42
1 Chemical probe rescues cells from ARTD10/PARP10 induced apoptosis and sensitizes cancer cells to DNA damage Harikanth Venkannagari 1 , Patricia Verheugd 2 , Jarkko Koivunen 1 , Teemu Haikarainen 1 , Ezeogo Obaji 1 , Yashwanth Ashok 1 , Mohit Narwal 1 , Taina Pihlajaniemi 1 , Bernhard Lüscher 2 & Lari Lehtiö 1 * 1 Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu, Oulu, 90014, Finland 2 Institute of Biochemistry and Molecular Biology, RWTH Aachen University, Aachen, 52074,Germany *Corresponding author: Lari Lehtiö ([email protected])

Chemical probe rescues cells from ARTD10/PARP10 induced ...jultika.oulu.fi/files/nbnfi-fe201701181176.pdfARTD10 regulates NF- B signaling, where both its ADP-ribosylation activity

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Chemical probe rescues cells from ARTD10/PARP10 induced ...jultika.oulu.fi/files/nbnfi-fe201701181176.pdfARTD10 regulates NF- B signaling, where both its ADP-ribosylation activity

1

Chemical probe rescues cells from ARTD10/PARP10 induced

apoptosis and sensitizes cancer cells to DNA damage

Harikanth Venkannagari1, Patricia Verheugd

2, Jarkko Koivunen

1, Teemu Haikarainen

1,

Ezeogo Obaji1, Yashwanth Ashok

1, Mohit Narwal

1, Taina Pihlajaniemi

1, Bernhard Lüscher

2

& Lari Lehtiö1*

1Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu, Oulu, 90014,

Finland

2Institute of Biochemistry and Molecular Biology, RWTH Aachen University, Aachen, 52074,Germany

*Corresponding author: Lari Lehtiö ([email protected])

Page 2: Chemical probe rescues cells from ARTD10/PARP10 induced ...jultika.oulu.fi/files/nbnfi-fe201701181176.pdfARTD10 regulates NF- B signaling, where both its ADP-ribosylation activity

2

SUMMARY

Members of the human diphtheria toxin-likeADP-ribosyltransferase (ARTD or PARP) family play

important roles in regulating biological activities by mediating either a mono-ADP-ribosylation

MARylation) of a substrate or a poly-ADP-ribosylation (PARylation). ARTD10/PARP10 belongs

to the MARylating ARTDs (mARTDs) subfamily, and plays important roles in biological processes

that range from cellular signaling, DNA repair, and cell proliferation to immune response. Despite

their biological and disease relevance, no selective inhibitors for mARTDs are available. Here we

describe a small-molecule ARTD10 inhibitor, OUL35, a selective and potent inhibitor for this

enzyme. We characterize its selectivity profile, model its binding, and demonstrate activity in HeLa

cells where OUL35 rescued cells from ARTD10 induced cell death. Using OUL35 as a cell biology

tool we show that ARTD10 inhibition sensitizes the cells to the hydroxyurea-induced genotoxic

stress. Our study supports the proposed role of ARTD10 in DNA-damage repair and

provides a tool compound for selective inhibition of ARTD10-mediated MARylation.

Page 3: Chemical probe rescues cells from ARTD10/PARP10 induced ...jultika.oulu.fi/files/nbnfi-fe201701181176.pdfARTD10 regulates NF- B signaling, where both its ADP-ribosylation activity

3

INTRODUCTION

The human diphtheria toxin-like ADP-ribosyltransferase (ARTD or PARP) family includes 17

members that share a conserved catalytic domain responsible for ADP-ribosylation of substrate

proteins (Amé et al., 2004; Hottiger et al., 2010; Otto et al., 2005). While some ARTDs modify

substrates by transferring iteratively multiple ADP-ribose units resulting in poly-ADP-ribosylation

(PARylation), most ARTDs mono-ADP-ribosylate (MARylate) their substrates (Kleine et al.,

2008). ARTDs function in DNA damage repair (Malanga and Althaus, 2005; Nicolae et al., 2014,

2015), the unfolded protein response (Jwa and Chang, 2012), apoptosis (Herzog et al., 2013; Koh et

al., 2005), heat shock (Petesch and Lis, 2008), cellular signaling (Feijs et al., 2013a; Verheugd et

al., 2013), cell division (Chang et al., 2004, 2005, 2009; Ha et al., 2012), as well as transcription

and chromatin regulation (Hassa and Hottiger, 2008; Schreiber et al., 2006). PARylating ARTDs

(pARTDs; ARTD1- 6), most prominently ARTD1, have been the focus of cancer related research

during the past two decades. Due to their sequence and structural similarity ARTD3 and ARTD4

are classified here as pARTDs, although their ability to form PAR chains is not well established and

they have also been postulated to be mARTDs (Kickhoefer et al., 1999; Loseva et al., 2010; Vyas et

al., 2014). An ARTD inhibitor, Olaparib, has been approved to treat ovarian cancer with BRCA

mutations. However, Olaparib as well as several other inhibitors developed for ARTD1 are not

specific, but they inhibit several ARTD enzymes (Wahlberg et al., 2012), which indicates that

further studies are required to evaluate the contribution of different ARTDs to the phenotypes

observed. This is relevant because recent studies have linked MARylating ARTDs (mARTDs) to

cancer biology (Vyas and Chang, 2014). Notably, ARTD10/PARP10 has been suggested to be a

potential drug target in cancer (Nicolae et al., 2014). Together these findings delineate the need to

understand the cellular roles of the different PARylating and MARylating ARTDs and to develop

tools to selectively modulate individual family members for functional studies and therapeutic

approaches.

Page 4: Chemical probe rescues cells from ARTD10/PARP10 induced ...jultika.oulu.fi/files/nbnfi-fe201701181176.pdfARTD10 regulates NF- B signaling, where both its ADP-ribosylation activity

4

MARylation, a covalent reversible post-translational modification of proteins, is associated with

various cellular processes (Bütepage et al., 2015; Feijs et al., 2013b). ARTD10 was first identified

as a binding partner of the oncoprotein c-Myc and it was the first member of the ARTD family to be

fully characterized as an mARTD (Kleine et al., 2008). ARTD10 has a conserved C-terminal ART

domain responsible for its enzymatic activity (Hottiger et al., 2010; Kleine et al., 2008; Otto et al.,

2005; Yu et al., 2005). In vitro screening of more than 8000 proteins identified 78 substrates for

ARTD10 (Feijs et al., 2013a), the majority of these being kinases. ADP-ribosylation of GSK3-β by

ARTD10 negatively regulates the kinase activity of GSK3-β in vitro and knockdown of ARTD10

increased the kinase activity of GSK3-β in cells (Feijs et al., 2013a).

In addition to the catalytic domain, which is homologous to other ARTDs, ARTD10 has a RNA

recognition motif at the N-terminus, a nuclear export sequence, two ubiquitin interaction motifs

(UIMs), a glycine rich region and a segment that mediates nuclear translocation. ARTD10 shuttles

between the nuclear and cytoplasmic compartments, regulates cell proliferation and is ubiquitously

expressed in essentially all tissues (Kaufmann et al., 2015). The UIMs of ARTD10 have been

shown to specifically interact with K63-linked poly-ubiquitin (K63-pUb) chains (Verheugd et al.,

2013). K63-pUB chains function as scaffolds in various signaling processes including the NF-B

signal transduction pathway (Chen and Sun, 2009; Fan et al., 2011; Xu et al., 2009). Indeed

ARTD10 regulates NF-B signaling, where both its ADP-ribosylation activity and its interaction

with K63-pUb by the UIMs are needed (Verheugd et al., 2013). ARTD10 was suggested to

MARylate NEMO and also physically prevent its modification by K63-pUb. NEMO is required to

assemble the IKK complex, which is critical for upstream signaling that activates IKKs and

modifies IB inhibitors (Oeckinghaus et al., 2011). Moreover, it was observed that a catalytically

Page 5: Chemical probe rescues cells from ARTD10/PARP10 induced ...jultika.oulu.fi/files/nbnfi-fe201701181176.pdfARTD10 regulates NF- B signaling, where both its ADP-ribosylation activity

5

inactive ARTD10 also efficiently inhibited the signaling cascade by physically interacting with the

K63-pUb via the UIMs and blocking the interaction of NEMO with K63-pUb.

A recent study has shown that ARTD10 interacts with PCNA and actively participates in S phase

DNA damage repair by responding to replication fork stalling and activating translesion DNA

synthesis (Nicolae et al., 2014). The exact mechanism of how ARTD10 is involved in this process

is not clearly understood, but catalytic activity is needed for its DNA damage related functions

(Nicolae et al., 2014).

In the present study we describe a selective small molecule inhibitor for ARTD10. This compound

is a useful chemical probe to study MARylation by ARTD10 and a novel chemical scaffold that

could also be utilized in the development of potent and selective probes for other mARTDs.

RESULTS

Discovery of a potent and selective ARTD10 inhibitor

Earlier we described a robust activity-based assay for ARTD10, which was validated for screening

of compound libraries (Venkannagari et al., 2013). Here we applied this assay for the screening of

compound libraries from the open chemical repository of the National Cancer Institute (NCI). In

total 2638 compounds were tested. The hits were re-tested to exclude false positives and a total of

19 hits were identified. Dose response measurements revealed two compounds with sub-micromolar

potencies: OUL47 (resorufin, 7-hydroxy-3H-phenoxazin-3-one, NSC12097) (IC50 = 482 nM) and

OUL35 (4-[(4-carbamoylcyclohexyl)oxy]cyclohexane-1-carboxamide, NSC39047) (IC50 = 329 nM)

(Figures 1A and 1B). OUL47 is widely used as an indicator of cell viability in assays where

resazurin is reduced to the highly fluorescent OUL47 by living cells (Jouanneau et al., 2014).

Notably, there were no previous publications or activity data on the OUL35 compound. Our initial

results indicated that OUL47 affected cell viability, and the toxicity was confirmed by tests with

Page 6: Chemical probe rescues cells from ARTD10/PARP10 induced ...jultika.oulu.fi/files/nbnfi-fe201701181176.pdfARTD10 regulates NF- B signaling, where both its ADP-ribosylation activity

6

HeLa cells (Figures S1A-S1G). Treatment of HeLa cells with 10 µM of OUL35 or OUL47 showed

that OUL35 had no cytotoxic effect, whereas OUL47 inhibited cell proliferation. Hence, OUL47

was excluded from further experiments and OUL35 was singled out as the most promising non-

toxic inhibitor of ARTD10 found in the screening. Furthermore it is the most potent inhibitor of any

human mARTD reported to date.

Profiling and selectivity

There are 17 ARTD enzymes in the human ARTD superfamily, which all contain a homologous

catalytic domain. Therefore it was essential to verify the selectivity of OUL35 for ARTD10. The

non-selectivity of the “PARP inhibitors” is a known issue and some efforts have recently been

devoted to understanding the inhibitor profile (Haikarainen et al., 2014; Wahlberg et al., 2012). We

used our established assay method (Narwal et al., 2012; Venkannagari et al., 2013) to profile the

selectivity of OUL35 against the ARTDs available in our laboratory (Table 1). Profiling

experiments revealed that OUL35, despite its small molecular weight and simple structure, was

highly selective towards ARTD10 over the other enzymes of the family (Table 1). OUL35 also

inhibited ARTD8 (23.4 µM), ARTD4 (22.6 µM), and ARTD15 (4.17 µM) but with modest potency

(Table 1).

Selectivity was further validated by differential scanning fluorimetry (DSF) to test for binding of

the compound. This activity independent method also allowed us to include enzymatically inactive

ARTD9 and ARTD13 (Karlberg et al., 2015) in the profiling. DSF verified binding of OUL35 to

the ARTD10 catalytic domain and its selectivity over other ARTDs. OUL35 stabilized the catalytic

domains of ARTD10, ARTD8 & ARTD15 as expected based on the enzymatic assays (Table 1).

Page 7: Chemical probe rescues cells from ARTD10/PARP10 induced ...jultika.oulu.fi/files/nbnfi-fe201701181176.pdfARTD10 regulates NF- B signaling, where both its ADP-ribosylation activity

7

OUL35 contains a nicotinamide mimicking motif like most of the ARTD inhibitors reported to date

(Wahlberg et al., 2012) (Figure 1A). It consists of two symmetrical benzamide motifs that could

compete with the binding of NAD+ in the nicotinamide binding site. Based on this hypothesis we

designed two small modifications to the compound yielding OUL77 (4-{[4-

(dimethylcarbamoyl)cyclohexyl]oxy}-N,N-dimethylcyclohexane-1-carboxamide) and OUL78 (N-

methyl-4-{[4-(methylcarbamoyl)cyclohexyl]oxy}cyclohexane-1-carboxamide), which we expected

to be inactive and useful as control compounds when evaluating OUL35 in biological systems

(Figure 1A). Neither of the methylated analogs at concentrations of 1 µM or 10 µM inhibited

ARTD10 supporting our hypothesis (Figure S2). Notably, an inactive mutant ARTD10-G888W

designed to block the nicotinamide binding site (Kleine et al., 2008) was not stabilized by OUL35

in the DSF experiment in agreement with our hypothesis of nicotinamide competition (Table 1).

Based on the biochemical and biophysical analysis OUL35 is a potentially useful tool to study

ARTD10 functions in cells.

Structural basis for the inhibition and selectivity

To understand the structural basis for inhibition and selectivity of OUL35 towards ARTD10 we

performed docking studies using the available crystal structure of an ARTD10 mutant (PDB id.

3HKV). There are no NAD+ complex structures with ARTDs available and our understanding of

the substrate binding is based on the NAD+ binding mode in the diphtheria toxin (Bell and

Eisenberg, 1996) (PDB id. 1TOX) and on NAD+ mimicking inhibitors (Haikarainen et al., 2014).

The crystal structure of ARTD10 contains the small general ARTD inhibitor 3-aminobenzamide

(3AB), which binds to the nicotinamide binding site of ARTDs (Figures 1A and S3). The chemical

structure of OUL35 suggests that the benzamide moiety would bind to the nicotinamide pocket in a

manner similar to the nicotinamide of the substrate NAD+ and the small general ARTD inhibitor

3AB (Figures 1A and S3). Based on this assumption, we expect that OUL35 extends from the

Page 8: Chemical probe rescues cells from ARTD10/PARP10 induced ...jultika.oulu.fi/files/nbnfi-fe201701181176.pdfARTD10 regulates NF- B signaling, where both its ADP-ribosylation activity

8

pocket towards the acceptor site where the ADP-ribosylation target proteins bind. The acceptor site

is deduced from the NAD+ analog found partially visible in the chicken ARTD1 structure (Ruf et

al., 1998) (Figure S3C). Docking studies revealed that the best ranked binding poses indeed

overlap with 3AB found in the co-crystal structure and that OUL35 extends towards the acceptor

site (Figures 2A and S3). Notably, all the nicotinamide mimicking ARTD inhibitors reported so far

extend along the NAD+ binding cleft towards the adenosine binding pocket and hence the docking

model indicates that OUL35 is a unique ARTD inhibitor (Ferraris, 2010). In the docking model,

OUL35 makes hydrogen bonds to the amide and carbonyl of Gly888 and to the side chain hydroxyl

of Ser927 (Figure 2A). OUL35 stacks between Tyr919 and Tyr932 like 3AB in the crystal structure

(Figure S3). In the model the 4-oxo-benzamide moiety extends towards the acceptor site and

interacts with hydrophobic residues Ile987 and Leu926. It also makes a putative nonoptimal

hydrogen bond with Arg931. We tested this by mutating Arg931 to alanine, but the mutation did

not affect the IC50 of OUL35 towards ARTD10 indicating that the interaction does not contribute

significantly to the binding affinity (Table 1).

In contrast to the nicotinamide site, the residues surrounding the acceptor site are not well

conserved throughout the ARTD family (Figures 2A-2F and S4A-S4E). OUL35 did not inhibit

ARTD1 (Table 1) and comparison of the binding pose in ARTD10 with the crystal structure of

ARTD1 suggested that the catalytic glutamate (Glu988), present in all PARylating enzymes,

overlaps with the compound. Furthermore Lys903 in ARTD1 also overlaps with the inhibitor

(Figure 2B). The unfavorable interactions with the catalytic glutamate of the so called H-Y-E motif

(Hottiger et al., 2010; Otto et al., 2005) and with Lys903 most likely explain why OUL35 does not

inhibit ARTD1, 2, 3, 5, and 6 (Figures 2B, 2C and S4A-S4C).

Page 9: Chemical probe rescues cells from ARTD10/PARP10 induced ...jultika.oulu.fi/files/nbnfi-fe201701181176.pdfARTD10 regulates NF- B signaling, where both its ADP-ribosylation activity

9

The catalytic glutamate on the β-strand outside the binding pocket (Glu988 in ARTD1) and the

lysine on the α-helix (Lys903 in ARTD1) on the other edge of the binding cavity are highly

conserved in pARTDs. ARTD4, initially proposed to have PARylation activity based on the

catalytically relevant H-Y-E signature, appears to be a mARTD (Bütepage et al., 2015), in which

the lysine is replaced by Thr484 (Figure S5). This difference could explain the modest potency of

OUL35 towards ARTD4 (Table 1). In ARTDs 7-17, the catalytic glutamate in H-Y-E is replaced

by a hydrophobic residue, usually leucine, isoleucine or valine, but is replaced by a threonine in

ARTD9 and a tyrosine in ARTD15 (Figure S5). The lysine on the α-helix in ARTDs 1, 2, 3, 5, and

6 (Figures 2B, 2C and S4A-S4C) is replaced by a leucine in ARTD10 and ARTD15 (Figures 2A

and 2E), isoleucine in ARTD16 and ARTD17 and tyrosine in rest of the mARTDs. In ARTD7 and

ARTD12 this tyrosine (Figures 2F and S4D) might prevent binding of OUL35. The tyrosine in

ARTD8 is oriented further away from the active site in the crystal structure in comparison to

ARTD7 and ARTD12 (Figure 2D) and the observed difference may explain the modest potency of

OUL35 towards ARTD8 (Table 1). To verify the effect of tyrosine on the selectivity, we studied an

ARTD10 tyrosine mutant (L926Y). While the activity of the mutant in the enzymatic assay was

only slightly lower than the wild type, the mutation decreased the potency of OUL35 by more than

10-fold (Table 1). On the other hand gain-of-binding mutations made for ARTD7 (Y576L) and

ARTD8 (Y1640L) increased the potency of the compound towards these enzymes (Table 1).

ARTD13 and ARTD9 are catalytically inactive and lack a glycine residue (Gly888 in ARTD10),

which is highly conserved in other ARTDs (Figure S5). ARTD13 also contains a tyrosine (Tyr826)

in the α-helix (Figure S4E), which together with the reported closed binding site could explain why

OUL35 does not bind to ARTD13 (Karlberg et al., 2015). OUL35 showed modest potency against

ARTD15 (Table 1). Unlike the other ARTDs, a serine residue in the active site (e.g. Ser927 in

ARTD10) is replaced by Ala190 in ARTD15 (Figure 2E). The catalytic glutamate found in

Page 10: Chemical probe rescues cells from ARTD10/PARP10 induced ...jultika.oulu.fi/files/nbnfi-fe201701181176.pdfARTD10 regulates NF- B signaling, where both its ADP-ribosylation activity

10

pARTDs is replaced with a tyrosine in ARTD15 (Tyr224) (Karlberg et al., 2012) (Figures 2E and

S5). While the benzamide would lose a hydrogen bond due to the absence of serine in ARTD15, the

possible stacking of the 4-oxo-benzamide moiety with the Tyr224 is likely to contribute to the

observed modest potency (Table 1, Figure 2E). In agreement with this a S927A ARTD10 mutant

has significantly lower affinity to OUL35 (Table 1). Together the docking and mutational studies

provide explanations for the observed differences in potency of OUL35 towards different ARTD

family members.

OUL35 rescues HeLa cells from ARTD10-induced cell death

Over-expression of wild-type ARTD10 but not the catalytically inactive mutant ARTD10-G888W

leads to cell death (Herzog et al., 2013). Colony formation assays were performed to assess whether

OUL35 can enter cells and inhibit ARTD10 and whether the toxic effect of ARTD10 could be

negated by inhibiting its ADP-ribosylation activity. As expected, overexpression of wild-type

ARTD10 but not ARTD10-G888W strongly inhibited cell proliferation (Figures 3A and 3B). The

treatment of HeLa cells overexpressing ARTD10 with OUL35 resulted in a complete rescue of cell

proliferation. Notably the inactive analogs OUL77 and OUL78 were ineffective and did not

facilitate colony formation (Figures 3A and 3B). Moreover none of the compounds showed any

toxicity in the control cells. Together these findings provide evidence that OUL35 enters cells and

inhibits the catalytic activity of ARTD10 in a dose dependent manner with an IC50 of 1.35 µM

(Figure 3C). To further substantiate this we performed cellular thermal shift assays (CETSA)

showing that endogenous ARTD10 in U2OS cells is stabilized by OUL35 (Figure 3D). FACS

analysis was performed to further evaluate the results of the colony formation assay.

Overexpression of ARTD10 induced an increase in cell death compared to control cells (Figures

4A-4D). Again cells were rescued from ARTD10 induced cell death by OUL35 while the inactive

analogs were ineffective.

