF and LTα3. Contrary to our hypothesis, we were unable to discover differences in signalling by TNFR1 in response to TNF and LTα3. Our results imply that the reasons for the conservation of LTα are most likely due either to differential regulation, the ability to signal through Herpes Virus Entry Mediator or the ability of LTα to form heterotrimers with LTβ.

Collaboration


Dive into the Ueli Nachbur's collaboration.

Top Co-Authors

Avatar

John Silke

University of Melbourne

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Aleksandra Bankovacki

Walter and Eliza Hall Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar

Nima Etemadi

Walter and Eliza Hall Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar

Andrew I. Webb

Walter and Eliza Hall Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar

James A Rickard

Walter and Eliza Hall Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Holly Anderton

Walter and Eliza Hall Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge