Network


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

Hotspot


Dive into the research topics where Karla L. H. Feijs is active.

Publication


Featured researches published by Karla L. H. Feijs.


Nature Structural & Molecular Biology | 2013

Macrodomain-containing proteins are new mono-ADP-ribosylhydrolases

Florian Rosenthal; Karla L. H. Feijs; Emilie Frugier; Mario Bonalli; Alexandra H. Forst; Ralph Imhof; Hans Christian Winkler; David Fischer; Amedeo Caflisch; Paul O. Hassa; Bernhard Lüscher; Michael O. Hottiger

ADP-ribosylation is an important post-translational protein modification (PTM) that regulates diverse biological processes. ADP-ribosyltransferase diphtheria toxin-like 10 (ARTD10, also known as PARP10) mono-ADP-ribosylates acidic side chains and is one of eighteen ADP-ribosyltransferases that catalyze mono- or poly-ADP-ribosylation of target proteins. Currently, no enzyme is known that reverses ARTD10-catalyzed mono-ADP-ribosylation. Here we report that ARTD10-modified targets are substrates for the macrodomain proteins MacroD1, MacroD2 and C6orf130 from Homo sapiens as well as for the macrodomain protein Af1521 from archaebacteria. Structural modeling and mutagenesis of MacroD1 and MacroD2 revealed a common core structure with Asp102 and His106 of MacroD2 implicated in the hydrolytic reaction. Notably, MacroD2 reversed the ARTD10-catalyzed, mono-ADP-ribose–mediated inhibition of glycogen synthase kinase 3β (GSK3β) in vitro and in cells, thus underlining the physiological and regulatory importance of mono-ADP-ribosylhydrolase activity. Our results establish macrodomain-containing proteins as mono-ADP-ribosylhydrolases and define a class of enzymes that renders mono-ADP-ribosylation a reversible modification.


Cell Communication and Signaling | 2013

ARTD10 substrate identification on protein microarrays: regulation of GSK3β by mono-ADP-ribosylation

Karla L. H. Feijs; Henning Kleine; Anne K. Braczynski; Alexandra H. Forst; Nicolas Herzog; Patricia Verheugd; Ulrike Linzen; Elisabeth Kremmer; Bernhard Lüscher

BackgroundAlthough ADP-ribosylation has been described five decades ago, only recently a distinction has been made between eukaryotic intracellular poly- and mono-ADP-ribosylating enzymes. Poly-ADP-ribosylation by ARTD1 (formerly PARP1) is best known for its role in DNA damage repair. Other polymer forming enzymes are ARTD2 (formerly PARP2), ARTD3 (formerly PARP3) and ARTD5/6 (formerly Tankyrase 1/2), the latter being involved in Wnt signaling and regulation of 3BP2. Thus several different functions of poly-ADP-ribosylation have been well described whereas intracellular mono-ADP-ribosylation is currently largely undefined. It is for example not known which proteins function as substrate for the different mono-ARTDs. This is partially due to lack of suitable reagents to study mono-ADP-ribosylation, which limits the current understanding of this post-translational modification.ResultsWe have optimized a novel screening method employing protein microarrays, ProtoArrays®, applied here for the identification of substrates of ARTD10 (formerly PARP10) and ARTD8 (formerly PARP14). The results of this substrate screen were validated using in vitro ADP-ribosylation assays with recombinant proteins. Further analysis of the novel ARTD10 substrate GSK3β revealed mono-ADP-ribosylation as a regulatory mechanism of kinase activity by non-competitive inhibition in vitro. Additionally, manipulation of the ARTD10 levels in cells accordingly influenced GSK3β activity. Together these data provide the first evidence for a role of endogenous mono-ADP-ribosylation in intracellular signaling.ConclusionsOur findings indicate that substrates of ADP-ribosyltransferases can be identified using protein microarrays. The discovered substrates of ARTD10 and ARTD8 provide the first sets of proteins that are modified by mono-ADP-ribosyltransferases in vitro. By studying one of the ARTD10 substrates more closely, the kinase GSK3β, we identified mono-ADP-ribosylation as a negative regulator of kinase activity.