Page 11: Chemical probe rescues cells from ARTD10/PARP10 induced ...jultika.oulu.fi/files/nbnfi-fe201701181176.pdfARTD10 regulates NF- B signaling, where both its ADP-ribosylation activity

11

Hydroxyurea sensitivity confirms the role of ARTD10 in genotoxic stress response

Hydroxyurea (HU) inhibits ribonucleotide reductase and thus interferes with deoxyribonucleotide

synthesis. This results in inhibition of S phase and causes stalled replication forks (Koç et al.,

2004). ARTD10 knockdown cells are more sensitive to HU-induced DNA damage as the catalytic

activity is necessary for its DNA repair functions (Nicolae et al., 2014). We investigated whether

we could reproduce this effect in HeLa cells using OUL35. HeLa cells treated with HU were more

sensitive to DNA damage in the presence of 5 µM OUL35 (Figures 5A and 5B). At 1 µM OUL35,

a concentration below the cellular IC50 value, no sensitization of the cells was measurable (Figure

5C). Similarly, cells treated with the inactive analog OUL77 grew comparably to control cells

(Figure 5D). Thus using a specific chemical probe targeting ARTD10 we were able to sensitize the

cells to the hydroxyurea induced genotoxic stress.

DISCUSSION

Growing interest in the development of ARTD inhibitors culminated in the recent approval of the

ARTD1-4 inhibitor Olaparib (Lynparza) for the treatment of ovarian cancer. The less studied

members of the superfamily, mARTDs, have recently attracted attention as potential new drug

targets (Andersson et al., 2012; Ekblad et al., 2015; Morgan et al., 2015; Wahlberg et al., 2012). It

is clear that the existing ARTD inhibitors are not selective and, although they inhibit certain

mARTDs, they can not be used to evaluate the cellular effects of inhibiting these enzymes

(Wahlberg et al., 2012). Intracellular MARylation plays multiple roles in cancer biology as it is

involved in cellular signaling events including stress and immune responses (Bütepage et al., 2015;

Nicolae et al., 2014; Scarpa et al., 2013; Verheugd et al., 2013). Hence there is a need to identify

new inhibitors to be used as research tools and to evaluate mARTDs as potential drug targets.

Page 12: Chemical probe rescues cells from ARTD10/PARP10 induced ...jultika.oulu.fi/files/nbnfi-fe201701181176.pdfARTD10 regulates NF- B signaling, where both its ADP-ribosylation activity

12

Here, the small molecule OUL35 was identified as a potent and selective inhibitor of one of the

mARTDs - ARTD10. The compound is able to enter cultured cells and inhibit ARTD10-dependent

cellular processes. Structural analysis suggested the basis of the selectivity over the other ARTD

enzymes and the docking model indicated that OUL35 would be the first ARTD/PARP inhibitor

extending towards the acceptor site. While OUL35 is selective towards ARTD10 over the other

tested enzymes it can also be used in the design inhibitors for other mARTDs due to the structural

similarities of the binding site. Notably, the identified mARTDs that show weak affinity to OUL35

are also suggested to be involved in cancer linked processes. ARTD8 plays a role in the survival of

cancerous multiple myeloma cells (Barbarulo et al., 2013) and promotes DNA damage repair

(Nicolae et al., 2015), while ARTD15 is a regulator of the unfolded protein response (Jwa and

Chang, 2012).

OUL35 is a simple low molecular weight inhibitor, which is available through chemical vendors.

OUL35 was not toxic to control cells in our experiments, but it efficiently rescued cells

overexpressing ARTD10 and reproduced the effects seen by ARTD10 knockdown using RNAi

(Herzog et al., 2013; Nicolae et al., 2014). ARTD10 shuttles between cytoplasm and nucleus

(Kleine et al., 2012) and the results provide additional evidence for the role of ARTD10 in S phase

DNA damage repair. OUL35 will aid further studies to assess the roles of ARTD10 in different

processes. In vitro screening identified 78 target proteins for MARylation by ARTD10 of which the

majority were kinases (Feijs et al., 2013a), highlighting the need to understand its roles in various

physiological and stress/disease related conditions.

SIGNIFICANCE

ARTD10 is a cellular mono-ADP-ribosyltransferase, which shuttles between the nucleus and the

cytosol and regulates signaling pathways that include inflammation and S phase DNA damage

Page 13: Chemical probe rescues cells from ARTD10/PARP10 induced ...jultika.oulu.fi/files/nbnfi-fe201701181176.pdfARTD10 regulates NF- B signaling, where both its ADP-ribosylation activity

13

repair. The study reports a chemical probe, OUL35, which was discovered during screening of

random chemical libraries. The compound is potent and selective towards ARTD10, it is non-toxic,

and inhibits ARTD10 enzymatic activity in cells. We also provide initial results on applying the

chemical probe to study the potential therapeutic applications of ARTD10 inhibition as OUL35

sensitizes cancer cells to a DNA damaging agent in line with published ARTD10 knockdown

studies. OUL35 is the first selective inhibitor for a human mono-ADP-ribosyltransferase and the

presented structural analysis can therefore be used as a basis for further drug discovery efforts.

EXPERIMENTAL PROCEDURES

Cloning and protein expression

All the proteins used in the study were cloned into one of the following four plasmids:

pNIC28-Bsa4 with a N-terminal His-tag, pNIC-CH with a C-terminal His-tag, pNH-TrxT with a N-

terminal His-tag and a thioredoxin fusion tag, and pNC-ZB with a N-terminal His-tag and a Z-basic

fusion tag. Details for the amino acid composition of each construct are described in the Tables S1

and S2.

All the proteins were expressed and purified with hexa-histidine fusion tags from E. coli Rosetta2

(DE3) cells. Cells were grown in auto-induction TB media supplemented with trace elements

(Formedium, UK) and further supplemented with 8 g/l glycerol. Kanamycin (50 µg/ml) and

chloramphenicol (34 µg/ml) were used as selection markers. 5 ml pre-cultures were used to

inoculate 1 l cultures in 5 l flasks. After inoculation the cells were grown at 37oC with shaking at

250 rpm until the OD600 reached between 1.0 and 1.5 after which the temperature was reduced to

18oC and the cultures were allowed to grow overnight (14-16 h). The cells were harvested by

centrifugation (5500×g, 20 min, and 4oC). The cell pellets were re-suspended in lysis buffer (100

mM HEPES, pH 7.5, 500 mM NaCl, 10% Glycerol, 10 mM Imidazole, 0.5 mM TCEP) at 1.5 ml

per 1 gram of pellet and stored at -20 oC.

Page 14: Chemical probe rescues cells from ARTD10/PARP10 induced ...jultika.oulu.fi/files/nbnfi-fe201701181176.pdfARTD10 regulates NF- B signaling, where both its ADP-ribosylation activity

14

Protein purification

The cell pellets were thawed in warm water and supplemented with 20 µg/ml DNAase (Roche), 0.1

mM pefabloc (Sigma-Aldrich), and 0.25 mg lysozyme (Fluka Analytical). The pellets were

incubated on ice for 5 min before sonication (Branson 450D sonifier) with 50% amplitude for two

minutes with five seconds on/off pulse using a ½ inch horn. After sonication, the cell lysates were

clarified by centrifugation at 35,000×g, 4oC for 20 min. The supernatant was filtered through a 0.45

µm filter syringe. Proteins were purified using affinity chromatography (Ni-NTA) and size-

exclusion chromatography. DNA binding ARTDs were further purified on a heparin column before

performing the size-exclusion chromatography. In some cases the 6x his-tag was cleaved from the

fusion proteins (ARTD12, ARTD15). The protocol described above summarizes the general

purification method followed for all the proteins used in this study. More details about constructs

used in different assays are given in the Tables S1 and S2.

Screening and validation

Screening of the compound libraries against ARTD10 was performed based on our previously

validated activity-based fluorescent assay (Venkannagari et al., 2013). Compound libraries were

obtained from the National Cancer Institute (NCI) repository (1 mM mechanistic diversity set, 10

mM diversity set III, 10 mM approved oncology drugs set III, and the natural products set II). In

total 2636 compounds were tested. The initial screening was performed at 1 µM for the mechanistic

diversity set and 10 µM for diversity set III, approved oncology drugs set III, and natural products

set II libraries. Phenanthridinone (Alexis Biochemicals), which is a known ARTD10 inhibitor

(Venkannagari et al., 2013), was used as a positive control in all the assay plates. The hits from the

initial screening were rescreened and dose response curves were measured for the confirmed hit

compounds. The hit compound OUL35 from the NCI compound library and the inactive analogs

Page 15: Chemical probe rescues cells from ARTD10/PARP10 induced ...jultika.oulu.fi/files/nbnfi-fe201701181176.pdfARTD10 regulates NF- B signaling, where both its ADP-ribosylation activity

15

OUL77 and OUL78 were later purchased from Chemdiv (Catalog numbers 0384-0062, 0384-0009

and 0384-0063, respectively).

Activity-based profiling

Profiling and dose response experiments were conducted as described previously (Narwal et al.,

2012; Venkannagari et al., 2013). As detailed in our earlier publication (Venkannagari et al., 2013)

some of the mARTDs are very sensitive to increasing concentrations of DMSO. In these cases we

tested a maximum concentration of 10 µM for compound OUL35. Details of the assay conditions

and expression constructs used for each ARTD are provided in Table S2.

Western blot based activity assay

To test that the mutations in ARTD10 (Table 1) do not affect its ability to modify the target protein

SRPK2, a western blot experiment was performed using biotinylated NAD+ as a substrate

(Trevigen, USA) (Figure S6). The enzymatic reaction contained 1 μM biotinylated NAD+ together

with 500 nM ARTD10 and 2 μM SRPK2 in 50 mM Tris pH 7.0. The reactions were carried out at

room temperature for 4 h, and they were stopped by adding 4× Laemmli buffer (Bio-Rad, USA) and

heating the mixture at 90 °C for 5 min. The samples were separated by SDS-PAGE and then

transferred onto a Trans-Blot® Turbo™ nitrocellulose transfer pack (Bio-Rad, USA). The

immobilized proteins on the membrane were blocked overnight at 4oC using 1% Casein in 1x TBS

(Bio-Rad, USA) before detecting the biotin ADP-ribosylated proteins with streptavidin conjugated

horseradish peroxidase (PerkinElmer, USA), which was diluted to 1:15,000.

Differential scanning fluorimetry

Differential scanning fluorimetry was performed with protein concentrations of 0.25 mg/ml. Details

of the expression constructs used for each ARTD are provided in Table S1. The concentration of

Page 16: Chemical probe rescues cells from ARTD10/PARP10 induced ...jultika.oulu.fi/files/nbnfi-fe201701181176.pdfARTD10 regulates NF- B signaling, where both its ADP-ribosylation activity

16

OUL35 was 100 µM. Control wells without the compound contained an equal amount of the

DMSO vehicle as the melting curves in the presence of OUL35. Sypro Orange (Life Technologies)

was used as the reporter dye with a final concentration of 20x or 5x. The experiment was performed

on a real time PCR machine (Applied Biosystems) with the temperature increasing from 21°C up to

90oC (70 cycles) with 1 degree increment per minute.

