Network


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

Hotspot


Dive into the research topics where Ruth E. Watkinson is active.

Publication


Featured researches published by Ruth E. Watkinson.


Nature Immunology | 2013

Intracellular antibody-bound pathogens stimulate immune signaling via the Fc receptor TRIM21

William A. McEwan; Jerry C. H. Tam; Ruth E. Watkinson; Susanna R. Bidgood; Donna L. Mallery; Leo C. James

During pathogen infection, antibodies can be carried into the infected cell, where they are detected by the ubiquitously expressed cytosolic antibody receptor TRIM21. Here we found that recognition of intracellular antibodies by TRIM21 activated immune signaling. TRIM21 catalyzed the formation of Lys63 (K63)-linked ubiquitin chains and stimulated the transcription factor pathways of NF-κB, AP-1, IRF3, IRF5 and IRF7. Activation resulted in the production of proinflammatory cytokines, modulation of natural killer stress ligands and induction of an antiviral state. Intracellular antibody signaling was abrogated by genetic deletion of TRIM21 and was restored by ectopic expression of TRIM21. The sensing of antibodies by TRIM21 was stimulated after infection by DNA or RNA nonenveloped viruses or intracellular bacteria. Thus, the antibody-TRIM21 detection system provides potent, comprehensive activation of the innate immune system independently of known pattern-recognition receptors.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Intracellular antibody receptor TRIM21 prevents fatal viral infection

Marina Vaysburd; Ruth E. Watkinson; Cooper H; Reed M; O'Connell K; Smith J; Cruickshanks J; Leo C. James

Host species have evolved mechanisms that can inhibit pathogen replication even after a cell has been successfully invaded. Here we show that tripartite-motif protein 21 (TRIM21), a ubiquitously expressed E3 ubiquitin ligase that targets viruses inside the cytosol, protects mice against fatal viral infection. Upon infection with mouse adenovirus-1, naive mice lacking TRIM21 succumb to encephalomyelitis within 7 d. In contrast, wild-type mice rapidly up-regulate TRIM21 and control viremia. Trim21 heterozygous mice have a haploinsufficiency phenotype in which reduced TRIM21 expression leads to a viral load that is higher than wild types but lower than knockouts. TRIM21 is a high-affinity antibody receptor that allows antibodies to operate inside an infected cell. In passive transfer experiments at high viral dose, antisera that fully protects wild-type mice fails to protect most Trim21 knockout animals. These results demonstrate that TRIM21 provides potent antiviral protection and forms an important part of the humoral immune response.


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

Sequential ubiquitination and deubiquitination enzymes synchronize the dual sensor and effector functions of TRIM21

Adam J. Fletcher; Donna L. Mallery; Ruth E. Watkinson; Claire F. Dickson; Leo C. James

Significance Tripartite motif (TRIM) proteins are a large family of E3 ubiquitin (Ub) ligases, with many members having important roles in innate immunity. TRIM21 is a cytosolic antibody receptor that recognizes the Fc portion of antibodies bound to incoming virions. On binding to these immune complexes, TRIM21 triggers the catastrophic disassembly of viral capsids at the proteasome, terminating viral infection. Simultaneously, TRIM21 “senses” the presence of the virus and provokes signaling cascades that activate the transcription factor NF-κB, ultimately alerting surrounding cells to the infection. Here, we uncover the stepwise ubiquitination mechanism catalyzed by TRIM21, as well as the various cofactors required, that allows these two antiviral activities to occur synchronously at the proteasome. Tripartite motif (TRIM) 21 is a cytosolic antibody receptor that neutralizes antibody-coated viruses that penetrate the cell and simultaneously activates innate immunity. Here we show that the conjugation of TRIM21 with K63-linked ubiquitin (Ub-63Ub) catalyzed by the sequential activity of nonredundant E2 Ub enzymes is required for its dual antiviral functions. TRIM21 is first labeled with monoubiquitin (monoUb) by the E2 Ube2W. The monoUb is a substrate for the heterodimeric E2 Ube2N/Ube2V2, resulting in TRIM21-anchored Ub-63Ub. Depletion of either E2 abolishes Ub-63Ub and Ub-48Ub conjugation of TRIM21, NF-κB signaling, and virus neutralization. The formation of TRIM21-Ub-63Ub precedes proteasome recruitment, and we identify an essential role for the 19S-resident and degradation-coupled deubiquitinase Poh1 in TRIM21 neutralization, signaling, and cytokine induction. This study elucidates a complex mechanism of step-wise ubiquitination and deubiquitination activities that allows contemporaneous innate immune signaling and neutralization by TRIM21.