Nature Communications | 2013

Regulation of NF-κB signalling by the mono-ADP-ribosyltransferase ARTD10

Patricia Verheugd; Alexandra H. Forst; Larissa Milke; Nicolas Herzog; Karla L. H. Feijs; Elisabeth Kremmer; Henning Kleine; Bernhard Lüscher

Adenosine diphosphate-ribosylation is a post-translational modification mediated by intracellular and membrane-associated extracellular enzymes and many bacterial toxins. The intracellular enzymes modify their substrates either by poly-ADP-ribosylation, exemplified by ARTD1/PARP1, or by mono-ADP-ribosylation. The latter has been discovered only recently, and little is known about its physiological relevance. The founding member of mono-ADP-ribosyltransferases is ARTD10/PARP10. It possesses two ubiquitin-interaction motifs, a unique feature among ARTD/PARP enzymes. Here, we find that the ARTD10 ubiquitin-interaction motifs bind to K63-linked poly-ubiquitin, a modification that is essential for NF-κB signalling. We therefore studied the role of ARTD10 in this pathway. ARTD10 inhibits the activation of NF-κB and downstream target genes in response to interleukin-1β and tumour necrosis factor-α, dependent on catalytic activity and poly-ubiquitin binding of ARTD10. Mechanistically ARTD10 interferes with poly-ubiquitination of NEMO, which interacts with and is a substrate of ARTD10. Our findings identify a novel regulator of NF-κB signalling and provide evidence for cross-talk between K63-linked poly-ubiquitination and mono-ADP-ribosylation.


FEBS Journal | 2013

Expanding functions of intracellular resident mono-ADP-ribosylation in cell physiology

Karla L. H. Feijs; Patricia Verheugd; Bernhard Lüscher

Poly‐ADP‐ribosylation functions in diverse signaling pathways, such as Wnt signaling and DNA damage repair, where its role is relatively well characterized. Contrarily, mono‐ADP‐ribosylation by for example ARTD10/PARP10 is much less understood. Recent developments hint at the involvement of mono‐ADP‐ribosylation in transcriptional regulation, the unfolded protein response, DNA repair, insulin secretion and immunity. Additionally, macrodomain‐containing hydrolases, MacroD1, MacroD2 and C6orf130/TARG1, have been identified that make mono‐ADP‐ribosylation reversible. Complicating further progress is the lack of tools such as mono‐ADP‐ribose‐specific antibodies. The currently known functions of mono‐ADP‐ribosylation are summarized here, as well as the available tools such as mass spectrometry to study this modification in vitro and in cells.


Cell Communication and Signaling | 2012

Dynamic subcellular localization of the mono-ADP-ribosyltransferase ARTD10 and interaction with the ubiquitin receptor p62

Henning Kleine; Andreas Herrmann; Trond Lamark; Alexandra H. Forst; Patricia Verheugd; Juliane Lüscher-Firzlaff; Barbara E. Lippok; Karla L. H. Feijs; Nicolas Herzog; Elisabeth Kremmer; Terje Johansen; Gerhard Müller-Newen; Bernhard Lüscher

