Network


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

Hotspot


Dive into the research topics where Tycho E. T. Mevissen is active.

Publication


Featured researches published by Tycho E. T. Mevissen.


Cell | 2013

OTU Deubiquitinases Reveal Mechanisms of Linkage Specificity and Enable Ubiquitin Chain Restriction Analysis

Tycho E. T. Mevissen; Manuela K. Hospenthal; Paul P. Geurink; Paul R. Elliott; Masato Akutsu; Nadia Arnaudo; Reggy Ekkebus; Yogesh Kulathu; Tobias Wauer; Farid El Oualid; Stefan M. V. Freund; Huib Ovaa; David Komander

Summary Sixteen ovarian tumor (OTU) family deubiquitinases (DUBs) exist in humans, and most members regulate cell-signaling cascades. Several OTU DUBs were reported to be ubiquitin (Ub) chain linkage specific, but comprehensive analyses are missing, and the underlying mechanisms of linkage specificity are unclear. Using Ub chains of all eight linkage types, we reveal that most human OTU enzymes are linkage specific, preferring one, two, or a defined subset of linkage types, including unstudied atypical Ub chains. Biochemical analysis and five crystal structures of OTU DUBs with or without Ub substrates reveal four mechanisms of linkage specificity. Additional Ub-binding domains, the ubiquitinated sequence in the substrate, and defined S1’ and S2 Ub-binding sites on the OTU domain enable OTU DUBs to distinguish linkage types. We introduce Ub chain restriction analysis, in which OTU DUBs are used as restriction enzymes to reveal linkage type and the relative abundance of Ub chains on substrates.


Nature Communications | 2013

Regulation of A20 and Other Otu Deubiquitinases by Reversible Oxidation

Yogesh Kulathu; Francisco J. Garcia; Tycho E. T. Mevissen; Martin Busch; Nadia Arnaudo; Kate S. Carroll; David Barford; David Komander

Protein ubiquitination is a highly versatile posttranslational modification that regulates as diverse processes as protein degradation and kinase activation. Deubiquitinases hydrolyse ubiquitin modifications from proteins and are hence key regulators of the ubiquitin system. Ovarian Tumour (OTU) deubiquitinases comprise a family of fourteen human enzymes, many of which regulate cellular signalling pathways. OTU deubiquitinases are cysteine proteases that cleave polyubiquitin (polyUb) chains in vitro and in cells, but little is currently known about their regulation. Here we show that OTU deubiquitinases are susceptible to reversible oxidation of the catalytic cysteine residue. High-resolution crystal structures of the catalytic domain of A20 in four different oxidation states reveal that the reversible form of A20 oxidation is a cysteine sulphenic acid intermediate, which is stabilised by the architecture of the catalytic centre. Using chemical tools to detect sulphenic acid intermediates, we show that many OTU deubiquitinases undergo reversible oxidation upon treatment with H2O2, revealing a new mechanism to regulate deubiquitinase activity.


Nature Structural & Molecular Biology | 2012

An ankyrin-repeat ubiquitin-binding domain determines TRABID's specificity for atypical ubiquitin chains.

Julien Licchesi; Juliusz Mieszczanek; Tycho E. T. Mevissen; Trevor J. Rutherford; Masato Akutsu; Satpal Virdee; Farid El Oualid; Jason W. Chin; Huib Ovaa; Mariann Bienz; David Komander

Eight different types of ubiquitin linkages are present in eukaryotic cells that regulate diverse biological processes. Proteins that mediate specific assembly and disassembly of atypical Lys6, Lys27, Lys29 and Lys33 linkages are mainly unknown. We here reveal how the human ovarian tumor (OTU) domain deubiquitinase (DUB) TRABID specifically hydrolyzes both Lys29- and Lys33-linked diubiquitin. A crystal structure of the extended catalytic domain reveals an unpredicted ankyrin repeat domain that precedes an A20-like catalytic core. NMR analysis identifies the ankyrin domain as a new ubiquitin-binding fold, which we have termed AnkUBD, and DUB assays in vitro and in vivo show that this domain is crucial for TRABID efficiency and linkage specificity. Our data are consistent with AnkUBD functioning as an enzymatic S1′ ubiquitin-binding site, which orients a ubiquitin chain so that Lys29 and Lys33 linkages are cleaved preferentially.