Immunological Reviews | 2015

TRIM21: a cytosolic Fc receptor with broad antibody isotype specificity

Stian Foss; Ruth E. Watkinson; Inger Sandlie; Leo C. James; Jan Terje Andersen

Antibodies are key molecules in the fight against infections. Although previously thought to mediate protection solely in the extracellular environment, recent research has revealed that antibody‐mediated protection extends to the cytosolic compartment of cells. This postentry viral defense mechanism requires binding of the antibody to a cytosolic Fc receptor named tripartite motif containing 21 (TRIM21). In contrast to other Fc receptors, TRIM21 shows remarkably broad isotype specificity as it does not only bind IgG but also IgM and IgA. When viral pathogens coated with these antibody isotypes enter the cytosol, TRIM21 is rapidly recruited and efficient neutralization occurs before the virus has had the time to replicate. In addition, inflammatory signaling is induced. As such, TRIM21 acts as a cytosolic sensor that engages antibodies that have failed to protect against infection in the extracellular environment. Here, we summarize our current understanding of how TRIM21 orchestrates humoral immunity in the cytosolic environment.


PLOS Pathogens | 2015

TRIM21 Promotes cGAS and RIG-I Sensing of Viral Genomes during Infection by Antibody-Opsonized Virus

Ruth E. Watkinson; William A. McEwan; Jerry C. H. Tam; Marina Vaysburd; Leo C. James

Encapsidation is a strategy almost universally employed by viruses to protect their genomes from degradation and from innate immune sensors. We show that TRIM21, which targets antibody-opsonized virions for proteasomal destruction, circumvents this protection, enabling the rapid detection and degradation of viral genomes before their replication. TRIM21 triggers an initial wave of cytokine transcription that is antibody, rather than pathogen, driven. This early response is augmented by a second transcriptional program, determined by the nature of the infecting virus. In this second response, TRIM21-induced exposure of the viral genome promotes sensing of DNA and RNA viruses by cGAS and RIG-I. This mechanism allows early detection of an infection event and drives an inflammatory response in mice within hours of viral challenge.


Journal of Virology | 2013

Simultaneous Neutralization and Innate Immune Detection of a Replicating Virus by TRIM21

Ruth E. Watkinson; Jerry C. H. Tam; Marina Vaysburd; Leo C. James

ABSTRACT Tripartite motif-containing 21 (TRIM21) is a cytosolic immunoglobulin receptor that mediates antibody-dependent intracellular neutralization (ADIN). Here we show that TRIM21 potently inhibits the spreading infection of a replicating cytopathic virus and activates innate immunity. We used a quantitative PCR (qPCR)-based assay to measure in vitro replication of mouse adenovirus type 1 (MAV-1), a virus that causes dose-dependent hemorrhagic encephalitis in mice. Using this assay, we show that genetic ablation of TRIM21 or chemical inhibition of either the AAA ATPase p97/valosin-containing protein (VCP) or the proteasome results in a >1,000-fold increase in the relative level of infection in the presence of immune serum. Moreover, the TRIM21-mediated ability of antisera to block replication was a consistent feature of the humoral immune response in immunized mice. In the presence of immune sera and upon infection, TRIM21 also activates a proinflammatory response, resulting in secretion of tumor necrosis factor alpha (TNF-α) and interleukin-6 (IL-6). These results demonstrate that TRIM21 provides a potent block to spreading infection and induces an antiviral state.


Journal of Clinical Immunology | 2014

Intracellular Antibody Immunity

Ruth E. Watkinson; William A. McEwan; Leo C. James

Antibodies allow the immune system to target pathogens despite their tremendous diversity and rapid evolution. Once bound to a pathogen, antibodies induce a broad range of effector mechanisms, including phagocytosis and complement. However, these mechanisms are all initiated in the extracellular space, meaning that pathogens like viruses evade them upon infection of their target cells. Recently, it has been shown that, in addition to mediating extracellular immune responses, antibodies also activate immunity inside infected cells. Antibodies that are bound to the surface of non-enveloped viruses or bacteria are carried into the cell during pathogen entry. Once inside the cell, these pathogen-attached antibodies are recognised by a highly conserved, high affinity cytosolic antibody receptor called TRIM21. TRIM21 initiates both sensor and effector responses that reduce viral replication and induce an antiviral state. These responses are an important part of antiviral immunity and the removal of TRIM21 results in uncontrolled viraemia and death in a mouse model of infection.


