Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

Article

Cite This: J. Nat. Prod. XXXX, XXX, XXX−XXX pubs.acs.org/jnp

Antiviral Compounds from Codiaeum peltatum Targeted by a Multi-


informative Molecular Networks Approach
Florent Olivon,† Simon Remy,† Gwendal Grelier,† Ceć ile Apel,† Ceć ilia Eydoux,‡
Jean-Claude Guillemot,‡ Johan Neyts,§ Leen Delang,§ David Touboul,† Fanny Roussi,†
and Marc Litaudon*,†

Institut de Chimie des Substances Naturelles, CNRS-ICSN, UPR 2301, Université Paris-Saclay, 91198, Gif-sur-Yvette, France

Aix Marseille University, CNRS, AFMB, AD2P, 163 Avenue de Luminy, 13288 Marseille Cedex 09, France
§
Laboratory for Virology and Experimental Chemotherapy, Rega Institute for Medical Research, KU Leuven, 3000 Leuven, Belgium
*
S Supporting Information
Downloaded from pubs.acs.org by IOWA STATE UNIV on 01/25/19. For personal use only.
J. Nat. Prod.

ABSTRACT: From a set of 292 Euphorbiaceae extracts, the use of a molecular networking (MN)-based prioritization
approach highlighted three clusters (MN1−3) depicting ions from the bark extract of Codiaeum peltatum. Based on their
putative antiviral potential and structural novelty, the MS-guided purification of compounds present in MN1 and MN2 afforded
two new daphnane-type diterpenoid orthoesters (DDO), codiapeltines A (1) and B (2), the new actephilols B (3) and C (4),
and four known 1,4-dioxane-fused phenanthrene dimers (5−8). The structures of the new compounds were elucidated by NMR
spectroscopic data analysis, and the absolute configurations of compounds 1 and 2 were deduced by comparison of
experimental and calculated ECD spectra. Codiapeltine B (2) is the first daphnane bearing a 9,11,13-orthoester moiety,
establishing a new major structural class of DDO. Compounds 1−8 and four recently reported monoterpenyl quinolones (9−
12) detected in MN3 were investigated for their selective activities against chikungunya virus replication and their
antipolymerase activities against the NS5 proteins of dengue and zika viruses. Compounds 3−8 exhibited strong inhibitory
activities on both dengue and zika NS5 in primary assays, but extensive biological analyses indicated that only actephilol B (3)
displayed a specific interaction with the NS5 targets.

R ecent outbreaks of chikungunya (CHIKV), dengue


(DENV), and zika (ZIKV) massive epidemics highlighted
the worldwide public health threat that emerging and
symptoms renders their differential diagnosis difficult.11,12 In
order to identify novel CHIKV inhibitors, several Euphorbia-
ceae species have been investigated in the past few years using
reemerging viruses represent.1 These viruses, which belong bioassay-guided purification procedures. This has resulted in
to the Alphavirus (CHIKV) and Flavivirus (ZIKV and DENV) the isolation of tigliane, jatrophane, flexibilane, daphnane, and
genera, are responsible for severe infectious diseases associated myrsinane diterpenoids showing micro- to nanomolar range
with fever, rashes, and headache.2,3 ZIKV is also associated activities.13−19 However, bioassay-guided fractionation has
with neurological disorders such as Guillain-Barre syndrome
several limitations. As a noteworthy example, its workflow,
and other congenital syndromes in newborns such as
which relies on bioactivity concerns only, does not take full
microcephaly.4−6 In Flaviviruses, the nonstructural protein 5
(NS5) polymerase plays an essential role in viral replication.7−9 advantage of the inherent structural diversity embedded in
Since DENV and ZIKV NS5 proteins harbor highly similar crude extract collections. Thus, this approach frequently leads
RdRp domains, these enzymes are considered as targets of to the reisolation of known bioactive compounds or analogues
particular interest in the prospect of discovering dual
inhibitors.10 The discovery of such dual inhibitors could be Received: September 20, 2018
particularly relevant given that the similarity of dengue and zika
© XXXX American Chemical Society and
American Society of Pharmacognosy A DOI: 10.1021/acs.jnatprod.8b00800
J. Nat. Prod. XXXX, XXX, XXX−XXX
Journal of Natural Products Article

Figure 1. Extract and compound prioritization strategy using multi-informative molecular networking: (A) Generation of a molecular network from
LC-MS2 analyses of 292 EtOAc extracts of New Caledonian Euphorbiaceae. (B) Detection of bioactive clusters by mapping CHIKV, DENV, and
ZIKV activities over networks. (C) Selected MN1, MN2, and MN3 with a taxonomical mapping consisting in overlaying the 20 Euphorbiaceae
genera represented in the extract collection to detect genus- or extract-specific clusters. Node sizes are proportional to MS1 peak area values.

and slows down the natural products (NPs) drug discovery dissimilarity within a taxonomically homogeneous set of
process. samples could imply chemical uniqueness, the generation of
Recently, we developed a molecular networking (MN)- these multi-informative maps unifying structural data,
based strategy consisting in deciphering the relationship taxonomical information, and bioassay results allows bio-
between spectral networks and biological activities and further activities to be associated with taxon-specific scaffolds. From a
exploiting it to prioritize the isolation of bioactive NPs.20,21 network made of approximately 1200 clusters and constructed
The core concept of this approach is based on the cross-linking from 292 LC-MS2 analyses of EtOAc Euphorbiaceae extracts,
of various information layers within a massive molecular this approach highlighted the original chemical composition of
network to spot bioactive scaffolds. Assuming that spectral Codiaeum peltatum, a species endemic from New Caledonia. A
B DOI: 10.1021/acs.jnatprod.8b00800
J. Nat. Prod. XXXX, XXX, XXX−XXX
Journal of Natural Products Article

Chart 1

first chemical investigation of this extract afforded four unusual percentage of inhibition at 50 μg·mL−1. For each target, the
chlorinated monoterpenyl quinolones (9−12).21 bioactivity results were first split up according to their activity
A careful analysis of the networks with CHIKV, DENV, and levels. The division into various bioactivity levels is much more
ZIKV bioactivity overlays revealed two additional clusters of informative than a simple active/nonactive categorization, as
interest (MN1 and MN2) associated with this C. peltatum bark has been shown in our previous study.20 The bioactive clusters
extract. This paper deals with the MS-guided purification of were highlighted by mapping these bioactivity degrees over
these compounds that led to the isolation of new bioactive networks using a color gradient applied to all nodes (Figure
daphnane-type diterpenoid orthoesters (DDO) (1 and 2) and 1B). CHIKV EC50 values were layered into five activity ranges
1,4-dioxane-fused phenanthrene dimers (3−8). (0 to 5; 5 to 10; 10 to 20; 20 to 100; and >100 μg·mL−1),

■ RESULTS AND DISCUSSION


A collection of 292 EtOAc extracts of New Caledonian
DENV NS5 percentages of inhibition were layered into five
groups (0 to 40; 40 to 50; 50 to 60; 60 to 80; and >80%),
whereas ZIKV NS5 percentages of inhibition were sliced into
Euphorbiaceae was first filtered over polyamide to remove five ranges (0 to 60; 60 to 70; 70 to 80; 80 to 90; and >90%).
polar compounds that might interact with biological assays and The significance levels of 40% for DENV NS5 and 60% for
induce false-positive responses. This set of samples was ZIKV NS5 roughly correspond to values above average plus
prepared from 107 different species, of which 93% are one standard deviation.
endemic. The extracts were prepared from leaves (47%), Starting from the whole Euphorbiaceae network showing
bark or twigs (48%), whole plants (3%), or fruits (2%). The 1231 constellations made of at least three nodes (Figure 1A),
Euphorbiaceae family was chosen for its large structural this first filtering step allowed the number of constellations of
diversity, characterized by unique classes of structurally diverse interest to be reduced to 27 for CHIKV, 28 for DENV, and 30
diterpenoids endowed with a wide range of biological activities, for ZIKV. The second step of the prioritization strategy
such as antiviral activity against human immunodeficiency consisted in targeting spectral uniqueness to detect uncommon
virus or modulation of P-glycoprotein and protein kinase secondary metabolites. Indeed, genus- or species-specific
C.22−29 compounds can be distinguished from other ubiquitous
The 292 samples were profiled by LC-HRMS2 and MS2 data metabolites in networks by using a taxonomical mapping.20,21
were organized as molecular networks to map the spectral Each Euphorbiaceae genus was assigned a specific color tag
space of the extract collection (Figure 1A). Concurrently, this (see Figure 1C), and this layout was used to highlight clusters
set was screened over three biological targets: (i) a CHIKV of nodes that would be restricted to a given taxon and thus a
cytopathogenic effect (CPE) inhibition assay affording EC50 given color.
values ranging from 0.02 to 100 μg.mL−1; (ii) a DENV NS5 This methodology stressed the bioactivity potential and the
inhibition assay affording a percentage of inhibition at 50 μg· original chemical composition of the EtOAc bark extract of
mL−1; and (iii) a ZIKV NS5 inhibition assay affording a Codiaeum peltatum (see Figure 1C). This extract was active
C DOI: 10.1021/acs.jnatprod.8b00800
J. Nat. Prod. XXXX, XXX, XXX−XXX
Journal of Natural Products Article

Figure 2. Key COSY (bold), HMBC (blue arrows), and NOESY or ROESY (red arrows) correlations for compounds 1 and 3.

toward the three targets studied and was thus selected for an 5-OH and 20-OH were supported by the downfield resonances
in-depth investigation. Among the high chemical diversity of of the C-5 methine (δH 4.18, s; δC 74.5) and C-20 methylene
this extract (Figure S1, Supporting Information) three groups. Correlations from 11-OH (δH 3.48, s) to C-11 (δC
constellations were spotted either for bioactivity (MN1) or 77.0) and C-18 (δC 24.6) revealed the presence of an unusual
bioactivity associated with a putative structural originality oxidation at C-11. Two benzoyloxy groups were located at C-3
(MN2 and MN3). and C-12, as supported by HMBC correlations from H-3 to C-
Besides sample prioritization, this multi-informative MN 1‴ (δC 168.1) and H-12 (δH 5.46, s) to C-1″ (δC 166.6). The
approach allows a targeted isolation of compounds of interest. three remaining oxygen atoms were assigned to C-9 (δC 84.3),
After a first semipreparative HPLC fractionation step directed C-13 (δC 86.1), and C-14 (δC 82.2). Eventually, the presence
toward the purification of MN1−3 associated compounds of the 9,13,14-orthobenzoate moiety was confirmed by the
traceable in the crude extract, all fractions obtained were downfield shifts of the latter together with HMBC correlations
analyzed in LC-MS2. As the amount of starting extract was from H-14 (δH 4.73, d) and H-3′/H-7′ (δH 7.79, m) to C-1′
relatively low (600 mg), only fractions showing ions detected (δC 118.2).
in high abundances and belonging to one of the targeted The relative configuration of 1 was confirmed by 1H NMR
clusters were selected for further isolation efforts. coupling constant values and key NOESY correlations as
The MN1 cluster displayed ions shared between bark depicted in Figure 2. The α-orientation of the 11-hydroxy
extracts of Codiaeum peltatum and C. oligogynum and bark and group was supported by the strong NOESY correlation
wood extracts of Trigonostemon cherrieri. This information between H3-18 (δH 1.54, s), H-12, and H-8 (δH 3.08, d).
together with the presence of chlorinated ions in the For establishing the absolute configurations, electronic circular
Trigonostemon subunit of MN1 allowed this cluster to be dichroism (ECD) calculations were carried out for compound
associated with DDO analogues.13,30,31 1 at the B3LYP/6-31G* level (Figure 3). Comparison of the
Compound 1 was obtained as a white, amorphous powder. experimental and calculated ECD spectra supported the
Its HRESIMS showed a protonated molecular ion peak at m/z absolute configuration of codiapeltine A (1)
727.2752 (calcd 727.2749), which, in conjunction with the 13C (2S,3S,4S,5S,6R,7S,8S,9R,10R,11R,12S,13R,14R).
NMR data, was consistent with the molecular formula Compound 2 showed a protonated molecular ion at m/z
C41H42O12. The 1H and 13C NMR data, along with the 727.2749 (calcd 727.2749) in the HRESIMS, corresponding to
HSQC spectrum, revealed three methyl groups, eight methine the molecular formula C41H42O12, the same as compound 1.
groups, of which five are oxygenated, three aromatic rings, two From this formula and similar spectroscopic data, it was
carbonyls, an exomethylene and an olefinic carbon, two apparent that 1 and 2 shared the same DDO core, but with a
methylene groups (one oxygenated), five oxygenated tertiary different C-ring substitution. Indeed, when compared with
carbons, and one deshielded sp3 trioxygenated carbon at δC compound 1, the upfield resonances of the oxygenated
118.2 typical of an orthobenzoate moiety.32 Additionally, two secondary carbon C-12 (δC 68.8) and oxygenated tertiary
protons showing no correlation in the HSQC spectrum were carbons C-13 (δC 81.8) and C-14 (δC 75.6) and the downfield
attributable to the exchangeable protons of two hydroxy resonances of the 8-methine and oxygenated tertiary carbons
groups. C-9 (δC 89.1) and C-11 (δC 84.1) of compound 2 suggested
The A−B−C rings of 1 can be constructed by comparison of that the orthobenzoate moiety is attached to different C-ring
NMR literature data and analysis of COSY and HMBC positions. From the HMBC correlation of H-12 (δH 5.24, s) to
correlations (Figure 2).33−35 The spin system connecting H2-1 carbonyl C-1″ (δC 166.6) it was confirmed that a benzoyloxy
(δH 2.07, m; 2.27, ddd), H-2 (δH 1.93, m), H-3 (δH 5.09, d), group is attached at C-12, as was the case in compound 1. In
H-10 (δH 3.12, dd), and H3-19 (δH 1.08, d) together with addition, strong COSY correlations between H-14 (δH 4.20,
HMBC correlations from H3-19 to C-1 (δC 34.7), C-2 (δC dd) and H-8 at δH 3.40 (d, J = 5.8 Hz) and a proton at δH 3.11
35.9), and C-3 (δC 81.9) and from 4-OH (δH 3.00, s) to C-4 (d, J = 12.5 Hz) showing no correlation in the HSQC
(δC 80.6) and C-10 (δC 48.2) indicated the presence of a spectrum, together with HMBC correlations from the latter
saturated cyclopentane oxygenated at C-3 for ring A. HMBC and C-8, C-13, and C-14, suggested the presence of a hydroxy
correlations from H-7 (δH 3.44, brs) to C-6 (δC 64.4), C-8 (δC group at C-14 (δC 75.6). These results and HMBC correlations
34.8), C-9 (δC 84.3), and C-20 (δC 66.6) and from H2-20 (δH from the 18-methyl protons to the deshielded carbons C-9 and
3.82, d; 3.87, d) to C-6 and C-7 (δC 64.4) confirmed the C-11 suggested that the orthobenzoate unit is attached at C-9,
presence of an epoxide moiety at C-6/C-7.34 The presence of C-11, and C-13. Coupling constants and NOESY correlations
D DOI: 10.1021/acs.jnatprod.8b00800
J. Nat. Prod. XXXX, XXX, XXX−XXX
Journal of Natural Products Article

Table 1. NMR Data for Compounds 1 and 2 (500 and 125


MHz, CDCl3)
1 2
pos. δC δH (J in Hz) δC δH (J in Hz)
1α 34.7 2.07 m 34.0 1.97 ma
1β 2.27 ddd (14.6, 13.2, 11.5) 2.20 m
2 35.9 1.93 m 36.0 1.99 ma
3 81.9 5.09 d (4.7) 82.7 4.98 ma
4 80.6 80.2
4-OH 3.00 s 3.06 s
5 74.5 4.18 s 73.9 4.19 sa
5-OH 3.94 ma
6 61.4 61.5
7 64.4 3.44 brs 62.9 3.58 s
8 34.8 3.08 d (2.7) 40.8 3.40 d (5.8)
9 84.3 89.1
10 48.2 3.12 dd (13.2, 6.8) 46.8 3.39 ma
11 77.0 84.1
11-OH 3.48 s
12 75.4 5.46 s 68.8 5.24 s
13 86.1 81.8
14 82.2 4.73 d (2.7) 75.6 4.20 ma
14-OH 3.11 d (12.5)
15 141.6 142.8
16-a 114.6 4.98 brs 114.4 4.97 brs
Figure 3. Experimental (black) and calculated (blue) ECD spectra of
codiapeltines A (1) and B (2) in MeOH. Vertical bars represent the 16-b 5.24 brs 5.19 brs
experimental Δε in L·mol−1·cm−1 (left) and the B3LYP/6-31G*- 17 19.9 1.78 s 19.5 1.75 s
computed rotational strengths in 10−40 cgs units (right). 18 24.6 1.54 s 18.1 1.65 s
19 13.5 1.08 d (6.7) 13.6 1.12 d (6.4)
20-a 66.6 3.82 d (12.3) 67.0 3.77 d (12.2)
observed for compound 2 were comparable to 1, allowing its
20-b 3.87 d (12.3) 3.93 d (12.2)
relative configuration to be depicted as shown. TDDFT
1′ 118.2 116.0
calculation of the ECD spectrum of 2 was conducted at the
2′ 134.8 135.4
B3LYP/6-31G* level. The absolute configuration of codia-
3′−7′ 126.6 7.79 m 126.3 7.82 m
peltine B (2) was defined as (2S,3S,4S,5S,6R,-
4′−6′ 128.5b 7.42 m 128.5 7.45 m
7S,8R,9R,10R,11R,12R,13R,14R) by comparison of its exper-
5′ 130.2 7.42 m 130.1 7.45 m
imental and calculated ECD curves (Figure 3).
1″ 166.6 166.6
Unlike all reported diterpenoids sharing the 5−7−6 tricyclic
2″ 129.4 129.5c
ring system, i.e., daphnane- and tigliane-type diterpenoids,
3″−7′′ 130.4 8.09 d (8.1) 130.3 8.04 d (8.0)
codiapeltines A (1) and B (2) bear an unusual but unique C-
4″−6′′ 128.6b 7.40 m 128.6d 7.40 m
ring 9,11,12,13,14-oxygenation pattern. Furthermore, com-
5″ 133.5 7.55 m 133.6 7.55 m
pound 2 is the first daphnane possessing a 9,11,13-orthoester
1‴ 168.1 168.4
linkage, establishing a new major structural class of
2‴ 129.4 129.6c
DDO.23,32,36
3‴−7″′ 129.8 7.97 d (8.1) 129.8 7.96 d (8.1)
The cluster MN2 was composed of ions found exclusively in
4‴−6″′ 128.9b 7.40 m 128.9d 7.40 m
bark extracts of the endemic C. oligogynum and C. peltatum
5‴ 133.9 7.55 m 133.9 7.55 m
species. Based on the molecular formula deduced from the four a b,c,d
protonated ions at m/z 581.219 and 609.253 of MN2, the Overlapping signals. Values are interchangeable.
known actephilol A (5), epi-actephilol A (6), fimbricalyx C
(7), and fimbricalyx D (8), which shared the original dioxane comparable to those of actephilol A (5) but revealed the
phenanthrene dimeric scaffold, were dereplicated and sub- presence of only one methoxy group and two additional
sequently isolated.37−39 Interestingly, these phenanthrene carbonyls at δC 178.6 and 182.4. The structure of compound 3
dimers showed characteristic absorption bands at 224, 247, may be divided in two different monomeric units, A and B
and 290 nm in their UV spectra. This typical UV profile has (Figure 1). While the 1H and 13C NMR chemical shifts of
also been observed for a third analogue (compound 3) that subunit A were close to those of compound 5, those of subunit
was not included in MN2 but appeared in a separated cluster B showed marked differences. The ortho-quinone motif was
in the massive MN representation. As shown in Figure S2, deduced through the observation of the key HMBC
Supporting Information, some spectral differences indicated correlations from H-8′ (δH 7.71, s) to C-6′ and a carbonyl
that 3 was not part of MN2. at δC 178.6 on one hand and from H3-11′ (δH 2.65, s) to C-1′,
Compound 3 was isolated as a brownish-red resin and was C-2′, C-10a′, and a carbonyl at δC 182.4, which established the
assigned the molecular formula C34H28O9 by analysis of its 9′,10′-dione function. Other HMBC correlations fully
positive HRESIMS (m/z 581.1796 [M + H]+, calcd for supported the proposed structure depicted in Figure 2 for
C34H29O9, 581.1806). The spectroscopic data of 3 were closely subunit B.
E DOI: 10.1021/acs.jnatprod.8b00800
J. Nat. Prod. XXXX, XXX, XXX−XXX
Journal of Natural Products Article

Table 2. NMR Data for Compounds 3 (600 and 150 MHz, DMSO-d6) and 4 (600 and 150 MHz, Methanol-d4)
3 (A unit) 3 (B unit) 4 (A unit) 4 (B unit)
position δC δH position δC δH position δC δH position δC δH
1 47.2 1′ 132.1 1 48.8 1′ 120.3
2 204.2 2′ 140.6 2 207.0 2′ 141.6
3 96.7 3′ 146.3 3 98.5 3′ 141.4
3-OMe 52.1 3.42 s 4′ 110.1 7.23 s 3-OMe 53.0 3.49 s 4′ 108.2 7.74 s
4 72.5 5.71 s 4′a 121.7 4 74.5 5.80 s 4′a 120.7
5 112.1 4′b 135.9 5 116.2 4′b 132.6
6 142.8 5′ 109.7 7.20 s 6 144.6 5′ 107.2 7.64 s
7 103.3 6.74 s 6′ 164.1 7 105.8 6.79 s 6′ 156.8
8 155.2 7′ 126.3 8 156.3 7′ 126.5
9 118.1 8′ 132.1 7.71 s 9 120.5 8′ 125.7 7.93 s
10 123.8 7.93 s 8′a 131.2 10 125.5 8.01 s 8′a 122.2
11 126.0 9′ 178.6 11 128.2 9′ 154.3
12 156.4 9′-OMe 12 159.9 9′-OMe 56.2 4.02 s
12-OMe 10′ 182.4 12-OMe 56.5 3.84 s 10′ 96.8 6.83 s
13 104.0 7.38 s 10′a 124.8 13 101.4 7.35 s 10′a 129.5
14 133.6 11′ 13.4 2.65 s 14 135.4 11′ 11.8 2.61 s
15 28.7 1.35 s 12′ 15.5 2.13 s 15 30.0 1.46 s 12′ 16.9 2.34 s
16 27.9 1.58 s 16 28.9 1.65 s
17 16.7 2.32 s 17 17.1 2.34 s

Table 3. Antimetabolic and Antiviral Activities of Compounds 1−12 in Vero Cells against CHIKV and Their Inhibitory
Activities against RNA-Dependent RNA NS5 of Dengue and Zika Viruses
CHIKV ZIKV DENV
a
compound CC50 (Vero) EC50a SI b
IC50a IC50a
1 52 ± 12 10.0 ± 2.3 5 >69 >69
2 49 ± 17 4.4 ± 0.5 11 >69 >69
3 >172 >172 8.1 ± 0.9 12.1 ± 1.9
4 ndc nd 10.9 ± 2.5 14.5 ± 2.2
5 >172 >86 4.8 ± 0.9 4.5 ± 0.7
6 >129 >172 8.1 ± 1.0 9.1 ± 1.2
7 nd nd 13.6 ± 1.2 21.7 ± 2.0
8 nd nd 12.7 ± 1.3 17.8 ± 3.1
9 >208 >277 >138 >119
10 >288 >288 nd nd
11 >239 >239 >138 >119
12 >277 >277 nd nd
chloroquine 89 ± 28 11 ± 7 8
3′dATP 0.14 ± 0.01 0.07 ± 0.005
a
Data in μM. Values are the median ± absolute deviation calculated from at least three independent assays. bSI or window for antiviral selectivity is
calculated as CC50 Vero/EC50 CHIKV. cnd = not determined.

The relative configuration of compound 3 was established ROESY correlation between 3-OMe (δH 3.49, s) and H-4 (δH
from the comparison of NMR data with those of compound 5 5.80, s).
and ROESY analysis (Figure 2). The cis-configuration of the C- As previously reported, the MS-guided purification of
3−C-4 ring junction was confirmed by a strong ROESY compounds displayed in MN3 afforded the first naturally
correlation between 3-OMe (δH 3.42, s) and H-4 (δH 5.71, s). occurring monoterpenyl quinolin-4-one isochloroaustralasine
Actephilol B (3) is the seventh member of the 1,4-dioxane- A (12) along with three exceptionally rare chlorinated
fused phenanthrene dimer family.37 monoterpenyl quinolin-2-ones, chloroaustralasines A−C (9−
Compound 4, displayed in MN2 (Figure 1C), was isolated 11, respectively).21
as a brownish-red resin, and its HRESIMS data showed a Compounds 1−12 were evaluated for their antiviral activity
protonated ion at m/z 595.2316 (calcd for C36H35O8, in a CHIKV cell-based assay (CPE inhibition) and against
595.2326). The spectroscopic data of compound 4 were DENV- and ZIKV-NS5 (Table 3). In the CHIKV assay,
close to those of actephilol A (5) and epi-actephilol A (6). compounds 1 and 2 showed anti-CHIKV activities compared
However, a strong HMBC correlation between a methyl group, to the reference compound, chloroquine, with EC50 values of
MeO-12 (δH 3.84, s), and C-12 (δC 159.9) indicated the 10 ± 2.3 μM (SI = 5) and 4.4 ± 0.5 μM (SI = 11),
presence of an additional methoxy in position 12. The relative respectively. Although the anti-CHIKV activity of DDO
configuration of actephilol C (4) was supported by the strong derivatives had already been reported,13,30,31 these results
F DOI: 10.1021/acs.jnatprod.8b00800
J. Nat. Prod. XXXX, XXX, XXX−XXX
Journal of Natural Products


Article

confirmed the predictions made from the multi-informative EXPERIMENTAL SECTION


MN approach used in this study. General Experimental Procedures. Optical rotations were
In the DENV and ZIKV assays, results also met our measured at 25 °C on an Anton Paar MCP 300 polarimeter. UV
expectations, as compounds 3−8 exhibited strong inhibitory spectra were recorded on a Varian Cary 100 UV−vis spectropho-
activities, although ∼100 times less potent than the reference tometer. ECD spectra were measured at 25 °C on a JASCO J-810
compound, 3′-dATP, on both enzymes with IC50 values in the spectropolarimeter. NMR spectra were recorded on a Bruker 500
μM range. To confirm these results, two series of experiments MHz instrument (Avance 500) for compounds 1, 2, 7, and 8, on a
Bruker 600 MHz instrument (Avance 600) for compounds 3 and 4,
were conducted. First, the IC50 values of compounds 3−8 were
and on a Bruker 300 MHz instrument (Avance 300) for compounds 5
determined in the presence of detergent, Triton X-100, at and 6. Chemical shifts are in ppm, and coupling constants are in Hz.
0.01% final concentration. Using a detergent in the assay below Kinetex analytical and semipreparative C18 columns (250 × 4.6 mm
the critical micellar concentration indicates whether a and 250 × 10 mm; 5 μm Phenomenex) and a Nucleodur PFP
compound is a potential aggregator for one or more partners semipreparative column (250 × 10 mm; 5 μm Macherey-Nagel) were
of the assay. Thus, a nonspecific inhibition would induce an used for HPLC separations using a Dionex system equipped with a
increase of the IC50 value.40 Under these conditions sample manager (Gilson 215 liquid handler), a column fluidics
organizer, a binary pump (Dionex HPG-3200BX), a UV−vis diode
compounds 4−8 lost their inhibitory potential (IC50 > 100
array detector (190−600 nm, Dionex UVD340U), and a PL-ELS
μM) on both NS5 proteins, while compound 3 showed IC50 1000 ELS Polymer Laboratory detector. HPLC separations were also
values of 36.0 ± 4.7 and 67.2 ± 20.0 μM on ZIKV-NS5 and undertaken using a Waters Alliance system equipped with a binary
DENV-NS5, respectively. pump (Waters 2525), a UV−vis diode array detector (190−600 nm,
Using an orthogonal assay based on fluorescence polar- Waters 2996), and an ELS detector (Waters 2424). Disconnected
ization, the apparent Kd values of these compounds on ZIKV- from ELSD, this system was also used in preparative mode to purify
NS5 and DENV-NS5 were determined. This assay shows small amounts of products. All solvents were purchased from Carlo
Erba (France) and SDS (Peypin, France).
whether a compound can have a specific interaction with the
Data-Dependent LC-ESI-HRMS2 Analysis. LC analyses were
enzyme by measuring its potential to shift the interaction performed with a Dionex Ultimate 3000 RSLC system equipped with
between the GTP bodipy (boron-dipyrromethene) and the an Accucore C18 column (2.1 × 100 mm; 2.6 μm, ThermoScientific).
protein. Compounds 4−8 showed no fixation on the enzyme The mobile phase consisted of H2O−MeCN with 0.1% formic acid
(Kdapp > 100 μM). These results, correlated with the loss of (20:80) during 5 min, then a gradient from 20:80 to 100:0 in 20 min,
inhibition in the detergent assays, confirmed that these at a flow rate of 350 μL·min−1. The temperature of the column oven
compounds act as aggregators and not inhibitors. was set at 30 °C and the injection volume at 5 μL.
LC-ESI-HRMS2 analyses were achieved by coupling the LC system
However, compound 3 showed a Kdapp of 32.1 ± 4.5 and 9.3
to an Agilent 6540 hybrid quadrupole TOF mass spectrometer
± 3.6 μM on ZIKV-NS5 and DENV-NS5, respectively. It (Agilent Technologies, Massy, France) equipped with an ESI dual
agreed with the detergent assays and indicated a specific source, operating in positive-ion mode. Source parameters were set as
interaction toward the NS5 targets. Taken together, these follows: capillary temperature at 325 °C, source voltage at 3500 V,
results indicate that actephilol B (3) is the first natural dual sheath gas flow rate at 7 L.min−1, nebulizer pressure at 30 psi, drying-
inhibitor of ZIKV and DENV NS5. gas flow rate at 10 L·min−1, drying-gas temperature at 350 °C, stealth-
Time and money spent in tedious bioassay-guided gas temperature at 350 °C, skimmer voltage at 45 V, fragmentor
fractionation and isolation processes represent major bottle- voltage at 150 V, and nozzle voltage at 500 V.
MS scans were operated in full-scan mode from m/z 100 to 1000
necks in NP drug discovery. Here, we reported an example of (0.1 s scan time) with a mass resolution of 20 000 at m/z 922. An
the multi-informative MN methodology set up to reduce MS1 scan was followed by MS2 scans of the five most intense ions
workload and associated costs of the NP hit detection above an absolute threshold of 3000 counts. Selected parent ions were
workflow. This strategy facilitated the extract prioritization fragmented with collision energy fixed at 35 eV and an isolation
step by highlighting the original chemical composition of C. window of 1.3 amu.41
peltatum and also permitted a straightforward MS-guided Calibration solution, containing two internal reference masses
purification of targeted compounds. The study of the C. (purine, C5H4N4, m/z 121.0509, and HP-921 [hexakis(1H,1H,3H-
tetrafluoropentoxy)phosphazene], C18H18O6N3P3F24, m/z 922.0098),
peltatum bark extract led to the characterization of three routinely led to mass accuracy below 2 ppm. LC-UV data were
different chemical series. Among the compounds isolated, analyzed with Chromeleon software (Dionex), and MS data
codiapeltine B (2) is the first daphnane bearing a 9,11,13- acquisition and processing were performed using MassHunter
orthoester moiety, establishing a new major structural class of Workstation software (Agilent Technologies, Massy, France).
daphnane-type diterpenoid orthoesters. Moreover, actephilol B MZmine 2 Data-Preprocessing Parameters. Networks were
(3), a member of the rare 1,4-dioxane-fused phenanthrene generated using the feature-finding-based methodology previously
dimer family, has been found as the first natural dual inhibitor described.42 The 292 MS2 data files were converted from the .d
(Agilent) standard data format to .mzXML format using the
of DENV and ZIKV NS5 yet characterized. MSConvert software, part of the ProteoWizard package.43 All
Without any prior chemotaxonomic knowledge or biblio- .mzxml were processed using MZmine 2 v31.44 The mass detection
graphic preconceived assumptions, the multi-informative MN was realized keeping the noise level at 0. The ADAP chromatogram
prioritization approach allows bioactive scaffolds to be spotted. builder was used using a minimum group size of scans of 4, a group
As a complement to bioactivity observations, the taxonomical intensity threshold of 3000, a minimum highest intensity of 4000, and
input pinpoints genus- or species-specific metabolites. m/z tolerance of 0.005 (or 20 ppm).45 The ADAP wavelets
Combining biological and chemical profiling results in an deconvolution algorithm was used with the following standard
settings: S/N threshold = 8, minimum feature height = 4000,
MN environment enables a clear view of the structural breadth coefficient/area threshold = 20, peak duration range 0.05−1 min, tR
and distribution within taxonomically homogeneous extract wavelet range 0.01−0.07 min. However, these parameters needed to
collections and drastically increases the chances of discovering be reoptimized for a few specific samples in this data set. MS2 scans
new lead compounds. were paired using an m/z tolerance range of 0.02 Da and tR tolerance

G DOI: 10.1021/acs.jnatprod.8b00800
J. Nat. Prod. XXXX, XXX, XXX−XXX
Journal of Natural Products Article

range of 0.5 min. Isotopologues were grouped using the isotopic peak [M + H] + (calcd for C 34 H 29 O 9 , 581.1806); GNPS ID:
grouper algorithm with an m/z tolerance of 0.005 (or 20 ppm) and a CCMSLIB00004681457 ([M + H]+).
tR tolerance of 0.2 min. Peak alignment was performed using the join Actephilol C (4): brownish-red resin; [α]24D +8 (c 0.1, MeOH);
aligner module [m/z tolerance = 0.004 (or 10 ppm), weight for m/z = UV (MeOH) λmax (log ε) 250 (5.13), 289 (4.86), 317 (4.49) nm; 1H
2, weight for tR = 1, absolute tR tolerance 0.5 min]. The peak list was and 13C NMR, see Table 2; HRESIMS m/z 595.2316 [M + H]+
gap-filled with the same tR and m/z range gap filler module [m/z (calcd for C36H35O8, 595.2326); GNPS ID: CCMSLIB00004681458
tolerance of 0.004 (or 10 ppm)]. ([M + H]+).
Eventually, the .mgf preclustered spectral data file and its Actephilol A (5): brownish-red resin; HRESIMS m/z 581.2179 [M
corresponding .csv metadata file (for tR, areas, and formulas + H ] + ( c a lc d f o r C 3 5 H 3 3 O 8 , 5 8 1 .2 1 70 ) ; G N P S I D :
integration) were exported using the dedicated “Export for GNPS” CCMSLIB00004681459 ([M + H]+).
and “Export to CSV file” built-in options. See further documentation Epi-actephilol A (6): brownish-red resin; HRESIMS m/z 581.2176
and tutorials at https://ccms-ucsd.github.io/GNPSDocumentation/ [M + H] + (calcd for C 35 H 33 O 8 , 581.2170); GNPS ID:
featurebasedmolecularnetworking/. CCMSLIB00004681460 ([M + H]+).
Molecular Network Analysis. The molecular networks were Fimbricalyx C (7): brownish-red resin; HRESIMS m/z 609.2467
created using the online workflow at Global Natural Products Social [M + H] + (calcd for C 37 H 37 O 8 , 609.2483); GNPS ID:
molecular networking (http://gnps.ucsd.edu).46 MS2 spectra were CCMSLIB00004681461 ([M + H]+).
window-filtered by choosing only the top six peaks in the ±50 Da Fimbricalyx D (8): brownish-red resin; HRESIMS m/z 609.2466
window throughout the spectrum. The data were not clustered with [M + H] + (calcd for C 37 H 37 O 8 , 609.2483); GNPS ID:
MS-Cluster. A network was created where edges were filtered to have CCMSLIB00004681462 ([M + H]+).
a cosine score above 0.7 and at least four matching peaks. Further ECD Calculation. Computational calculations were performed
edges between two nodes were kept in the network if and only if each using the Gaussian ’09 software package, revision D01.47 Conforma-
of the nodes appeared in each other’s respective top 10 most similar tional analyses and geometry optimizations were performed at the
nodes. The library spectra were filtered in the same manner as the B3LYP/6-31G* level in MeOH. The ECD calculations were
input data. All matches kept between network spectra and library performed using TDDFT at the B3LYP/6-31G* level in MeOH.
spectra were required to have a score above 0.7 and at least 4 matched The ECD spectra were obtained by weighing the Boltzmann
peaks. distribution rate of each geometric conformation with a bandwidth
Plant Material. Stem and bark of Codiaeum peltatum were σ of 0.3 eV. Conformer structures of 1 and 2 and their optimized
collected at “Mont Aoupinié” (New Caledonia) at an altitude of 487 coordinates at the B3LYP/6-31G* level in MeOH are available in
m in November 2001 and authenticated by V. Dumontet. A voucher Figures S3 and S4, Supporting Information.
specimen (DUM-0109) has been deposited at the Herbier IRD de Virus-Cell-Based Anti-Chikungunya Assay. The antiviral
Nouméa, New Caledonia. activity of the extracts and pure compounds against Chikungunya
virus was determined using an MTS [3-(4,5-dimethylthiazol-2-yl)-5-
Extraction and Isolation. The plant material (200 g, dry wt) was
(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium]-based
extracted with EtOAc (3 × 0.5 L each). The EtOAc extract was
cytopathic effect reduction assay. Chikungunya virus (Indian Ocean
concentrated in vacuo at 40 °C to yield 1.5 g of residue. This extract
strain 899), kindly provided by Prof. C. Drosten (Institute of
was dissolved in MeCN (0.25 L) and subjected to a liquid/liquid
Virology, University of Bonn, Germany), was grown on Vero (African
partition with n-heptane (2 × 0.25 L) to afford 0.6 g of an MeCN-
green monkey kidney) cells. Antiviral assays were performed in
soluble fraction. This fraction was subjected to a semipreparative
DMEM medium [MEM Rega3 (cat. no. 19993013; Invitrogen), 2%
HPLC (Kinetex C18, H2O−MeCN 35:65 + 0.1% formic acid to 0:100 FCS (Integro), 5 mL of 200 mM L-glutamine, and 5 mL of 7.5%
in 45 min at 4.5 mL·min−1) to afford 20 fractions of decreasing NaHCO3]. Cells were seeded in a 96-well plate (Falcon, BD
polarity, F1−F20. Purification of fraction F12 (9.2 mg) by Biosciences) and allowed to adhere overnight. Serial dilutions of the
semipreparative HPLC (Nucleodur PFP, H2O−MeCN 45:55 + test compounds were made in 100 μL of assay medium in a 96-well
0.1% formic acid at 4.5 mL·min−1) afforded compounds 1 (2.5 mg) microtiter. Subsequently, 50 μL of a 4× virus dilution in assay
and 2 (1.5 mg) (tR: 17.5 and 15.5 min, respectively). Fraction F7 (9.1 medium was added (MOI 0.01), followed by 50 μL of a cell
mg) was purified by semipreparative HPLC (Nucleodur PFP, H2O− suspension. This suspension, with a cell density of 25 000 cells/50 μL,
MeCN 50:50 + 0.1% formic acid at 4.5 mL·min−1) to afford was prepared from a Vero A cell line subcultured in cell growth
compound 3 (1.1 mg) (tR: 18.0 min). Fraction F5 (17.4 mg) was medium (MEM Rega3, supplemented with 10% FCS, 5 mL of L-
purified by semipreparative HPLC (Nucleodur PFP, H2O−MeCN glutamine, and 5 mL of NaHCO3) in a 1:4 ratio. The assay plates
30:70 + 0.1% formic acid at 4.5 mL.min−1) to afford compound 4 (1.4 were returned to the incubator (37 °C, 5% CO2, 95−99% relative
mg) (tR: 13.4 min). Fraction F9 (20.9 mg) was subjected to a humidity) for 5 days, a time at which maximal virus-induced cell death
semipreparative HPLC (Nucleodur PFP, H2O−MeCN 45:55 + 0.1% or CPE is observed in untreated, infected controls. Subsequently, the
formic acid at 4.5 mL.min−1) to afford compound 5 (2.9 mg) and assay medium was aspirated, replaced with 75 μL of a 5% MTS
compound 6 (4.1 mg) (tR: 18.5 and 20.5 min, respectively). Fraction (Promega) solution (MTS-phenazinemethosulfate (MTS/PMS)
F4 (27.8 mg) was purified by semipreparative HPLC (Nucleodur stock solution [(2 g·L−1 MTS (Promega, Leiden, The Netherlands)
PFP, H2O−MeCN 18:82 + 0.1% formic acid at 4.5 mL·min−1) to and 46 mg·L−1 PMS (Sigma−Aldrich, Bornem, Belgium) in PBS at
afford compounds 7 (0.62 mg) and 8 (0.47 mg) (tR: 11.3 and 12.0 pH 6−6.5)], diluted 1/20 in MEM (Life Technologies, Gent,
min, respectively). Belgium) in phenol-red-free medium, and incubated for 1.5 h.
Codiapeltine A (1): white, amorphous powder; [α]24D −53 (c 0.1, Absorbance was measured at a wavelength of 498 nm (Safire2,
MeOH); UV (MeOH) λmax (log ε) 230 (4.44) nm; 1H and 13C Tecan), with the optical densities (OD values) reaching 0.6−0.8 for
NMR, see Table 1; HRESIMS m/z 727.2752 [M + H]+ (calcd for the untreated, uninfected controls. The optical density values were
C41H43O12, 727.2749); GNPS ID: CCMSLIB00004681455 ([M + converted to control percentages, and logarithmic interpolation was
H]+). used to calculate the EC50 and CC50. The EC50 was defined as the
Codiapeltine B (2): white, amorphous powder; [α]24D −43 (c 0.1, compound concentration that protected 50% of cells from virus-
MeOH); UV (MeOH) λmax (log ε) 230 (4.46) nm; 1H and 13C induced CPE. Adverse effects of the drug on the host cell were also
NMR, see Table 1; HRESIMS m/z 727.2749 [M + H]+ (calcd for assessed by means of the MTS method, by exposing uninfected cells
C41H43O12, 727.2749); GNPS ID: CCMSLIB00004681456 ([M + to the same concentrations of the compounds. The CC50 was defined
H]+). as the compound concentration that reduced the number of viable
Actephilol B (3): brownish-red resin; [α]24D −9 (c 0.1, MeOH); cells by 50%. All assay conditions producing an antiviral effect that
UV (MeOH) λmax (log ε) 224 (4.45), 247 (4.40), 290 (4.31), 337 exceeded 50% were checked microscopically for signs of a cytopathic
(3.86) nm; 1H and 13C NMR, see Table 2; HRESIMS m/z 581.1796 effect or adverse effects on the host cell (i.e., altered cell or monolayer

H DOI: 10.1021/acs.jnatprod.8b00800
J. Nat. Prod. XXXX, XXX, XXX−XXX
Journal of Natural Products Article

morphology). A sample was considered to elicit a selective antiviral (av(neg) − av(pos)). Compounds leading to a 70% inhibition or
effect on virus replication only when, following microscopic quality more at 50 μg·mL−1 and presenting a dose/response effect were
control, at least at one concentration no CPE or any adverse effect qualified as hits. Selected hits were then confirmed in triplicate, and
was observed (image resembling untreated, uninfected cells). The IC50 values determined only on pure compounds.
antiviral experiments have been performed in a biosafety screening IC50 Determination Based on Fluorescent Picogreen Assay. The
facility that has been validated for handling of Chikungunya virus as concentrations of the compounds leading to 50% inhibition of NS5-
well as the manipulation of molecules of unknown chemical safety mediated RNA synthesis were determined in IC50 buffer (50 mM
risk. All studies have been performed by trained staff. HEPES pH 8.0, 10 mM KCl, 2 mM MnCl2, 2 mM MgCl2, 10 mM
DENV and ZIKV Assays. Products and Reagents. Homopoly- DTT) containing 100 or 40 nM homopolymeric uridine RNA
meric uridine (Poly U) RNA template was obtained from GE template for Zika NS5 and DENV-2 NS5, respectively, seven various
Healthcare. Natural extracts or pure compounds were resuspended at concentrations of compound, and 40 nM of enzyme. For the IC50
10 or 1 mg·mL−1 in 100% DMSO and stored at −20 °C. The Quant-it determination in the presence of detergent, Triton X100 was added at
Picogreen dsDNA assay kit (ref P11496) was obtained from 0.01% final concentration in the assay. Five ranges of inhibitor were
Molecular Probes-Invitrogen. available (0.01 to 5 μM; 0.1 to 50 μM; 0.5 to 50 μM; 1 to 100 μM; 5
Expression and Purification of Zika and Dengue 2 NS5 to 400 μM). According to the inhibitory potency of the compound
Recombinant Proteins. The viral NS5 proteins of Zika and Dengue tested, a range was selected to obtain a more accurate IC50 based on
serotype 2 (DENV-2) virus coding sequences were cloned in fusion the best repartition of the points surrounding the range. Reactions
with a N-terminus hexahistidine tag in gateway plasmids (pQE30). were conducted in 40 μL volume on a 96-well Nunc plate. All
The proteins were expressed in E. coli cells and purified following a experiments were robotized by using a BioMek 4000 Automate
previously described protocol.48 (Beckman). A 2 μL amount of each diluted compound in 100%
Determination of the Inhibitory Potential of Compounds by a DMSO was added to the wells to the chosen concentration (5%
Picogreen Assay on Zika and Dengue NS5. Principle of the Assay. DMSO final concentration). For each assay, the enzyme mix was
A picogreen kinetic assay was based on polymerase activity of NS5
distributed in wells. Reactions were started by the addition of the
protein, which catalyzes de novo reaction of a poly(U) template and
nucleotide mix (100 and 80 μM ATP for Zika NS5 and DENV-2
adenosine triphosphate (ATP). The reaction was carried out at 30 °C
NS5, respectively) and were incubated at 30 °C for 10 min. Reaction
with 50 mM Hepes pH 8, 10 mM DTT, 10 mM KCl, 2 mM MgCl2, 2
assays were stopped by the addition of 20 μL of EDTA (100 mM).
mM MnCl2, and the selected concentrations in substrate (Poly U and
Positive and negative controls consisted respectively of a reaction mix
ATP) and enzyme. Each reaction assay was robotized on a Biomek
with 5% DMSO final concentration or EDTA (100 mM) instead of
4000 in 40 μL final volume for screening assay and IC50. Control
compounds. Reaction mixes were then transferred to a Greiner plate
plates, using only DMSO at 5% final concentration instead of
using a Biomek I5 Automate (Beckman). Picogreen fluorescent
compounds, were used to determine the Z′ factor, the background,
and the gain value of the assay. Quality of measurements was assessed reagent was diluted to 1/800 in TE buffer according to the
by calculating the Z′ factor for each plate: Z′ factor = 1 − ([3SD(neg) manufacturer’s data, and 60 μL of reagent was distributed into each
+ 3SD(pos)]/[av(neg) − av(pos)]), where SD(neg) and SD(pos) well of the Greiner plate. The plate was incubated for 5 min in the
stand for the standard deviation obtained for negative and positive dark at room temperature, and the fluorescence signal was read at 480
controls, respectively, and where av(neg) and av(pos) are averages for nm (excitation) and 530 nm (emission) using a TecanSafire2. The
negative and positive controls, respectively. Assay plates contained IC50 value was determined using the following equation: % of active
positive and negative controls distributed in the first and the last enzyme = 100/(1 + I2/IC50), where I is the concentration of inhibitor
columns, respectively. Positive and negative controls consisted of the and 100% activity is the fluorescence intensity without inhibitor. IC50
reaction mixture supplemented by ATP and 5% DMSO or 20 mM was determined from curve-fitting using Prism software. For each
EDTA, respectively. In the case of the screening assay, 3′dATP was value, results were obtained using triplicates in a single experiment.
also used as an internal control in the last column at 10 μM final Apparent Kd Determination of Compounds by a Fluorescence
concentration. Time incubation (from 10 to 30 min), substrate Polarization Assay. The concentration of compounds leading to a
concentrations (poly U and ATP), and enzyme concentration were shifting of the fixation of GTP bodipy to the D2 or Zika NS5 protein
optimized, based on the previous experimental results to obtain the was determined in the binding buffer (50 mM Tris pH 7.5, 50 mM
best reproducibility (Z′ > 0.6; background <10%, gain value <120). NaCl, 0.01% NP40, 1 mM DTT) containing 50 nM GTP bodipy, 300
Screening Assay Determination Based on the Fluorescent nM D2 NS5, or 150 nM Zika NS5 and seven various concentrations
Picogreen Assay. The assay was organized in 96-well Nunc plates, of compound. Reactions were conducted in 50 μL volume on a black
each containing 80 samples that are natural extracts or pure 96-half-well Greiner plate. Each diluted compound (2.5 μL) in 100%
compounds. Each of 80 samples was added to the assay using a DMSO was added in plate wells to the chosen concentration (5%
BioMek I5 workstation (Beckman) to a final concentration of 50, 10, DMSO final concentration). For each assay, 10 μL of a 5×
or 1 μg·mL−1 and/or 50, 10, and 1 μM in 5% DMSO. concentration of the enzyme was distributed in plate wells. Reactions
Reactions were conducted in 40 μL final volume. For each assay, 20 were started by the addition of the GTP bodipy (10 μL of a 5×
μL of the enzyme mix was first distributed in plate wells using a concentration) and were incubated at room temperature for 15 min.
Biomek 4000 (Beckman), containing 2 μL of the samples. Reactions Total fluorescence and fluorescence polarization were measured using
were initiated by addition of 18 μL of the nucleotide mix (100 and 80 a microplate reader (Pherastar) with a fluorescence polarization
μM ATP for Zika NS5 and DENV-2 NS5, respectively) and incubated module (485 nm excitation and 520 emission wavelengths).
at 30 °C for 20 and 30 min for Zika NS5 and DENV-2 NS5, Instrument settings were fixed as follows: 100mP target mP, 200
respectively. Assays were stopped by the addition of 20 μL of EDTA flashes per well, 0.5 s settling time. Focus and gain values were
(100 mM). adjusted prior to measurement.
Reaction mixes were transferred to a Greiner plate using a Biomek The positive control consisted of a reaction mix with 5% DMSO
I5 Automate (Beckman). Picogreen fluorescent reagent was diluted to final concentration in place of the compound. GTP bodipy alone (n =
1/800 in TE buffer (10 mM Tris-HCl, 1 mM EDTA, pH 7.5, with no 8) was used to determine the minimal fluorescence polarization
NaCl added) according to manufacturer’s data, and 60 μL of reagent intensity, and the average of this background value was subtracted
was distributed into each well of the Greiner plate. The plate was from each value of the assay. The fluorescence polarization of the
incubated for 5 min in the dark at room temperature, and the positive control is 100% fixation, and the residual fluorescence
fluorescence signal was read at 480 nm (excitation) and 530 nm polarization signal (mP %) is determined for each concentration of
(emission) using a TecanSafire2. the compound. The apparent Kd (Kdapp) of the compound was
For each compound, the percentage of inhibition was calculated as determined from curve-fitting using Prism software. For each value,
follows: Inhibition % = 100(raw_data_of_compound − av(pos)/ results were obtained using triplicates in a single experiment.

I DOI: 10.1021/acs.jnatprod.8b00800
J. Nat. Prod. XXXX, XXX, XXX−XXX
Journal of Natural Products


Article

ASSOCIATED CONTENT (9) Coutard, B.; Barral, K.; Lichière, J.; Selisko, B.; Martin, B.;
Aouadi, W.; Lombardia, M. O.; Debart, F.; Vasseur, J.-J.; Guillemot, J.
*
S Supporting Information
C.; Canard, B.; Decroly, E. J. Virol. 2017, 91, DOI: 10.1128/
The Supporting Information is available free of charge on the JVI.02202-16.
ACS Publications website at DOI: 10.1021/acs.jnat- (10) Godoy, A. S.; Lima, G. M. A.; Oliveira, K. I. Z.; Torres, N. U.;
prod.8b00800. Maluf, F. V.; Guido, R. V. C.; Oliva, G. Nat. Commun. 2017, 8,
1
H, 13C, HSQC, HMBC, COSY, and ROESY NMR 14764−14764.
spectra and HRESIMS data for compounds 1−8; (11) Göertz, G. P.; Vogels, C. B. F.; Geertsema, C.; Koenraadt, C. J.
M.; Pijlman, G. P. PLoS Neglected Trop. Dis. 2017, 11, e0005654.
conformer structures of 1 and 2 and their optimized (12) Dupont-Rouzeyrol, M.; O’Connor, O.; Calvez, E.; Daurès, M.;
coordinates at the B3LYP/6-31G* level in MeOH John, M.; Grangeon, J.-P.; Gourinat, A.-C. Emerging Infect. Dis. 2015,
(PDF) 21, 381−382.


(13) Allard, P.-M.; Leyssen, P.; Martin, M.-T.; Bourjot, M.;
AUTHOR INFORMATION Dumontet, V.; Eydoux, C.; Guillemot, J.-C.; Canard, B.; Poullain,
C.; Guéritte, F.; Litaudon, M. Phytochemistry 2012, 84, 160−168.
Corresponding Author (14) Bourjot, M.; Delang, L.; Nguyen, V. H.; Neyts, J.; Guéritte, F.;
*Tel: +33 1 69 82 30 85. E-mail: marc.litaudon@cnrs.fr. Leyssen, P.; Litaudon, M. J. Nat. Prod. 2012, 75, 2183−2187.
ORCID (15) Olivon, F.; Palenzuela, H.; Girard-Valenciennes, E.; Neyts, J.;
David Touboul: 0000-0003-2751-774X Pannecouque, C.; Roussi, F.; Grondin, I.; Leyssen, P.; Litaudon, M. J.
Nat. Prod. 2015, 78, 1119−1128.
Fanny Roussi: 0000-0002-5941-9901 (16) Corlay, N.; Delang, L.; Girard-Valenciennes, E.; Neyts, J.;
Marc Litaudon: 0000-0002-0877-8234 Clerc, P.; Smadja, J.; Guéritte, F.; Leyssen, P.; Litaudon, M.
Notes Fitoterapia 2014, 97, 87−91.
The authors declare no competing financial interest. (17) Esposito, M.; Nothias, L.-F.; Retailleau, P.; Costa, J.; Roussi, F.;


Neyts, J.; Leyssen, P.; Touboul, D.; Litaudon, M.; Paolini, J. J. Nat.
ACKNOWLEDGMENTS Prod. 2017, 80, 2051−2059.
(18) Nothias-Scaglia, L.-F.; Gallard, J.; Dumontet, V.; Roussi, F.;
The authors are grateful to the North Province of New Costa, J.; Iorga, B.; Paolini, J.; Litaudon, M. J. Nat. Prod. 2015, 78,
Caledonia, which has facilitated our field investigation, and V. 2423−2431.
Dumontet for his contribution to plant material identification (19) Nothias-Scaglia, L.-F.; Retailleau, P.; Paolini, J.; Pannecouque,
and species collection. We thank C. Collard and C. C.; Neyts, J.; Dumontet, V.; Roussi, F.; Leyssen, P.; Costa, J.;
Vanderheydt for their excellent technical assistance in the Litaudon, M. J. Nat. Prod. 2014, 77, 1505−1512.
acquisition of the antiviral data. This work has benefited from (20) Olivon, F.; Allard, P.-M.; Koval, A.; Righi, D.; Genta-Jouve, G.;
an “Investissement d’Avenir” grant managed by Agence Neyts, J.; Apel, C.; Pannecouque, C.; Nothias, L.-F.; Cachet, X.;
Nationale de la Recherche (CEBA, ref ANR-10-LABX-25- Marcourt, L.; Roussi, F.; Katanaev, V. L.; Touboul, D.; Wolfender, J.-
L.; Litaudon, M. ACS Chem. Biol. 2017, 12, 2644−2651.
01). The tree and leaves icons in the graphical abstract were (21) Olivon, F.; Apel, C.; Retailleau, P.; Allard, P. M.; Wolfender, J.
created by Sayan Mukerjhee, Alice Noir, Iejank, Lance L.; Touboul, D.; Roussi, F.; Litaudon, M.; Desrat, S. Org. Chem. Front.
Hancock, and LSE Designs from the Noun Project.


2018, 5, 2171−2178.
(22) Vasas, A.; Hohmann, J. Chem. Rev. 2014, 114, 8579−8612.
REFERENCES (23) Wang, H.-B.; Wang, X.-Y.; Liu, L.-P.; Qin, G.-W.; Kang, T.-G.
(1) Lazear, H. M.; Stringer, E. M.; de Silva, A. M. JAMA 2016, 315, Chem. Rev. 2015, 115, 2975−3011.
1945−1946. (24) Nothias-Scaglia, L.-F.; Pannecouque, C.; Renucci, F.; Delang,
(2) Burt, F. J.; Rolph, M. S.; Rulli, N. E.; Mahalingam, S.; Heise, M. L.; Neyts, J.; Roussi, F.; Costa, J.; Leyssen, P.; Litaudon, M.; Paolini, J.
T. Lancet 2012, 379, 662−671. J. Nat. Prod. 2015, 78, 1277−1283.
(3) Staples, J. E.; Breiman, R. F.; Powers, A. M. Clin. Infect. Dis. (25) Esposito, M.; Nim, S.; Nothias, L.-F.; Gallard, J.-F.; Rawal, M.
2009, 49, 942−948. K.; Costa, J.; Roussi, F.; Prasad, R.; Di Pietro, A.; Paolini, J.; Litaudon,
(4) Cao-Lormeau, V.-M.; Blake, A.; Mons, S.; Lastère, S.; Roche, C.; M. J. Nat. Prod. 2017, 80, 479−487.
Vanhomwegen, J.; Dub, T.; Baudouin, L.; Teissier, A.; Larre, P.; Vial, (26) Lanzotti, V. Diterpenes for Therapeutic Use. In Natural
A.-L.; Decam, C.; Choumet, V.; Halstead, S. K.; Willison, H. J.; Products: Phytochemistry, Botany and Metabolism of Alkaloids, Phenolics
Musset, L.; Manuguerra, J.-C.; Despres, P.; Fournier, E.; Mallet, H.-P.; and Terpenes; Ramawat, K. G.; Mérillon, J.-M., Eds.; Springer Berlin
Musso, D.; Fontanet, A.; Neil, J.; Ghawché, F. Lancet 2016, 387, Heidelberg: Berlin, Heidelberg, 2013; pp 3173−3191.
1531−1539. (27) Mwine, T. J.; Van Damme, P. J. Med. Plants Res. 2011, 5, 652−
(5) Ventura, C. V.; Maia, M.; Bravo-Filho, V.; Góis, A. L.; Belfort, R., 662.
Jr. Lancet 2016, 387, 228. (28) Shi, Q.-W.; Su, X.-H.; Kiyota, H. Chem. Rev. 2008, 108, 4295−
(6) Mlakar, J.; Korva, M.; Tul, N.; Popović, M.; Poljšak-Prijatelj, M.; 4327.
Mraz, J.; Kolenc, M.; Resman Rus, K.; Vesnaver Vipotnik, T.; Fabjan (29) Xu, J.-B.; Yue, J.-M. Org. Chem. Front. 2014, 1, 1225−1252.
Vodušek, V.; Vizjak, A.; Pižem, J.; Petrovec, M.; Avšič Ž upanc, T. N. (30) Allard, P.-M.; Martin, M.-T.; Tran Huu Dau, M.-E.; Leyssen,
Engl. J. Med. 2016, 374, 951−958. P.; Guéritte, F.; Litaudon, M. Org. Lett. 2012, 14, 342−345.
(7) Malet, H.; Massé, N.; Selisko, B.; Romette, J.-L.; Alvarez, K.; (31) Bourjot, M.; Leyssen, P.; Neyts, J.; Dumontet, V.; Litaudon, M.
Guillemot, J. C.; Tolou, H.; Yap, T. L.; Vasudevan, S. G.; Lescar, J.; Molecules 2014, 19, 3617−3627.
Canard, B. Antiviral Res. 2008, 80, 23−35. (32) Liao, S.-G.; Chen, H.-D.; Yue, J.-M. Chem. Rev. 2009, 109,
(8) Bollati, M.; Alvarez, K.; Assenberg, R.; Baronti, C.; Canard, B.; 1092−1140.
Cook, S.; Coutard, B.; Decroly, E.; de Lamballerie, X.; Gould, E. A.; (33) Li, L.-Z.; Song, S.-J.; Gao, P.-Y.; Li, F.-F.; Wang, L.-H.; Liu, Q.-
Grard, G.; Grimes, J. M.; Hilgenfeld, R.; Jansson, A. M.; Malet, H.; B.; Huang, X.-X.; Li, D.-Q.; Sun, Y. RSC Adv. 2015, 5, 4143−4152.
Mancini, E. J.; Mastrangelo, E.; Mattevi, A.; Milani, M.; Moureau, G.; (34) Li, S.-F.; Di, Y.-T.; Li, S.-L.; Zhang, Y.; Yang, F.-M.; Sun, Q.-Y.;
Neyts, J.; Owens, R. J.; Ren, J.; Selisko, B.; Speroni, S.; Steuber, H.; Simo, J. M.; He, H.-P.; Hao, X.-J. J. Nat. Prod. 2011, 74, 464−469.
Stuart, D. I.; Unge, T.; Bolognesi, M. Antiviral Res. 2010, 87, 125− (35) Lin, B.-D.; Han, M.-L.; Ji, Y.-C.; Chen, H.-D.; Yang, S.-P.;
148. Zhang, S.; Geng, M.-Y.; Yue, J.-M. J. Nat. Prod. 2010, 73, 1301−1305.

J DOI: 10.1021/acs.jnatprod.8b00800
J. Nat. Prod. XXXX, XXX, XXX−XXX
Journal of Natural Products Article

(36) Weidong, H.; Miroslav, C.; Giovanni, A.; Luc Van, P.; Josee, E. Millam, J. M.; Klene, M.; Adamo, C.; Cammi, R.; Ochterski, J. W.;
L.; Norbert De, K. Mini-Rev. Med. Chem. 2002, 2, 185−200. Martin, R. L.; Morokuma, K.; Farkas, O.; Foresman, J. B.; Fox, D. J.
(37) Ovenden, S. P. B.; Yew, A. L. S.; Glover, R. P.; Ng, S.; Rossant, Gaussian09, Revision D.01; Gaussian Inc.: Wallingford, CT, 2016.
C. J.; Regalado, J. C.; Soejarto, D. D.; Buss, A. D.; Butler, M. S. (48) Potisopon, S.; Ferron, F.; Fattorini, V.; Selisko, B.; Canard, B.
Tetrahedron Lett. 2001, 42, 7695−7697. Antiviral Res. 2017, 140, 25−36.
(38) Seephonkai, P.; Pyne, S. G.; Willis, A. C.; Lie, W. J. Nat. Prod.
2013, 76, 1358−1364.
(39) Li, S.-F.; He, H.-P.; Hao, X.-J. Nat. Prod. Res. 2015, 29, 1845−
1849.
(40) Tritsch, D.; Zinglé, C.; Rohmer, M.; Grosdemange-Billiard, C.
Bioorg. Chem. 2015, 59, 140−144.
(41) Olivon, F.; Roussi, F.; Litaudon, M.; Touboul, D. Anal. Bioanal.
Chem. 2017, 409, 5767−5778.
(42) Olivon, F.; Grelier, G.; Roussi, F.; Litaudon, M.; Touboul, D.
Anal. Chem. 2017, 89, 7836−7840.
(43) Chambers, M. C.; Maclean, B.; Burke, R.; Amodei, D.;
Ruderman, D. L.; Neumann, S.; Gatto, L.; Fischer, B.; Pratt, B.;
Egertson, J.; Hoff, K.; Kessner, D.; Tasman, N.; Shulman, N.; Frewen,
B.; Baker, T. A.; Brusniak, M.-Y.; Paulse, C.; Creasy, D.; Flashner, L.;
Kani, K.; Moulding, C.; Seymour, S. L.; Nuwaysir, L. M.; Lefebvre, B.;
Kuhlmann, F.; Roark, J.; Rainer, P.; Detlev, S.; Hemenway, T.;
Huhmer, A.; Langridge, J.; Connolly, B.; Chadick, T.; Holly, K.;
Eckels, J.; Deutsch, E. W.; Moritz, R. L.; Katz, J. E.; Agus, D. B.;
MacCoss, M.; Tabb, D. L.; Mallick, P. Nat. Biotechnol. 2012, 30, 918−
920.
(44) Pluskal, T.; Castillo, S.; Villar-Briones, A.; Orešič, M. BMC
Bioinf. 2010, 11, 395.
(45) Myers, O. D.; Sumner, S. J.; Li, S.; Barnes, S.; Du, X. Anal.
Chem. 2017, 89, 8696−8703.
(46) Wang, M.; Carver, J. J.; Phelan, V. V.; Sanchez, L. M.; Garg, N.;
Peng, Y.; Nguyen, D. D.; Watrous, J.; Kapono, C. A.; Luzzatto-Knaan,
T.; Porto, C.; Bouslimani, A.; Melnik, A. V.; Meehan, M. J.; Liu, W.-
T.; Crusemann, M.; Boudreau, P. D.; Esquenazi, E.; Sandoval-
Calderon, M.; Kersten, R. D.; Pace, L. A.; Quinn, R. A.; Duncan, K.
R.; Hsu, C.-C.; Floros, D. J.; Gavilan, R. G.; Kleigrewe, K.; Northen,
T.; Dutton, R. J.; Parrot, D.; Carlson, E. E.; Aigle, B.; Michelsen, C. F.;
Jelsbak, L.; Sohlenkamp, C.; Pevzner, P.; Edlund, A.; McLean, J.; Piel,
J.; Murphy, B. T.; Gerwick, L.; Liaw, C.-C.; Yang, Y.-L.; Humpf, H.-
U.; Maansson, M.; Keyzers, R. A.; Sims, A. C.; Johnson, A. R.;
Sidebottom, A. M.; Sedio, B. E.; Klitgaard, A.; Larson, C. B.; Boya P,
C. A.; Torres-Mendoza, D.; Gonzalez, D. J.; Silva, D. B.; Marques, L.
M.; Demarque, D. P.; Pociute, E.; O’Neill, E. C.; Briand, E.; Helfrich,
E. J. N.; Granatosky, E. A.; Glukhov, E.; Ryffel, F.; Houson, H.;
Mohimani, H.; Kharbush, J. J.; Zeng, Y.; Vorholt, J. A.; Kurita, K. L.;
Charusanti, P.; McPhail, K. L.; Nielsen, K. F.; Vuong, L.; Elfeki, M.;
Traxler, M. F.; Engene, N.; Koyama, N.; Vining, O. B.; Baric, R.; Silva,
R. R.; Mascuch, S. J.; Tomasi, S.; Jenkins, S.; Macherla, V.; Hoffman,
T.; Agarwal, V.; Williams, P. G.; Dai, J.; Neupane, R.; Gurr, J.;
Rodriguez, A. M. C.; Lamsa, A.; Zhang, C.; Dorrestein, K.; Duggan, B.
M.; Almaliti, J.; Allard, P.-M.; Phapale, P.; Nothias, L.-F.; Alexandrov,
T.; Litaudon, M.; Wolfender, J.-L.; Kyle, J. E.; Metz, T. O.; Peryea, T.;
Nguyen, D.-T.; VanLeer, D.; Shinn, P.; Jadhav, A.; Muller, R.; Waters,
K. M.; Shi, W.; Liu, X.; Zhang, L.; Knight, R.; Jensen, P. R.; Palsson,
B. O.; Pogliano, K.; Linington, R. G.; Gutierrez, M.; Lopes, N. P.;
Gerwick, W. H.; Moore, B. S.; Dorrestein, P. C.; Bandeira, N. Nat.
Biotechnol. 2016, 34, 828−837.
(47) Frisch, M. J.; Trucks, G. W.; Schlegel, H. B.; Scuseria, G. E.;
Robb, M. A.; Cheeseman, J. R.; Scalmani, G.; Barone, V.; Petersson,
G. A.; Nakatsuji, H.; Li, X.; Caricato, M.; Marenich, A. V.; Bloino, J.;
Janesko, B. G.; Gomperts, R.; Mennucci, B.; Hratchian, H. P.; Ortiz, J.
V.; Izmaylov, A. F.; Sonnenberg, J. L.; Williams-Young, D.; Ding, F.;
Lipparini, F.; Egidi, F.; Goings, J.; Peng, B.; Petrone, A.; Henderson,
T.; Ranasinghe, D.; Zakrzewski, V. G.; Gao, J.; Rega, N.; Zheng, G.;
Liang, W.; Hada, M.; Ehara, M.; Toyota, K.; Fukuda, R.; Hasegawa, J.;
Ishida, M.; Nakajima, T.; Honda, Y.; Kitao, O.; Nakai, H.; Vreven, T.;
Throssell, K.; Montgomery, J. A., Jr.; Peralta, J. E.; Ogliaro, F.;
Bearpark, M. J.; Heyd, J. J.; Brothers, E. N.; Kudin, K. N.; Staroverov,
V. N.; Keith, T. A.; Kobayashi, R.; Normand, J.; Raghavachari, K.;
Rendell, A. P.; Burant, J. C.; Iyengar, S. S.; Tomasi, J.; Cossi, M.;

K DOI: 10.1021/acs.jnatprod.8b00800
J. Nat. Prod. XXXX, XXX, XXX−XXX

You might also like