Journal of Biological Chemistry | 2014

Lysine 27 Ubiquitination of the Mitochondrial Transport Protein Miro Is Dependent on Serine 65 of the Parkin Ubiquitin Ligase

Nicol Birsa; Rosalind Norkett; Tobias Wauer; Tycho E. T. Mevissen; Hsiu-Chuan Wu; Thomas Foltynie; Kailash P. Bhatia; Warren D. Hirst; David Komander; Helene Plun-Favreau; Josef T. Kittler

Background: Miro is a mitochondrial protein involved in mitochondrial trafficking. Results: Mitochondrial damage drives rapid Miro ubiquitination in a manner dependent on Ser-65 in Parkin; however, Miro degradation is delayed. Conclusion: Ubiquitination of Miro, rather than its degradation, could act as a signal for mitochondrial arrest and clearance. Significance: Disruption of the mitochondrial transport machinery could be implicated in Parkinson disease. Mitochondrial transport plays an important role in matching mitochondrial distribution to localized energy production and calcium buffering requirements. Here, we demonstrate that Miro1, an outer mitochondrial membrane (OMM) protein crucial for the regulation of mitochondrial trafficking and distribution, is a substrate of the PINK1/Parkin mitochondrial quality control system in human dopaminergic neuroblastoma cells. Moreover, Miro1 turnover on damaged mitochondria is altered in Parkinson disease (PD) patient-derived fibroblasts containing a pathogenic mutation in the PARK2 gene (encoding Parkin). By analyzing the kinetics of Miro1 ubiquitination, we further demonstrate that mitochondrial damage triggers rapid (within minutes) and persistent Lys-27-type ubiquitination of Miro1 on the OMM, dependent on PINK1 and Parkin. Proteasomal degradation of Miro1 is then seen on a slower time scale, within 2–3 h of the onset of ubiquitination. We find Miro ubiquitination in dopaminergic neuroblastoma cells is independent of Miro1 phosphorylation at Ser-156 but is dependent on the recently identified Ser-65 residue within Parkin that is phosphorylated by PINK1. Interestingly, we find that Miro1 can stabilize phospho-mutant versions of Parkin on the OMM, suggesting that Miro is also part of a Parkin receptor complex. Moreover, we demonstrate that Ser-65 in Parkin is critical for regulating Miro levels upon mitochondrial damage in rodent cortical neurons. Our results provide new insights into the ubiquitination-dependent regulation of the Miro-mediated mitochondrial transport machinery by PINK1/Parkin and also suggest that disruption of this regulation may be implicated in Parkinson disease pathogenesis.


Nature Protocols | 2015

Deubiquitinase-based analysis of ubiquitin chain architecture using Ubiquitin Chain Restriction (UbiCRest)

Manuela K. Hospenthal; Tycho E. T. Mevissen; David Komander

Protein ubiquitination is a versatile protein modification that regulates virtually all cellular processes. This versatility originates from polyubiquitin chains, which can be linked in eight distinct ways. The combinatorial complexity of eight linkage types in homotypic (one chain type per polymer) and heterotypic (multiple linkage types per polymer) chains poses significant problems for biochemical analysis. Here we describe UbiCRest, in which substrates (ubiquitinated proteins or polyubiquitin chains) are treated with a panel of linkage-specific deubiquitinating enzymes (DUBs) in parallel reactions, followed by gel-based analysis. UbiCRest can be used to show that a protein is ubiquitinated, to identify which linkage type(s) are present on polyubiquitinated proteins and to assess the architecture of heterotypic polyubiquitin chains. DUBs used in UbiCRest can be obtained commercially; however, we include details for generating a toolkit of purified DUBs and for profiling their linkage preferences in vitro. UbiCRest is a qualitative method that yields insights into ubiquitin chain linkage types and architecture within hours, and it can be performed on western blotting quantities of endogenously ubiquitinated proteins.


Annual Review of Biochemistry | 2017

Mechanisms of Deubiquitinase Specificity and Regulation

Tycho E. T. Mevissen; David Komander

Protein ubiquitination is one of the most powerful posttranslational modifications of proteins, as it regulates a plethora of cellular processes in distinct manners. Simple monoubiquitination events coexist with more complex forms of polyubiquitination, the latter featuring many different chain architectures. Ubiquitin can be subjected to further posttranslational modifications (e.g., phosphorylation and acetylation) and can also be part of mixed polymers with ubiquitin-like modifiers such as SUMO (small ubiquitin-related modifier) or NEDD8 (neural precursor cell expressed, developmentally downregulated 8). Together, cellular ubiquitination events form a sophisticated and versatile ubiquitin code. Deubiquitinases (DUBs) reverse ubiquitin signals with equally high sophistication. In this review, we conceptualize the many layers of specificity that DUBs encompass to control the ubiquitin code and discuss examples in which DUB specificity has been understood at the molecular level. We further discuss the many mechanisms of DUB regulation with a focus on those that modulate catalytic activity. Our review provides a framework to tackle lingering questions in DUB biology.