Docking of OUL35

GOLD (Jones et al., 1997) was used to analyze the binding mode of OUL35 to the catalytic domain

of ARTD10. The available ARTD10 crystal structure (3HKV) contains mutations, but they are

located outside the expected binding pocket and do not contribute to the binding of OUL35. The

binding pocket was defined with an 8 Å radius based on the nicotinamide mimicking 3AB present

in the crystal structure. Hydrogen atoms were added, ligands and water molecules removed, and the

program defaults were used with the ChemPLP scoring function. The top three binding poses

produced by the genetic algorithm were essentially the same indicating a likely solution in the

docking experiment. The poses also fitted our hypothesis of benzamide acting as a nicotinamide

mimetic.

Colony formation assay

Three hundred HeLa cells designed to express the wild-type ARTD10 and catalytically inactive

ARTD10 (G888W) in response to doxycycline were seeded in 6 cm well culture plates. The cells

were grown in DMEM culture media with 10% FCS, and the plates were incubated at 37oC with 5%

CO2. After 24 h of seeding, over expression of ARTD10 was induced by adding 500 ng/ml

doxycycline and the media were also supplemented with the tested compounds at 10 μM

concentrations. The cells were allowed to proliferate for the next 10 days, after which they were

stained with methylene blue to count the number of surviving colonies. The compounds and

Page 17: Chemical probe rescues cells from ARTD10/PARP10 induced ...jultika.oulu.fi/files/nbnfi-fe201701181176.pdfARTD10 regulates NF- B signaling, where both its ADP-ribosylation activity

17

doxycycline were replenished every 48 h while fresh media (DMEM supplemented with 10% FCS)

was added every four days. The IC50 value was calculated and fitted using 4-parameters with

GraphPad Prism 5.04.

Cellular Thermal Stability Assay

U2OS cells were used for the CETSA experiments (Jafari et al., 2014) as U2OS cells have

endogenous ARTD10 protein levels that are detectable by Western blotting. We established

conditions under which in control cell lysates ARTD10 precipitated upon heat treatment. For this

U2OS cells were lysed in a buffer containing 50 mM Tris, pH 7.5, 150 mM NaCl, 1 mM EDTA,

10% glycerol, 1% NP-40, 1 mM DTT, and protein inhibitor cocktail. One hundred µl aliquots were

supplemented with OUL35 or DMSO only and heat treated in a gradient from 54°C - 65.5°C. Then

the precipitated proteins were pelleted by centrifugation (in an Eppendorf centrifuge at 14.000 rpm,

4°C, 25 min). The cleared supernatants were separated by SDS-PAGE and ARTD10 was detected

by immunoblotting (the anti-ARTD10 antibody was generated against a GST-ARTD10(1-400)

fusion protein and purified). For the in cell analysis, U2OS cells were seeded on tissue culture

plates and subsequently incubated in the presence of DMSO or OUL35 for 12 hours. Cells were

lysed in the buffer described above and aliquots of the lysates treated and analyzed as described

before.

Fluorescence-activated cell sorting

10 cm culture plates were seeded with 200,000 HeLa cells expressing an inducible ARTD10-GFP

fusion protein in Dulbecco’s modified Eagle’s medium (DMEM) culture media with 10% fetal

bovine serum (FCS). The plates were incubated at 37oC with 5% CO2. After 24 h ARTD10-GFP

expression was induced by adding 500 ng/ml doxycycline after which the compounds were added.

The cells in the control plates were not induced with doxycycline. The concentration of the

Page 18: Chemical probe rescues cells from ARTD10/PARP10 induced ...jultika.oulu.fi/files/nbnfi-fe201701181176.pdfARTD10 regulates NF- B signaling, where both its ADP-ribosylation activity

18

compounds was 3 μM, which is approximately 10 times the IC50 value of OUL35 towards

ARTD10. The cells were analyzed after 24 h of induction. The compound did not show any toxicity

towards cells in the control plates. Apoptotic cells were detected by staining with Annexin V while

cells dying from necrosis were detected by propidium iodide.

DNA damage experiments

HeLa cells were grown in DMEM supplemented with 10% FCS (Lonza), 1x penicillin and

streptomycin (Sigma) at 37°C in a 5% CO2 atmosphere. 4000 cells/well were plated on 96-well

plates. The cells were allowed to attach for 6 hours before treatment with hydroxyurea combined

with 1 or 5 μM OUL35 or DMSO vehicle in serum free medium. The DMSO content was kept

below 0.05% in all experiments. The 96-well plates were placed in an IncuCyte ZOOM live cell

imaging system (Essen BioScience) to follow cell proliferation. The plates were scanned in the

IncuCyte at 2 hour intervals for 72 hours. Data were analyzed by the IncuCyte Zoom software

(Essen BioScience). The results are representative of three (cell proliferation curves) or five

independent experiments (72 h end points) each done in six replicates. Statistics were analyzed with

Graphpad Prism 5 software using a two-tailed, unpaired Student’s t-test. P values less than 0.05

were considered as significant (n.s = not statistically significant, *P < 0.05, **P < 0.01).

ACKNOWLEDGEMENTS

The research was funded by Biocenter Oulu, the Academy of Finland (287063 and 294085 to LL

and 266922 to TH), a Centre of Excellence Grant 2012–2017 of the Academy of Finland (284605 to

TP), the Sigrid Jusélius Foundation, the Jane and Aatos Erkko Foundation, the German Science

Foundation (DFG LU466/15-1 to BL) and the IZKF Aachen (SP01-2011 and O2-1-2014 to BL) of

the Medical School of the RWTH Aachen University. HV was supported by an EMBO short term

fellowship for his visit to the RWTH Aachen University to perform cell-based assays. We thank

Page 19: Chemical probe rescues cells from ARTD10/PARP10 induced ...jultika.oulu.fi/files/nbnfi-fe201701181176.pdfARTD10 regulates NF- B signaling, where both its ADP-ribosylation activity

19

Ekaterina Biterova for cloning the ARTD15 catalytic domain, Barbara E. Lippok for technical

assistance, Max Kaufmann for the ARTD10-GFP cell line, Justina Wozniak for the initial cell-

based assays and Virpi Glumoff for help with the FACS analysis. We also thank SGC, Stockholm

for some of the ARTD expression constructs.

AUTHOR CONTRIBUTION

HV: Measured and analyzed data and wrote the manuscript. PV: Designed and carried out some of

the cell-based assays and analyzed the data. TH: Performed ARTD profiling and some of the

mutagenesis experiments. EO and MN: carried out ARTD profiling assays and analyzed the data.

YA: Performed some of the mutagenesis experiments and ARTD profiling. JK and TP designed

and performed DNA damage and toxicity assays. BL: designed the cell-based assays LL:

Conceived the study and wrote the manuscript. All authors contributed to the final version of the

manuscript.

CONFLICT OF INTEREST

HV, PV, BL and LL declare a financial interest due to a patent application concerning the OUL35

compound scaffold.

Page 20: Chemical probe rescues cells from ARTD10/PARP10 induced ...jultika.oulu.fi/files/nbnfi-fe201701181176.pdfARTD10 regulates NF- B signaling, where both its ADP-ribosylation activity

20

REFERENCES

Amé, J.-C., Spenlehauer, C., and de Murcia, G. (2004). The PARP superfamily. BioEssays 26, 882–

893.

Andersson, C.D., Karlberg, T., Ekblad, T., Lindgren, A.E.G., Thorsell, A.-G., Spjut, S.,

Uciechowska, U., Niemiec, M.S., Wittung-Stafshede, P., Weigelt, J., et al. (2012). Discovery of

ligands for ADP-ribosyltransferases via docking-based virtual screening. J. Med. Chem. 55, 7706–

7718.

Aoyagi-Scharber, M., Gardberg, A.S., Yip, B.K., Wang, B., Shen, Y., and Fitzpatrick, P.A. (2014).

Structural basis for the inhibition of poly(ADP-ribose) polymerases 1 and 2 by BMN 673, a potent

inhibitor derived from dihydropyridophthalazinone. Acta Crystallogr. Sect. F Struct. Biol.

Commun. 70, 1143–1149.

Barbarulo, A., Iansante, V., Chaidos, A., Naresh, K., Rahemtulla, A., Franzoso, G., Karadimitris,

A., Haskard, D.O., Papa, S., and Bubici, C. (2013). Poly(ADP-ribose) polymerase family member

14 (PARP14) is a novel effector of the JNK2-dependent pro-survival signal in multiple myeloma.

Oncogene 32, 4231–4242.

Bell, C.E., and Eisenberg, D. (1996). Crystal structure of diphtheria toxin bound to nicotinamide

adenine dinucleotide. Biochemistry (Mosc.) 35, 1137–1149.

Bütepage, M., Eckei, L., Verheugd, P., and Lüscher, B. (2015). Intracellular Mono-ADP-

Ribosylation in Signaling and Disease. Cells 4, 569–595.

Chang, P., Jacobson, M.K., and Mitchison, T.J. (2004). Poly(ADP-ribose) is required for spindle

assembly and structure. Nature 432, 645–649.

Chang, P., Coughlin, M., and Mitchison, T.J. (2005). Tankyrase-1 polymerization of poly(ADP-

ribose) is required for spindle structure and function. Nat. Cell Biol. 7, 1133–1139.

Chang, P., Coughlin, M., and Mitchison, T.J. (2009). Interaction between Poly(ADP-ribose) and

NuMA contributes to mitotic spindle pole assembly. Mol. Biol. Cell 20, 4575–4585.

Chen, Z.J., and Sun, L.J. (2009). Nonproteolytic functions of ubiquitin in cell signaling. Mol. Cell

33, 275–286.

Ekblad, T., Lindgren, A.E.G., Andersson, C.D., Caraballo, R., Thorsell, A.-G., Karlberg, T., Spjut,

S., Linusson, A., Schüler, H., and Elofsson, M. (2015). Towards small molecule inhibitors of mono-

ADP-ribosyltransferases. Eur. J. Med. Chem. 95, 546–551.

Fan, Y., Yu, Y., Mao, R., Zhang, H., and Yang, J. (2011). TAK1 Lys-158 but not Lys-209 is

required for IL-1β-induced Lys63-linked TAK1 polyubiquitination and IKK/NF-κB activation.

Cell. Signal. 23, 660–665.

Feijs, K.L., Kleine, H., Braczynski, A., Forst, A.H., Herzog, N., Verheugd, P., Linzen, U.,

Kremmer, E., and Lüscher, B. (2013a). ARTD10 substrate identification on protein microarrays:

regulation of GSK3β by mono-ADP-ribosylation. Cell Commun. Signal. 11, 5.