BackgroundADP-ribosylation is a posttranslational modification catalyzed in cells by ADP-ribosyltransferases (ARTD or PARP enzymes). The ARTD family consists of 17 members. Some ARTDs modify their substrates by adding ADP-ribose in an iterative process, thereby synthesizing ADP-ribose polymers, the best-studied example being ARTD1/PARP1. Other ARTDs appear to mono-ADP-ribosylate their substrates and are unable to form polymers. The founding member of this latter subclass is ARTD10/PARP10, which we identified as an interaction partner of the nuclear oncoprotein MYC. Biochemically ARTD10 uses substrate-assisted catalysis to modify its substrates. Our previous studies indicated that ARTD10 may shuttle between the nuclear and cytoplasmic compartments. We have now addressed this in more detail.ResultsWe have characterized the subcellular localization of ARTD10 using live-cell imaging techniques. ARTD10 shuttles between the cytoplasmic and nuclear compartments. When nuclear, ARTD10 can interact with MYC as measured by bimolecular fluorescence complementation. The shuttling is controlled by a Crm1-dependent nuclear export sequence and a central ARTD10 region that promotes nuclear localization. The latter lacks a classical nuclear localization sequence and does not promote full nuclear localization. Rather this non-conventional nuclear localization sequence results in an equal distribution of ARTD10 between the cytoplasmic and the nuclear compartments. ARTD10 forms discrete and dynamic bodies primarily in the cytoplasm but also in the nucleus. These contain poly-ubiquitin and co-localize in part with structures containing the poly-ubiquitin receptor p62/SQSTM1. The co-localization depends on the ubiquitin-associated domain of p62, which mediates interaction with poly-ubiquitin.ConclusionsOur findings demonstrate that ARTD10 is a highly dynamic protein. It shuttles between the nuclear and cytosolic compartments dependent on a classical nuclear export sequence and a domain that mediates nuclear uptake. Moreover ARTD10 forms discrete bodies that exchange subunits rapidly. These bodies associate at least in part with the poly-ubiquitin receptor p62. Because this protein is involved in the uptake of cargo into autophagosomes, our results suggest a link between the formation of ARTD10 bodies and autophagy.Lay abstractPost-translational modifications refer to changes in the chemical appearance of proteins and occur, as the name implies, after proteins have been synthesized. These modifications frequently affect the behavior of proteins, including alterations in their activity or their subcellular localization. One of these modifications is the addition of ADP-ribose to a substrate from the cofactor NAD+. The enzymes responsible for this reaction are ADP-ribosyltransferases (ARTDs or previously named PARPs). Presently we know very little about the role of mono-ADP-ribosylation of proteins that occurs in cells. We identified ARTD10, a mono-ADP-ribosyltransferase, as an interaction partner of the oncoprotein MYC. In this study we have analyzed how ARTD10 moves within a cell. By using different live-cell imaging technologies that allow us to follow the position of ARTD10 molecules over time, we found that ARTD10 shuttles constantly in and out of the nucleus. In the cytosol ARTD10 forms distinct structures or bodies that themselves are moving within the cell and that exchange ARTD10 subunits rapidly. We have identified the regions within ARTD10 that are required for these movements. Moreover we defined these bodies as structures that interact with p62. This protein is known to play a role in bringing other proteins to a structure referred to as the autophagosome, which is involved in eliminating debris in cells. Thus our work suggests that ARTD10 might be involved in and is regulated by ADP-riboslyation autophagosomal processes.


FEBS Journal | 2013

Caspase-dependent cleavage of the mono-ADP-ribosyltransferase ARTD10 interferes with its pro- apoptotic function

Nicolas Herzog; Jörg Hartkamp; Patricia Verheugd; Fabian Treude; Alexandra H. Forst; Karla L. H. Feijs; Barbara E. Lippok; Elisabeth Kremmer; Henning Kleine; Bernhard Lüscher

ADP‐ribosylation is a post‐translational modification that regulates various physiological processes, including DNA damage repair, gene transcription and signal transduction. Intracellular ADP‐ribosyltransferases (ARTDs or PARPs) modify their substrates either by poly‐ or mono‐ADP‐ribosylation. Previously we identified ARTD10 (formerly PARP10) as a mono‐ADP‐ribosyltransferase, and observed that exogenous ARTD10 but not ARTD10‐G888W, a catalytically inactive mutant, interferes with cell proliferation. To expand on this observation, we established cell lines with inducible ARTD10 or ARTD10‐G888W. Consistent with our previous findings, induction of the wild‐type protein but not the mutant inhibited cell proliferation, primarily by inducing apoptosis. During apoptosis, ARTD10 itself was targeted by caspases. We mapped the major cleavage site at EIAMD406↓S, a sequence that was preferentially recognized by caspase–6. Caspase‐dependent cleavage inhibited the pro‐apoptotic activity of ARTD10, as ARTD10(1–406) and ARTD10(407–1025), either alone or together, were unable to induce apoptosis, despite catalytic activity of the latter. Deletion of the N–terminal RNA recognition motif in ARTD10(257–1025) also resulted in loss of pro‐apoptotic activity. Thus our findings indicate that the RNA recognition motif contributes to the pro‐apoptotic effect, together with the catalytic domain. We suggest that these two domains must be physically linked to stimulate apoptosis, possibly targeting ARTD10 through the RNA recognition motif to specific substrates that control cell death. Moreover, we established that knockdown of ARTD10 reduced apoptosis in response to DNA‐damaging agents. Together, these findings indicate that ARTD10 is involved in the regulation of apoptosis, and that, once apoptosis is activated, ARTD10 is cleaved as part of negative feedback regulation.