Chemistry & Biology | 2016

Non-hydrolyzable Diubiquitin Probes Reveal Linkage-Specific Reactivity of Deubiquitylating Enzymes Mediated by S2 Pockets

Dennis Flierman; Gerbrand J. van der Heden van Noort; Reggy Ekkebus; Paul P. Geurink; Tycho E. T. Mevissen; Manuela K. Hospenthal; David Komander; Huib Ovaa

Summary Ubiquitin chains are important post-translational modifications that control a large number of cellular processes. Chains can be formed via different linkages, which determines the type of signal they convey. Deubiquitylating enzymes (DUBs) regulate ubiquitylation status by trimming or removing chains from attached proteins. DUBs can contain several ubiquitin-binding pockets, which confer specificity toward differently linked chains. Most tools for monitoring DUB specificity target binding pockets on opposing sides of the active site; however, some DUBs contain additional pockets. Therefore, reagents targeting additional pockets are essential to fully understand linkage specificity. We report the development of active site-directed probes and fluorogenic substrates, based on non-hydrolyzable diubiquitin, that are equipped with a C-terminal warhead or a fluorogenic activity reporter moiety. We demonstrate that various DUBs in lysates display differential reactivity toward differently linked diubiquitin probes, as exemplified by the proteasome-associated DUB USP14. In addition, OTUD2 and OTUD3 show remarkable linkage-specific reactivity with our diubiquitin-based reagents.


Nature | 2016

Molecular basis of Lys11-polyubiquitin specificity in the deubiquitinase Cezanne

Tycho E. T. Mevissen; Yogesh Kulathu; Monique P. C. Mulder; Paul P. Geurink; Sarah L. Maslen; Malte Gersch; Paul Elliott; John E. Burke; Bianca D. M. van Tol; Masato Akutsu; Farid El Oualid; Masato Kawasaki; Stefan M. V. Freund; Huib Ovaa; David Komander

The post-translational modification of proteins with polyubiquitin regulates virtually all aspects of cell biology. Eight distinct chain linkage types co-exist in polyubiquitin and are independently regulated in cells. This ‘ubiquitin code’ determines the fate of the modified protein. Deubiquitinating enzymes of the ovarian tumour (OTU) family regulate cellular signalling by targeting distinct linkage types within polyubiquitin, and understanding their mechanisms of linkage specificity gives fundamental insights into the ubiquitin system. Here we reveal how the deubiquitinase Cezanne (also known as OTUD7B) specifically targets Lys11-linked polyubiquitin. Crystal structures of Cezanne alone and in complex with monoubiquitin and Lys11-linked diubiquitin, in combination with hydrogen–deuterium exchange mass spectrometry, enable us to reconstruct the enzymatic cycle in great detail. An intricate mechanism of ubiquitin-assisted conformational changes activates the enzyme, and while all chain types interact with the enzymatic S1 site, only Lys11-linked chains can bind productively across the active site and stimulate catalytic turnover. Our work highlights the plasticity of deubiquitinases and indicates that new conformational states can occur when a true substrate, such as diubiquitin, is bound at the active site.


ChemBioChem | 2016

Development of Diubiquitin-Based FRET Probes To Quantify Ubiquitin Linkage Specificity of Deubiquitinating Enzymes.

Paul P. Geurink; Bianca D. M. van Tol; Duco van Dalen; Paul J. G. Brundel; Tycho E. T. Mevissen; Jonathan N. Pruneda; Paul R. Elliott; Gabriëlle Ba van Tilburg; David Komander; Huib Ovaa

Deubiquitinating enzymes (DUBs) are proteases that fulfill crucial roles in the ubiquitin (Ub) system, by deconjugation of Ub from its targets and disassembly of polyUb chains. The specificity of a DUB towards one of the polyUb chain linkages largely determines the ultimate signaling function. We present a novel set of diubiquitin FRET probes, comprising all seven isopeptide linkages, for the absolute quantification of chain cleavage specificity of DUBs by means of Michaelis–Menten kinetics. Each probe is equipped with a FRET pair consisting of Rhodamine110 and tetramethylrhodamine to allow the fully synthetic preparation of the probes by SPPS and NCL. Our synthetic strategy includes the introduction of N,N′‐Boc‐protected 5‐carboxyrhodamine as a convenient building block in peptide chemistry. We demonstrate the value of our probes by quantifying the linkage specificities of a panel of nine DUBs in a high‐throughput manner.


Molecular Biology of the Cell | 2015

Efficient APC/C substrate degradation in cells undergoing mitotic exit depends on K11 ubiquitin linkages

Mingwei Min; Tycho E. T. Mevissen; Maria De Luca; David Komander; Catherine Lindon

K11- and K48-linked polyubiquitin constitute alternative destruction signals on substrates of the APC/C. Substrates targeted late in mitotic exit, including Aurora kinases, Polo-like kinase, and KIFC1, all depend on K11 linkages for degradation. APC/C coactivator Cdh1 directs K11 linkage assembly via UBE2S in a substrate-specific manner.

Collaboration


Dive into the Tycho E. T. Mevissen's collaboration.

Top Co-Authors

Avatar

David Komander

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar

Huib Ovaa

Netherlands Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Paul P. Geurink

Netherlands Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Manuela K. Hospenthal

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar

Tobias Wauer

Laboratory of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Farid El Oualid

Netherlands Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Masato Akutsu

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hsiu-Chuan Wu

UCL Institute of Neurology

View shared research outputs
Researchain Logo
Decentralizing Knowledge