Feijs, K.L.H., Verheugd, P., and Lüscher, B. (2013b). Expanding functions of intracellular resident

mono-ADP-ribosylation in cell physiology. FEBS J. 280, 3519–3529.

Page 21: Chemical probe rescues cells from ARTD10/PARP10 induced ...jultika.oulu.fi/files/nbnfi-fe201701181176.pdfARTD10 regulates NF- B signaling, where both its ADP-ribosylation activity

21

Ferraris, D.V. (2010). Evolution of poly(ADP-ribose) polymerase-1 (PARP-1) inhibitors. From

concept to clinic. J. Med. Chem. 53, 4561–4584.

Ha, G.-H., Kim, H.-S., Go, H., Lee, H., Seimiya, H., Chung, D.H., and Lee, C.-W. (2012).

Tankyrase-1 function at telomeres and during mitosis is regulated by Polo-like kinase-1-mediated

phosphorylation. Cell Death Differ. 19, 321–332.

Haikarainen, T., Narwal, M., Joensuu, P., and Lehtiö, L. (2014). Evaluation and Structural Basis for

the Inhibition of Tankyrases by PARP Inhibitors. ACS Med. Chem. Lett. 5, 18–22.

Hassa, P.O., and Hottiger, M.O. (2008). The diverse biological roles of mammalian PARPS, a small

but powerful family of poly-ADP-ribose polymerases. Front. Biosci. J. Virtual Libr. 13, 3046–3082.

Herzog, N., Hartkamp, J.D.H., Verheugd, P., Treude, F., Forst, A.H., Feijs, K.L.H., Lippok, B.E.,

Kremmer, E., Kleine, H., and Lüscher, B. (2013). Caspase-dependent cleavage of the mono-ADP-

ribosyltransferase ARTD10 interferes with its pro-apoptotic function. FEBS J. 280, 1330–1343.

Hottiger, M.O., Hassa, P.O., Lüscher, B., Schüler, H., and Koch-Nolte, F. (2010). Toward a unified

nomenclature for mammalian ADP-ribosyltransferases. Trends Biochem. Sci. 35, 208–219.

Jafari, R., Almqvist, H., Axelsson, H., Ignatushchenko, M., Lundbäck, T., Nordlund, P., and

Martinez Molina, D. (2014). The cellular thermal shift assay for evaluating drug target interactions

in cells. Nat. Protoc. 9, 2100–2122.

Jones, G., Willett, P., Glen, R.C., Leach, A.R., and Taylor, R. (1997). Development and validation

of a genetic algorithm for flexible docking. J. Mol. Biol. 267, 727–748.

Jouanneau, S., Recoules, L., Durand, M.J., Boukabache, A., Picot, V., Primault, Y., Lakel, A.,

Sengelin, M., Barillon, B., and Thouand, G. (2014). Methods for assessing biochemical oxygen

demand (BOD): A review. Water Res. 49, 62–82.

Jwa, M., and Chang, P. (2012). PARP16 is a tail-anchored endoplasmic reticulum protein required

for the PERK- and IRE1α-mediated unfolded protein response. Nat. Cell Biol. 14, 1223–1230.

Karlberg, T., Thorsell, A.-G., Kallas, A., and Schuler, H. (2012). Crystal structure of human ADP-

ribose transferase ARTD15/poly-ADP-ribose polymerase-16 (PARP16) reveals a novel putative

regulatory domain. J. Biol. Chem. jbc.M112.379289.

Karlberg, T., Klepsch, M., Thorsell, A.-G., Andersson, C.D., Linusson, A., and Schüler, H. (2015).

Structural Basis for Lack of ADP-ribosyltransferase Activity in Poly(ADP-ribose) Polymerase-

13/Zinc Finger Antiviral Protein. J. Biol. Chem. 290, 7336–7344.

Kaufmann, M., Feijs, K.L.H., and Lüscher, B. (2015). Function and regulation of the mono-ADP-

ribosyltransferase ARTD10. Curr. Top. Microbiol. Immunol. 384, 167–188.

Kickhoefer, V.A., Siva, A.C., Kedersha, N.L., Inman, E.M., Ruland, C., Streuli, M., and Rome,

L.H. (1999). The 193-kD vault protein, VPARP, is a novel poly(ADP-ribose) polymerase. J. Cell

Biol. 146, 917–928.

Kirby, C.A., Cheung, A., Fazal, A., Shultz, M.D., and Stams, T. (2012). Structure of human

tankyrase 1 in complex with small-molecule inhibitors PJ34 and XAV939. Acta Crystallograph.

Sect. F Struct. Biol. Cryst. Commun. 68, 115–118.

Page 22: Chemical probe rescues cells from ARTD10/PARP10 induced ...jultika.oulu.fi/files/nbnfi-fe201701181176.pdfARTD10 regulates NF- B signaling, where both its ADP-ribosylation activity

22

Kleine, H., Poreba, E., Lesniewicz, K., Hassa, P.O., Hottiger, M.O., Litchfield, D.W., Shilton, B.H.,

and Lüscher, B. (2008). Substrate-Assisted Catalysis by PARP10 Limits Its Activity to Mono-ADP-

Ribosylation. Mol. Cell 32, 57–69.

Kleine, H., Herrmann, A., Lamark, T., Forst, A.H., Verheugd, P., Lüscher-Firzlaff, J., Lippok, B.,

Feijs, K.L., Herzog, N., Kremmer, E., et al. (2012). Dynamic subcellular localization of the mono-

ADP-ribosyltransferase ARTD10 and interaction with the ubiquitin receptor p62. Cell Commun.

Signal. 10, 28.

Koç, A., Wheeler, L.J., Mathews, C.K., and Merrill, G.F. (2004). Hydroxyurea Arrests DNA

Replication by a Mechanism That Preserves Basal dNTP Pools. J. Biol. Chem. 279, 223–230.

Koh, D.W., Dawson, T.M., and Dawson, V.L. (2005). Mediation of cell death by poly(ADP-ribose)

polymerase-1. Pharmacol. Res. 52, 5–14.

Loseva, O., Jemth, A.-S., Bryant, H.E., Schüler, H., Lehtiö, L., Karlberg, T., and Helleday, T.

(2010). PARP-3 is a mono-ADP-ribosylase that activates PARP-1 in the absence of DNA. J. Biol.

Chem. 285, 8054–8060.

Malanga, M., and Althaus, F.R. (2005). The role of poly(ADP-ribose) in the DNA damage

signaling network. Biochem. Cell Biol. Biochim. Biol. Cell. 83, 354–364.

Morgan, R.K., Carter-O’Connell, I., and Cohen, M.S. (2015). Selective inhibition of PARP10 using

a chemical genetics strategy. Bioorg. Med. Chem. Lett.

Narwal, M., Fallarero, A., Vuorela, P., and Lehtiö, L. (2012). Homogeneous screening assay for

human tankyrase. J. Biomol. Screen. 17, 593–604.

Nicolae, C.M., Aho, E.R., Vlahos, A.H.S., Choe, K.N., De, S., Karras, G.I., and Moldovan, G.-L.

(2014). The ADP-ribosyltransferase PARP10/ARTD10 Interacts with Proliferating Cell Nuclear

Antigen (PCNA) and Is Required for DNA Damage Tolerance. J. Biol. Chem. 289, 13627–13637.

Nicolae, C.M., Aho, E.R., Choe, K.N., Constantin, D., Hu, H.-J., Lee, D., Myung, K., and

Moldovan, G.-L. (2015). A novel role for the mono-ADP-ribosyltransferase PARP14/ARTD8 in

promoting homologous recombination and protecting against replication stress. Nucleic Acids Res.

43, 3143–3153.

Oeckinghaus, A., Hayden, M.S., and Ghosh, S. (2011). Crosstalk in NF-κB signaling pathways.

Nat. Immunol. 12, 695–708.

Otto, H., Reche, P.A., Bazan, F., Dittmar, K., Haag, F., and Koch-Nolte, F. (2005). In silico

characterization of the family of PARP-like poly(ADP-ribosyl)transferases (pARTs). BMC

Genomics 6, 139.

Petesch, S.J., and Lis, J.T. (2008). Rapid, transcription-independent loss of nucleosomes over a

large chromatin domain at Hsp70 loci. Cell 134, 74–84.

Ruf, A., Rolli, V., de Murcia, G., and Schulz, G.E. (1998). The mechanism of the elongation and

branching reaction of poly(ADP-ribose) polymerase as derived from crystal structures and

mutagenesis. J. Mol. Biol. 278, 57–65.

Page 23: Chemical probe rescues cells from ARTD10/PARP10 induced ...jultika.oulu.fi/files/nbnfi-fe201701181176.pdfARTD10 regulates NF- B signaling, where both its ADP-ribosylation activity

23

Scarpa, E.S., Fabrizio, G., and Di Girolamo, M. (2013). A role of intracellular mono-ADP-

ribosylation in cancer biology. FEBS J. 280, 3551–3562.

Schreiber, V., Dantzer, F., Ame, J.-C., and de Murcia, G. (2006). Poly(ADP-ribose): novel

functions for an old molecule. Nat. Rev. Mol. Cell Biol. 7, 517–528.

Venkannagari, H., Fallarero, A., Feijs, K.L.H., Lüscher, B., and Lehtiö, L. (2013). Activity-based

assay for human mono-ADP-ribosyltransferases ARTD7/PARP15 and ARTD10/PARP10 aimed at

screening and profiling inhibitors. Eur. J. Pharm. Sci. 49, 148–156.

Verheugd, P., Forst, A.H., Milke, L., Herzog, N., Feijs, K.L.H., Kremmer, E., Kleine, H., and

Lüscher, B. (2013). Regulation of NF-κB signalling by the mono-ADP-ribosyltransferase ARTD10.

Nat. Commun. 4, 1683.

Vyas, S., and Chang, P. (2014). New PARP targets for cancer therapy. Nat. Rev. Cancer 14, 502–

509.

Vyas, S., Matic, I., Uchima, L., Rood, J., Zaja, R., Hay, R.T., Ahel, I., and Chang, P. (2014).

Family-wide analysis of poly(ADP-ribose) polymerase activity. Nat. Commun. 5, 4426.

Wahlberg, E., Karlberg, T., Kouznetsova, E., Markova, N., Macchiarulo, A., Thorsell, A.-G., Pol,

E., Frostell, Å., Ekblad, T., Öncü, D., et al. (2012). Family-wide chemical profiling and structural

analysis of PARP and tankyrase inhibitors. Nat. Biotechnol. 30, 283–288.

Xu, M., Skaug, B., Zeng, W., and Chen, Z.J. (2009). A ubiquitin replacement strategy in human

cells reveals distinct mechanisms of IKK activation by TNFalpha and IL-1beta. Mol. Cell 36, 302–

314.

Yu, M., Schreek, S., Cerni, C., Schamberger, C., Lesniewicz, K., Poreba, E., Vervoorts, J.,

Walsemann, G., Grötzinger, J., Kremmer, E., et al. (2005). PARP-10, a novel Myc-interacting

protein with poly(ADP-ribose) polymerase activity, inhibits transformation. Oncogene 24, 1982–

1993.

Page 24: Chemical probe rescues cells from ARTD10/PARP10 induced ...jultika.oulu.fi/files/nbnfi-fe201701181176.pdfARTD10 regulates NF- B signaling, where both its ADP-ribosylation activity

24

Figure legends

Figure 1. Compounds used in the study and potency of OUL35

(A) Chemical structures for compounds OUL47, OUL35, OUL77, OUL78, 3AB, and NAD+. (B)

Dose-response curve for OUL35 against the catalytic domain of bacterially expressed and purified

ARTD10.

Figure 2. Docking model of OUL35 to the ARTD10 crystal structure and structural

comparisons to other ARTD enzymes

(A) OUL35 docking pose in the catalytic domain of ARTD10. Hydrogen bonds are indicated with

dashed lines. Docking pose superposed on (B) ARTD1 (PDB id. 4PJT (Aoyagi-Scharber et al.,

2014)), (C) ARTD5 (PDB id. 3UH2 (Kirby et al., 2012)), (D) ARTD8 (PDB id. 3GOY (Wahlberg

et al., 2012)), (E) ARTD15 (PDB id. 4F0D (Karlberg et al., 2012)), and (F) ARTD7 (PDB id.

3GEY (Karlberg et al., 2015)).

Figure 3. OUL35 rescues ARTD10 overexpressing cells

(A) Images of the wells from colony formation experiments. ARTD10 expression was not induced

in cells in the control wells, while the expressions of ARTD10 and the inactive mutant ARTD10-

GW (G888W) were induced with doxycycline (Dox, 500 ng/ml) in the rest of the wells. The

different compounds were added as indicated with a final concentration of 10 µM. These were

replenished every 48 h. (B) Quantification of the colony formation results. The data represent mean

values of three independent experiments performed in duplicates with standard deviations. (C)

Dose-response curve of OUL35 rescue. (D) Cellular thermal stability assay demonstrating that

OUL35 (3 µM) binds to endogenous ARTD10 in U2OS cells.

Page 25: Chemical probe rescues cells from ARTD10/PARP10 induced ...jultika.oulu.fi/files/nbnfi-fe201701181176.pdfARTD10 regulates NF- B signaling, where both its ADP-ribosylation activity

25

Figure 4. ARTD10 induced cell death is antagonized by OUL35

Representative images of FACS scans of HeLa cells with and without induced ARTD10-GFP

expression. The cells were stained with Annexin V and propidium iodide (PI). (A) Uninduced

control cells with DMSO, (B) doxycycline induced cells with DMSO, (C) doxycycline induced

cells with OUL35. (D) A summary of the data from three independent experiments indicating the

percent of apoptotic and dead cells stained for Annexin V and with propidium iodide (PI). The data

represent the mean ± SEM of three independent experiments.

Figure 5. The effect of OUL35 on HeLa cells treated with hydroxyurea

(A) Proliferation curves of HeLa cells treated with DMSO vehicle or OUL35 (5 µM) and different

concentrations of HU (n=3, data present mean ± SEM). (B) Cell confluence at 72 hours (n = 5, data

present mean ± SEM , n.s = not significant, *P < 0.05, **P < 0.01). (C) Proliferation curves of

HeLa cells treated with DMSO vehicle or OUL35 (1 µM) and different concentrations of HU (n=5,

data present mean ± SEM). (D) Proliferation curves of HeLa cells treated with DMSO vehicle or

OUL77 (5 µM) and different concentrations of HU (n=3, data present mean ± SEM).

Page 26: Chemical probe rescues cells from ARTD10/PARP10 induced ...jultika.oulu.fi/files/nbnfi-fe201701181176.pdfARTD10 regulates NF- B signaling, where both its ADP-ribosylation activity

26

Table 1. IC50’s and thermal stabilization by OUL35 against a panel of human ARTD enzymes

ARTD / PARP IC50 (pIC50±SEM) ΔTm (DSF), oC ±STDEV

ARTD1 / PARP1 >100 µM 0.42 ± 0.09

ARTD2 / PARP2 >100 µM 0.15 ± 0.12

ARTD3 / PARP3 >100 µM 0.09 ± 0.32

ARTD4 / PARP4 22.6 µM (4.65 ± 0.17) 0.84 ± 0.14

ARTD5 / PARP5a >100 µM 0.45 ± 0.01

ARTD6 / PARP5b >100 µM 0.15 ± 0.12

ARTD7 / PARP15 > 10 µMa 1.19 ± 0.10

ARTD8 / PARP14 23.4 µM (4.63 ± 0.09) 2.68 ± 0.22

ARTD9 / PARP9 Inactive -0.77 ± 0.16

ARTD10 / PARP10 329 nM (6.48 ± 0.04) 3.76 ± 0.10

ARTD12 / PARP12 > 100 µM 0.87 ± 0.42

ARTD13 / PARP13 Inactive -0.21 ± 0.04

ARTD15 / PARP16 4.17 µM (5.38 ± 0.06) 2.45 ± 0.11

ARTD10 / PARP10 (G888W) Inactive -0.66 ± 0.68

ARTD10 / PARP10 (L926Y) 3.84 µM (5.41 ± 0.05) 2.63 ± 0.09

ARTD10 / PARP10 (R931A) 597 nM (6.422 ± 0.14) 3.73 ± 0.19

ARTD10 / PARP10 (S927A) 2.25 µM (5.65 ± 0.005) 2.10 ± 0.12

ARTD7 / PARP15 (Y576L) 295 nM (6.53 ± 0.05) 4.09 ± 0.10

ARTD8 / PARP14 (Y1640L) 1.63 µM (5.79 ± 0.14) 4.66 ± 0.07

a Compound concentration limited by ARTD DMSO sensitivity. Less than 50 % inhibition was

observed when tested at 20 µM concentration.

Page 27: Chemical probe rescues cells from ARTD10/PARP10 induced ...jultika.oulu.fi/files/nbnfi-fe201701181176.pdfARTD10 regulates NF- B signaling, where both its ADP-ribosylation activity

27

Figure 1

Page 28: Chemical probe rescues cells from ARTD10/PARP10 induced ...jultika.oulu.fi/files/nbnfi-fe201701181176.pdfARTD10 regulates NF- B signaling, where both its ADP-ribosylation activity

28

Figure 2

Page 29: Chemical probe rescues cells from ARTD10/PARP10 induced ...jultika.oulu.fi/files/nbnfi-fe201701181176.pdfARTD10 regulates NF- B signaling, where both its ADP-ribosylation activity

29

Figure 3

Page 30: Chemical probe rescues cells from ARTD10/PARP10 induced ...jultika.oulu.fi/files/nbnfi-fe201701181176.pdfARTD10 regulates NF- B signaling, where both its ADP-ribosylation activity

30

Figure 4

Page 31: Chemical probe rescues cells from ARTD10/PARP10 induced ...jultika.oulu.fi/files/nbnfi-fe201701181176.pdfARTD10 regulates NF- B signaling, where both its ADP-ribosylation activity

31

Figure 5

Page 32: Chemical probe rescues cells from ARTD10/PARP10 induced ...jultika.oulu.fi/files/nbnfi-fe201701181176.pdfARTD10 regulates NF- B signaling, where both its ADP-ribosylation activity

CONTENTS

Figure S1: Toxicity effects of DMSO, OUL35 and OUL47 on Hela cells.

Figure S2: Activity of ARTD10 with compounds OUL35, OUL77 and OUL78.

Figure S3: Comparison of the crystal structure of ARTD10 in complex with 3AB and the docking of OUL35 to

ARTD10.

Figure S4: Structural comparisons of the OUL35 pose with ARTD2, ARTD3, ARTD6, ARTD12 and ARTD13 crystal

structures.

Figure S5: Sequence alignment of the ART domains.

Figure S6: Western blot activity assays for ARTD10 WT and mutants.

Table S1: The constructs used in the thermal stability assays.

Table S2: The constructs and conditions used for the activity assays.

References

Page 33: Chemical probe rescues cells from ARTD10/PARP10 induced ...jultika.oulu.fi/files/nbnfi-fe201701181176.pdfARTD10 regulates NF- B signaling, where both its ADP-ribosylation activity

Figure S1, related to Figure 1. Toxicity effects of DMSO, OUL35 and OUL47 on HeLa cells.

Images of Hela cells at 24 h (A, B, C) and at 48 h (D, E, F). Treatment of Hela cells with DMSO vehicle (A and D), 10

µM of OUL35 (B and E) or OUL47 (C and F) indicated that OUL35 has no cytotoxic effect in the absence of HU

whereas OUL47 completely inhibited the growth of Hela cells. (G) Proliferation curves showing % of cell confluence

(error bars ± SEM, n=2).

Page 34: Chemical probe rescues cells from ARTD10/PARP10 induced ...jultika.oulu.fi/files/nbnfi-fe201701181176.pdfARTD10 regulates NF- B signaling, where both its ADP-ribosylation activity

Figure S2, related to Figures 1, 3, and 5. The activity of ARTD10 is compared in the presence of OUL35 and its

inactive analogs OUL77 and OUL78. The compounds were tested at 1 and 10 µM concentrations. Data shown are mean values of triplicates ± SD.

% A

ctiv

ity

DM

SO

Contr

ol

OU

L35

OU

L77

OU

L78

0

5 0

1 0 0

1 0 µ M

1 µ M

Page 35: Chemical probe rescues cells from ARTD10/PARP10 induced ...jultika.oulu.fi/files/nbnfi-fe201701181176.pdfARTD10 regulates NF- B signaling, where both its ADP-ribosylation activity

Figure S3, related to Figure 2. Comparison of the docking pose with the crystal structure of ARTD10.

(A) 3AB in complex with ARTD10 (PDB id. 3HKV). (B) Docking pose of OUL35 with ARTD10 (this figure is

identical to Fig. 2a and is shown here to allow direct comparison to panel A). OUL35 binds to the nicotinamide site in a

similar manner to 3AB. (C) NAD+ binding to ARTD10 based on superposition of the diphtheria toxin NAD

+ co-crystal

structure (PDB id. 1TOX (Ruf et al., 1998)). Nicotinamide site (NI), Adenosine site (ADE) as well as donor and

acceptor NAD+ sites are labelled. OUL35 docking pose is shown as reference.

Page 36: Chemical probe rescues cells from ARTD10/PARP10 induced ...jultika.oulu.fi/files/nbnfi-fe201701181176.pdfARTD10 regulates NF- B signaling, where both its ADP-ribosylation activity

Figure S4, related to Figure 2. Comparison of OUL35 binding to ARTD10 with other enzymes. (A) ARTD2 (PDB id. 3KCZ (Karlberg et al., 2010)), (B) ARTD3 (PDB id. 3FHB (Lehtiö et al., 2009)), (C) ARTD6

(PDB id. 3U9H (Narwal et al., 2012)), (D) ARTD12 (PDB id. 2PQF (Karlberg et al., 2015)), (E) ARTD13 (PDB id.

4X52). His810 of ARTD13 effectively closes the binding pocket (Karlberg et al., 2015).

Page 37: Chemical probe rescues cells from ARTD10/PARP10 induced ...jultika.oulu.fi/files/nbnfi-fe201701181176.pdfARTD10 regulates NF- B signaling, where both its ADP-ribosylation activity

01|P09874|788-1014 --DPIDVNYEKLKTD---IKVVDRDSEEAEIIRKYVKNTHA------TTHNAYDLEVIDI

02|Q9UGN5|356-583 --HPLDQHYRNLHCA---LRPLDHESYEFKVISQYLQSTHA------PTHSDYTMTLLDL

03|Q9Y6F1|313-533 --HPLDRDYQLLKCQ---LQLLDSGAPEYKVIQTYLEQTGS-------NHRCPTLQH--I

04|Q9UKK3|369-573 --PPSLAKYRALRCK---IEHVEQNTEEFLRVRKEVLQNHH------------SKSPVDV

05|O95271|1112-1317 --APEDKEYQSVEEEMQSTIREHRDGGNAGGIFNRY-----------NVIRIQKVVNKKL

06|Q9H2K2|959-1164 --SPDDKEFQSVEEEMQSTVREHRDGGHAGGIFNRY-----------NILKIQKVCNKKL

07|Q460N3|482-678 --LPEHWTDMN--HQLFCMVQLEPGQSEYNTIKDKFTRTCS--S--YAIEKIERIQNAFL

08|Q460N5|1605-1801 --IPAHWSDMK--QQNFCVVELLPSDPEYNTVASKFNQTCS--H--FRIEKIERIQNPDL

09|Q8IXQ6|628-850 --IQQQKTQDEMKENIIFLKCPVPPTQELLDQKKQFEKCGL------QVLKVEKIDNEVL

10|Q53GL7|806-1025 ----PTLAGQTLKGPWNNLERLAENTGEFQEVVRAFYDTLDAARSSIRVVRVERVSHPLL

11|Q9NR21|116-331 IPMPPHWENVNTQVP-YQLIPLHNQTHEYNEVANLFGKTMD--R--NRIKRIQRIQNLDL

12|Q9H0J9|484-698 --IPDYWDSSALPDPGFQKITLSSSSEEYQKVWNLFNRTLP--F—YFVQKIERVQNLAL

13|Q7Z2W4|716-902 --PQEDFCFLS--SKKYKLSEIHHLHPEYVRVSEHFKASMK--N--FKIEKIKKIENSEL

14|Q7Z3E1|449-657 --YPETWVYMH-PSQDFIQVPVSAEDKSYRIIYNLFHKTVPEFK--YRILQILRVQNQFL

15|Q8N5Y8|94-279 -----------------VLTIHSAGKAEFEKIQKLTGAPHT---------PVPAPDFLFE

16|Q8N3A8|617-844 --EMTQAPYLEIKKQMDKQDPLAHPLLQWVISSNRSHIVKLPVN---RQLKFMHTPHQFL

17|Q2NL67|394-620 --EMTQGSYLEIKKQMDKLDPLAHPLLQWIISSNRSHIVKLPL----SRLKFMHTSHQFL

01|P09874|788-1014 FKIEREGECQRYKPFKQL---HNRRLLWHGSRTTNFAGILSQGLRIAPPEA--PVTG---

02|Q9UGN5|356-583 FEVEKDGEKEAFREDL-----HNRMLLWHGSRMSNWVGILSHGLRIAPPEA--PITG---

03|Q9Y6F1|313-533 WKVNQEGEEDRFQAHSKL---GNRKLLWHGTNMAVVAAILTSGL----RIM--PHSG---

04|Q9UKK3|369-573 LQIFRVGRVNETTEFLSKL--GNVRPLLHGSPVQNIVGILCRGL-LLPKVV--EDRGVQR

05|O95271|1112-1317 RERFCHRQKEVSEENHNH---HNERMLFHGSP--FINAIIHKGF--DERHA---YIG---

06|Q9H2K2|959-1164 WERYTHRRKEVSEENHNH---ANERMLFHGSP--FVNAIIHKGF--DERHA---YIG---

07|Q460N3|482-678 WQSYQVKKRQMDIKNDHK---NNERLLFHGTDADSVPYVNQHGF--NRSCA--GKNA---

08|Q460N5|1605-1801 WNSYQAKKKTMDAKNGQT---MNEKQLFHGTDAGSVPHVNRNGF--NRSYA--GKNA---

09|Q8IXQ6|628-850 MAAFQRKKKMMEEKLHRQ---PVSHRLFQQVPYQFCNVVCRVGF--QRMYS--TPCD---

10|Q53GL7|806-1025 QQQYELYRERLLQRCERR---PVEQVLYHGTTAPAVPDICAHGF--NRSFC--GRNA---

11|Q9NR21|116-331 WEFFCRKKAQLKKKRGVPQ--INEQMLFHGTSSEFVEAICIHNF--DWRIN--GIHG---

12|Q9H0J9|484-698 WEVYQWQKGQMQKQNGGKA--VDERQLFHGTSAIFVDAICQQNF--DWRVC--GVHG---

13|Q7Z2W4|716-902 LDKFTWKKSQMK---------EEGKLLFYATSRAYVESICSNNF--DSFLH--ETHE---

14|Q7Z3E1|449-657 WEKYKRKKEYMNRKMFGRDRIINERHLFHGTSQDVVDGICKHNF--DPRVC--GKHA---

15|Q8N5Y8|94-279 IEYFDPANAKFYETKGE----RDLIYAFHGSRLENFHSIIHNGL-----HC--HLNKT--

16|Q8N3A8|617-844 LLSSPPAKESNFRAAKKL---FGSTFAFHGSHIENWHSILRNGL-VVASNTRLQLHG---

17|Q2NL67|394-620 LLSSPPAKEARFRTAKKL---YGSTFAFHGSHIENWHSILRNGL-VNASYTKLQLHG---

01|P09874|788-1014 ---YMFGKGIYFADMVSKSANYCH--------TSQGDPIGL-----------ILLGEVAL

02|Q9UGN5|356-583 ---YMFGKGIYFADMSSKSANYCF--------ASRLKNTGL-----------LLLSEVAL

03|Q9Y6F1|313-533 ---GRVGKGIYFASENSKSAGYVI------GMKCGAHHVGY-----------MFLGEVAL

04|Q9UKK3|369-573 TDVGNLGSGIYFSDSLSTSIKYSH--------PGETDGTRL-----------LLICDVAL

05|O95271|1112-1317 ---GMFGAGIYFAENSSKSNQYVYGIGGGTGCPTHKDRSCY-----------ICHRQMLF

06|Q9H2K2|959-1164 ---GMFGAGIYFAENSSKSNQYVYGIGGGTGCPVHKDRSCY-----------ICHRQLLF

07|Q460N3|482-678 ---VSYGKGTYFAVDASYSAKDTY-------SKPDSNGRKH-----------MYVVRVLT

08|Q460N5|1605-1801 ---VAYGKGTYFAVNANYSANDTY-------SRPDANGRKH-----------VYYVRVLT

09|Q8IXQ6|628-850 ---PKYGAGIYFTKNLKNLAEKAK-------KISAADKLIY-----------VFEAEVLT

10|Q53GL7|806-1025 ---TVYGKGVYFARRASLSVQDRY-------SPPNADGHKA-----------VFVARVLT

11|Q9NR21|116-331 ---AVFGKGTYFARDAAYSSRFCK-DDIKHGNTFQIHGVSLQQRHLFRTYKSMFLARVLI

12|Q9H0J9|484-698 ---TSYGKGSYFARDAAYSHHYSK----------SDTQTHT-----------MFLARVLV

13|Q7Z2W4|716-902 ---NKYGKGIYFAKDAIYSHKNCP----------YDAKNVV-----------MFVAQVLV

14|Q7Z3E1|449-657 ---TMFGQGSYFAKKASYSHNFSK---------KSSKGVHF-----------MFLAKVLT

15|Q8N5Y8|94-279 ---SLFGEGTYLTSDLSLALIYSP----------HGHGWQH-----------SLLGPILS

16|Q8N3A8|617-844 ---AMYGSGIYLSPMSSISFGYSG-----------MNKKQK-----------VSAKDEPA

17|Q2NL67|394-620 ---AAYGKGIYLSPISSISFGYSG----------MGKGQHR-----------MPSKDELV

Page 38: Chemical probe rescues cells from ARTD10/PARP10 induced ...jultika.oulu.fi/files/nbnfi-fe201701181176.pdfARTD10 regulates NF- B signaling, where both its ADP-ribosylation activity

01|P09874|788-1014 GNMYELKHASHISKLPKGKHSV----KGLGKTTPDPSANISLDGVDVPLGTGISSGV---

02|Q9UGN5|356-583 GQ-CNELLEANPKAEGLLQGKHS--TKGLGKMAPSSAHFVTLNGSTVPLGPASDTGILNP

03|Q9Y6F1|313-533 GR-EHHINTDNPSLKSPPPGFDSVIARGHTEPDPTQDTELELDGQQVVVPQGQPVPCPEF

04|Q9UKK3|369-573 GK-CMDLHEKDFSLTEAPPGYDS--VHGVSQTASVTTDF---------------------

05|O95271|1112-1317 CR-VTLGKSFLQFSTMKMAHAP----PGHHSVIGRPSVN---------------------

06|Q9H2K2|959-1164 CR-VTLGKSFLQFSAMKMAHSP----PGHHSVTGRPSVN---------------------

07|Q460N3|482-678 GV-FTKGRAGLVTPPPKNPHNPT---DLFDSVTNNTRSP---------------------

08|Q460N5|1605-1801 GI-YTHGNHSLIVPPSKNPQNPT---DLYDTVTDNVHHP---------------------

09|Q8IXQ6|628-850 GF-FCQGHPLNIVPPPLSPGAI----DGHDSVVDNVSSP---------------------

10|Q53GL7|806-1025 GD-YGQGRRGLRAPPLRGPGHVL---LRYDSAVDCICQP---------------------

11|Q9NR21|116-331 GD-YINGDSKYMRPPSKDGSYV----NLYDSCVDDTWNP---------------------

12|Q9H0J9|484-698 GE-FVRGNASFVRPPAKEGWSN----AFYDSCVNSVSDP---------------------

13|Q7Z2W4|716-902 GK-FTEGNITYTSPP-----------PQFDSCVDTRSNP---------------------

14|Q7Z3E1|449-657 GR-YTMGSHGMRRPPPVNPGSVT--SDLYDSCVDNFFEP---------------------

15|Q8N5Y8|94-279 CV-AVCEVIDHPDVKCQTKKKDS---KEIDRRRARIKHS---------------------

16|Q8N3A8|617-844 SS-SKSSNTSQSQKKGQQSQFLQ--SRNLKCIALCEVIT--------------------S

17|Q2NL67|394-620 QR-YNRMNT-IPQTRSIQSRFLQ--SRNLNCIALCEVIT--------------------S

01|P09874|788-1014 NDTSLLYNEYIVYDIAQVNLKYLLKLKFNFKTSLW--------------------

02|Q9UGN5|356-583 DGYTLNYNEYIVYNPNQVRMRYLLKVQFNF-LQLW--------------------

03|Q9Y6F1|313-533 SSSTFSQSEYLIYQESQCRLRYLLEVHL---------------------------

04|Q9UKK3|369-573 -----EDDEFVVYKTNQVKMKYIIKFSMPGDQIKD--------------------

05|O95271|1112-1317 ---GLAYAEYVIYRGEQAYPEYLITYQIMKPEAPS--------------------

06|Q9H2K2|959-1164 ---GLALAEYVIYRGEQAYPEYLITYQIMRPEGMV--------------------

07|Q460N3|482-678 -------KLFVVFFDNQAYPEYLITFTA---------------------------

08|Q460N5|1605-1801 -------SLFVAFYDYQAYPEYLITFRK---------------------------

09|Q8IXQ6|628-850 -------ETFVIFSGMQAIPQYLWTCTQEYVQSQDYSSGPMRPFAQHPWRGFASG

10|Q53GL7|806-1025 -------SIFVIFHDTQALPTHLITCEHVPRASPDDPSGLPGRSPDT--------

11|Q9NR21|116-331 -------KIFVVFDANQIYPEYLIDFH----------------------------

12|Q9H0J9|484-698 -------SIFVIFEKHQVYPEYVIQYTTSSKPSVTPSILLALGSLFS--------

13|Q7Z2W4|716-902 -------SVFVIFQKDQVYPQYVIEYTEDKACVIS--------------------

14|Q7Z3E1|449-657 -------QIFVIFNDDQSYPYFVIQYEEVSNTVSI--------------------

15|Q8N5Y8|94-279 EGGDIPPKYFVVTNNQLLRVKYLLVYSQKPPKRA---------------------

16|Q8N3A8|617-844 SDLHKHGEIWVVPNTDHVCTRFFFVYEDGQVGDANINTQEGGIHKEIL-------

17|Q2NL67|394-620 KDLQKHGNIWVCPVSDHVCTRFFFVYEDGQVGDANINTQDPKIQKEIM-------

Figure S5, related to Figure 2: Sequence alignment for the catalytic domains of the 17 different ARTDs (ARTD

numbering). Uniprot code used for each protein sequence is indicated. The conserved regions critical for OUL35 selectivity around

the active site region are highlighted. The residues on the β-strand are highlighted in yellow while the conserved

residues on the alpha-helix are highlighted in grey. Histidine and glycine in the active site region are highlighted in blue

while the catalytic glutamate and its replacements are highlighted in green. Sequence alignment was performed using

MUSCLE (Edgar, 2004).

Page 39: Chemical probe rescues cells from ARTD10/PARP10 induced ...jultika.oulu.fi/files/nbnfi-fe201701181176.pdfARTD10 regulates NF- B signaling, where both its ADP-ribosylation activity

Figure S6, related to Table 1. The enzymatic reactions contained 1 μM biotinylated NAD+ together with 500 nM

ARTD10 wt or mutants and 2 μM SRPK2. All the enzymes were robustly ADP-ribosylating SRPK2 and showed much

less automodification of ARTD10.

Page 40: Chemical probe rescues cells from ARTD10/PARP10 induced ...jultika.oulu.fi/files/nbnfi-fe201701181176.pdfARTD10 regulates NF- B signaling, where both its ADP-ribosylation activity

Table S1, related to Table 1. Constructs used for performing thermal stability assay.

ARTD Amino Acid composition Vector

ARTD1 654 - 1013 pNIC28-Bsa4

ARTD2 234 - 579 pNIC28-Bsa4

ARTD3 176 - 532 pNIC28-Bsa4

ARTD4 250 - 565 pNIC28-Bsa4

ARTD5 1091 - 1325 pNIC28-Bsa4

ARTD6 952 - 1161 pNIC28-Bsa4

ARTD7 460 - 656 pNIC28-Bsa4

ARTD7 (Y576L) 460 - 656 pNIC28-Bsa4

ARTD8 1535 - 1801 pNIC28-Bsa4

ARTD8 (Y1640L) 1535 - 1801 pNIC28-Bsa4

ARTD9 631 - 834 pNIC-CH

ARTD10 809 - 1017 pNIC-CH

ARTD10 (G888W) 809 - 1017 pNIC-CH

ARTD10 (L926Y) 809 - 1017 pNIC-CH

ARTD10 (R931A) 809 - 1017 pNIC-CH

ARTD10 (S927A) 809 - 1017 pNIC-CH

ARTD12 489 - 684 pNIC28-Bsa4

ARTD13 726 - 896 pNIC28-Bsa4

ARTD15 1 - 280 pNIC28-Bsa4

Page 41: Chemical probe rescues cells from ARTD10/PARP10 induced ...jultika.oulu.fi/files/nbnfi-fe201701181176.pdfARTD10 regulates NF- B signaling, where both its ADP-ribosylation activity

Table S2, related to Table 1. Constructs and conditions used in the activity assays.

ARTD Amino Acids Vector Assay Conditions

ARTD1 1 - 1014 pNIC-Bsa4

5 nM ARTD1 and NAD+ 500 nM

50 mM Tris pH 8, 2 mM Mg2+, 10 µg/mL activated DNA

1.5 h shaking at RT

ARTD2 1 - 583 pNH-TrxT

5 nM ARTD2 and NAD+ 500 nM

50 mM Tris pH 8, 5 mM Mg2+, 10 µg/mL activated DNA

1.5 h shaking at RT

ARTD3 1 - 533 pNH-TrxT

200 nM ARTD3 and 500 nM NAD+

50 mM HEPES pH 7, 10 µg/mL DNA, 8 mM MgCl2

3 h shaking at RT

ARTD4 250 - 565 pNIC28-Bsa4

200 nM ARTD4 and 500 nM NAD+

50 mM Na-P buffer pH 7.5, 0.5 mM TCEP, 1 mg/mL BSA

2.5 h shaking at RT

ARTD5 1030 - 1317 pNIC28-Bsa4

20 nM ARTD5 and 500 nM NAD+

50 mM BisTris propane pH 7, 0.5 mM TCEP,

0.01% Triton X-100

18 h shaking at RT

ARTD6 873 – 1161 pNIC-ZB

20 nM ARTD6 and 500 nM NAD+

50 mM BisTris propane pH 7, 0.5 mM TCEP,

0.01% Triton X-100

18 h shaking at RT /300rpm

ARTD7 460 - 656 pNIC28-Bsa4 200 nM ARTD7, 500 nM SRPK2 and 250 nM NAD+

50 mM Na-P pH 7

8 h shaking at RT

ARTD7

(Y576L) 460 - 656 pNIC28-Bsa4

150 nM ARTD7(Y576L), 500 nM SRPK2 and 250 nM NAD+

50 mM Na-P pH 7

2.5 h shaking at RT

ARTD8 1535 - 1801 pNH-TrxT 500 nM ARTD8 and 500 nM NAD+

50 mM Na-P pH 7.0

21 h shaking at RT

ARTD8

(Y1640L) 1535 - 1801 pNH-TrxT

500 nM ARTD8 (Y1640L) and 500 nM NAD+

50 mM Na-P pH 7.0

22 h shaking at RT

ARTD10 809 - 1017 pNIC-CH 100 nM ARTD10, 2 μM SRPK2 and 500 nM NAD+

50 mM Tris pH 7.0

13 h shaking at RT

ARTD10

(L926Y) 809 - 1017 pNIC-CH

200 nM ARTD10(L926Y), 2 μM SRPK2 and 500 nM NAD+

50 mM Tris pH 7.0

21 h shaking at RT

ARTD10

(G888W) 809 - 1017 pNIC-CH N/A

Page 42: Chemical probe rescues cells from ARTD10/PARP10 induced ...jultika.oulu.fi/files/nbnfi-fe201701181176.pdfARTD10 regulates NF- B signaling, where both its ADP-ribosylation activity

Table S2 contd.

ARTD Amino Acids Vector Assay Conditions

ARTD10

(R931A) 809 - 1017 pNIC-CH

200 nM ARTD10(R931A), 2 μM SRPK2 and 500 nM NAD+

50 mM Tris pH 7.0

21 h shaking at RT

ARTD10

(S927A) 809 - 1017 pNIC-CH

200 nM ARTD10(S927A), 2 μM SRPK2 and 500 nM NAD+

50 mM Tris pH 7.0

21 h shaking at RT

ARTD12 489 - 701 pNH-TrxT 500 nM ARTD12 and 500 nM NAD+

50 mM Na-P pH 7

20 h shaking at RT

ARTD15 1 - 280 pNIC28-Bsa4 2 μM ARTD15 and 500 nM NAD+

50 mM HEPES ph 7, 2 mM NiCl2

24 h shaking at RT

References

Edgar, R.C. (2004). MUSCLE: multiple sequence alignment with high accuracy and high throughput. Nucleic Acids

Res. 32, 1792–1797.

Karlberg, T., Hammarström, M., Schütz, P., Svensson, L., and Schüler, H. (2010). Crystal structure of the catalytic

domain of human PARP2 in complex with PARP inhibitor ABT-888. Biochemistry (Mosc.) 49, 1056–1058.

Karlberg, T., Klepsch, M., Thorsell, A.-G., Andersson, C.D., Linusson, A., and Schüler, H. (2015). Structural Basis for

Lack of ADP-ribosyltransferase Activity in Poly(ADP-ribose) Polymerase-13/Zinc Finger Antiviral Protein. J. Biol.

Chem. 290, 7336–7344.

Lehtiö, L., Jemth, A.-S., Collins, R., Loseva, O., Johansson, A., Markova, N., Hammarström, M., Flores, A., Holmberg-

Schiavone, L., Weigelt, J., et al. (2009). Structural basis for inhibitor specificity in human poly(ADP-ribose)

polymerase-3. J. Med. Chem. 52, 3108–3111.

Narwal, M., Venkannagari, H., and Lehtiö, L. (2012). Structural basis of selective inhibition of human tankyrases. J.

Med. Chem. 55, 1360–1367.

Ruf, A., Rolli, V., de Murcia, G., and Schulz, G.E. (1998). The mechanism of the elongation and branching reaction of

poly(ADP-ribose) polymerase as derived from crystal structures and mutagenesis. J. Mol. Biol. 278, 57–65.