Journal of Immunology | 2016

TRIM21 Immune Signaling Is More Sensitive to Antibody Affinity Than Its Neutralization Activity.

Stian Foss; Ruth E. Watkinson; Algirdas Grevys; Martin B. McAdam; Malin Bern; Lene Støkken Høydahl; Bjørn Dalhus; Terje E. Michaelsen; Inger Sandlie; Leo C. James; Jan Terje Andersen

Ab-coated viruses can be detected in the cytosol by the FcR tripartite motif-containing 21 (TRIM21), which rapidly recruits the proteasomal machinery and triggers induction of immune signaling. As such, TRIM21 plays a key role in intracellular protection by targeting invading viruses for destruction and alerting the immune system. A hallmark of immunity is elicitation of a balanced response that is proportionate to the threat, to avoid unnecessary inflammation. In this article, we show how Ab affinity modulates TRIM21 immune function. We constructed a humanized monoclonal IgG1 against human adenovirus type 5 (AdV5) and a panel of Fc-engineered variants with a wide range of affinities for TRIM21. We found that IgG1-coated viral particles were neutralized via TRIM21, even when affinity was reduced by as much as 100-fold. In contrast, induction of NF-κB signaling was more sensitive to reduced affinity between TRIM21 and the Ab variants. Thus, TRIM21 mediates neutralization under suboptimal conditions, whereas induction of immune signaling is balanced according to the functional affinity for the incoming immune stimuli. Our findings have implications for engineering of antiviral IgG therapeutics with tailored effector functions.


Scientific Reports | 2016

Antibody-antigen kinetics constrain intracellular humoral immunity.

Maria Bottermann; Heidrun Elisabeth Lode; Ruth E. Watkinson; Stian Foss; Inger Sandlie; Jan Terje Andersen; Leo C. James

During infection with non-enveloped viruses, antibodies stimulate immunity from inside cells by activating the cytosolic Fc receptor TRIM21. This intracellular humoral response relies on opsonized viral particles reaching the cytosol intact but the antigenic and kinetic constraints involved are unknown. We have solved the structure of a potent TRIM21-dependent neutralizing antibody in complex with human adenovirus 5 hexon and show how these properties influence immune activity. Structure-guided mutagenesis was used to generate antibodies with 20,000-fold variation in affinity, on-rates that differ by ~50-fold and off-rates by >175-fold. Characterization of these variants during infection revealed that TRIM21-dependent neutralization and NFκB activation was largely unaffected by on-rate kinetics. In contrast, TRIM21 antiviral activity was exquisitely dependent upon off-rate, with sub-μM affinity antibodies nevertheless unable to stimulate signaling because of fast dissociation kinetics. These results define the antibody properties required to elicit an efficient intracellular immune response during viral infection.


Scientific Reports | 2017

Corrigendum: Antibody-antigen kinetics constrain intracellular humoral immunity

Maria Bottermann; Heidrun Elisabeth Lode; Ruth E. Watkinson; Stian Foss; Inger Sandlie; Jan Terje Andersen; Leo C. James

Scientific Reports 6: Article number: 37457; published online: 24 November 2016; updated: 06 April 2017 This Article contains a typographical error in the Methods section under the subheading ‘Crystallization’, where the Protein Data Bank accession code ‘5LDN’ was incorrectly given as ‘5LDV’.

Collaboration


Dive into the Ruth E. Watkinson's collaboration.

Top Co-Authors

Avatar

Leo C. James

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

William A. McEwan

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar

Donna L. Mallery

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar

Jerry C. H. Tam

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar

Marina Vaysburd

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar

Maria Bottermann

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar

Susanna R. Bidgood

Laboratory of Molecular Biology

View shared research outputs
Researchain Logo
Decentralizing Knowledge