European Journal of Pharmaceutical Sciences | 2013

Activity-based assay for human mono-ADP-ribosyltransferases ARTD7/PARP15 and ARTD10/PARP10 aimed at screening and profiling inhibitors.

Harikanth Venkannagari; Adyary Fallarero; Karla L. H. Feijs; Bernhard Lüscher; Lari Lehtiö

Poly(ADP-ribose) polymerases (PARPs) or diphtheria toxin like ADP-ribosyl transferases (ARTDs) are enzymes that catalyze the covalent modification of proteins by attachment of ADP-ribose units to the target amino acid residues or to the growing chain of ADP-ribose. A subclass of the ARTD superfamily consists of mono-ADP-ribosyl transferases that are thought to modify themselves and other substrate proteins by covalently adding only a single ADP-ribose moiety to the target. Many of the ARTD enzymes are either established or potential drug targets and a functional activity assay for them will be a valuable tool to identify selective inhibitors for each enzyme. Existing assays are not directly applicable for screening of inhibitors due to the different nature of the reaction and different target molecules. We modified and applied a fluorescence-based assay previously described for PARP1/ARTD1 and tankyrase/ARTD5 for screening of PARP10/ARTD10 and PARP15/ARTD7 inhibitors. The assay measures the amount of NAD(+) present after chemically converting it to a fluorescent analog. We demonstrate that by using an excess of a recombinant acceptor protein the performance of the activity-based assay is excellent for screening of compound libraries. The assay is homogenous and cost effective, making it possible to test relatively large compound libraries. This method can be used to screen inhibitors of mono-ARTDs and profile inhibitors of the enzyme class. The assay was optimized for ARTD10 and ARTD7, but it can be directly applied to other mono-ARTDs of the ARTD superfamily. Profiling of known ARTD inhibitors against ARTD10 and ARTD7 in a validatory screening identified the best inhibitors with submicromolar potencies. Only few of the tested ARTD inhibitors were potent, implicating that there is a need to screen new compound scaffolds. This is needed to create small molecules that could serve as biological probes and potential starting points for drug discovery projects against mono-ARTDs.


Nature Reviews Molecular Cell Biology | 2013

Macrodomain-containing proteins: regulating new intracellular functions of mono(ADP-ribosyl)ation

Karla L. H. Feijs; Alexandra H. Forst; Patricia Verheugd; Bernhard Lüscher


Structure | 2013

Recognition of Mono-ADP-Ribosylated ARTD10 Substrates by ARTD8 Macrodomains.

Alexandra H. Forst; Tobias Karlberg; Nicolas Herzog; Ann-Gerd Thorsell; Annika Gross; Karla L. H. Feijs; Patricia Verheugd; Petri Kursula; Bianca Nijmeijer; Elisabeth Kremmer; Henning Kleine; Andreas G. Ladurner; Herwig Schüler; Bernhard Lüscher


Scientific Reports | 2018

Nucleolar-nucleoplasmic shuttling of TARG1 and its control by DNA damage-induced poly-ADP-ribosylation and by nucleolar transcription

Mareike Bütepage; Christian Preisinger; Alexander von Kriegsheim; Anja Scheufen; Eva Lausberg; Jinyu Li; Ferdinand Kappes; Regina Feederle; Sabrina Ernst; Laura Eckei; Sarah Krieg; Gerhard Müller-Newen; Giulia Rossetti; Karla L. H. Feijs; Patricia Verheugd; Bernhard Lüscher

Collaboration


Dive into the Karla L. H. Feijs's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Elisabeth Kremmer

German Center for Neurodegenerative Diseases

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge