You are on page 1of 30

Article

Selective control of parasitic nematodes


using bioactivated nematicides
­­­
https://doi.org/10.1038/s41586-023-06105-5 Andrew R. Burns1,2 ✉, Rachel J. Baker3, Megan Kitner4, Jessica Knox1,2, Brittany Cooke1,2,
Jonathan R. Volpatti2,5, Aditya S. Vaidya6,7, Emily Puumala2, Bruna M. Palmeira8,
Received: 18 May 2021
Elizabeth M. Redman8, Jamie Snider1, Sagar Marwah9,10, Sai W. Chung9,10,
Accepted: 20 April 2023 Margaret H. MacDonald11, Jens Tiefenbach1, Chun Hu1,2, Qi Xiao12, Constance A. M. Finney12,
Henry M. Krause1,2, Sonya A. MacParland9,10, Igor Stagljar1,2,13,14, John S. Gilleard8,
Published online: 24 May 2023
Leah E. Cowen2, Susan L. F. Meyer11, Sean R. Cutler6,7, James J. Dowling2,5, Mark Lautens3,
Check for updates Inga Zasada4 & Peter J. Roy1,2,15 ✉

Parasitic nematodes are a major threat to global food security, particularly as the
world amasses 10 billion people amid limited arable land1–4. Most traditional
nematicides have been banned owing to poor nematode selectivity, leaving farmers
with inadequate means of pest control4–12. Here we use the model nematode
Caenorhabditis elegans to identify a family of selective imidazothiazole nematicides,
called selectivins, that undergo cytochrome-p450-mediated bioactivation in
nematodes. At low parts-per-million concentrations, selectivins perform comparably
well with commercial nematicides to control root infection by Meloidogyne incognita,
a highly destructive plant-parasitic nematode. Tests against numerous phylogenetically
diverse non-target systems demonstrate that selectivins are more nematode-selective
than most marketed nematicides. Selectivins are first-in-class bioactivated nematode
controls that provide efficacy and nematode selectivity.

Global food demand will be increasingly difficult to meet as the human tioxazafen and cyclobutrifluram. Unfortunately, iprodione has already
population approaches 10 billion people1–4. There is a scarcity of arable been banned in Europe owing to its carcinogenic potential and risk
land for agricultural expansion, and land-conversion efforts are con- to aquatic life10. Meanwhile, the market release of a tioxazafen-based
strained by social and ecological factors. Maximizing production from seed treatment has been postponed owing to reports of skin irritation
currently cultivated land will be crucial to ensure global food security1–4. in individuals handling this nematicide. There is a pressing need for
Farmers rely on agrochemicals to maximize yields by controlling new nematicides with improved selectivity.
crop pathogens4,5. Plant-parasitic nematodes (PPNs) are especially
destructive pathogens that cause more than US $100 billion in crop
losses every year6,7. Synthetic nematicides have played an essential Selective imidazothiazole nematicides
part in PPN control for decades; however, concerns over environmental From a library of uncharacterized compounds that disrupt the growth
toxicity and human safety have justifiably prompted bans on the most of the free-living nematode C. elegans14, we identified three new
commonly used nematicides8,9. The lack of available agents to control imidazothiazole-containing molecules that selectively kill nematodes
PPNs is stark. Of the 20 key nematicides used in the twentieth century, (Fig. 1a). At low micromolar concentrations, these selective imidazothia-
only 4 are currently approved for use in the European Union and only 3 zole nematicides, which we call selectivin-A, selectivin-B and selectivin-C,
are used in the USA without restriction9–11. Although warranted, these killed all four of the free-living nematode species that we can easily cul-
withdrawals leave farmers with limited options and no control meas- ture in the laboratory. Moreover, these selectivins were able to kill eggs of
ures for several PPNs8,12. a nematode parasite of cattle and the infective juveniles of at least one out
Despite the need for more selective PPN control measures, only of three distinct species of root-knot nematode that infect crops (Fig. 1b).
seven non-fumigant synthetic nematicides have been developed These eight nematode species span five genera and two evolutionary
in the past 25 years9,13. These next-generation nematicides are flu- clades, which suggests that diverse nematode species, including para-
azaindolizine, fluensulfone, fluopyram, iprodione, spirotetramat, sites, are susceptible to the selectivins. By contrast, these compounds

1
The Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada. 2Department of Molecular Genetics, University of Toronto, Toronto, Ontario,
Canada. 3Davenport Research Laboratories, Department of Chemistry, University of Toronto, Toronto, Ontario, Canada. 4USDA-ARS Horticultural Crops Research Laboratory, Corvallis, OR, USA.
5
Division of Neurology and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada. 6Institute for Integrative Genome Biology, University of California,
Riverside, Riverside, CA, USA. 7Department of Botany and Plant Sciences, University of California, Riverside, Riverside, CA, USA. 8Department of Comparative Biology and Experimental
Medicine, Host–Parasite Interactions Program, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada. 9Ajmera Transplant Centre, Toronto General Research Institute,
University Health Network, Toronto, Ontario, Canada. 10Department of Laboratory Medicine and Pathobiology and Immunology, University of Toronto, Toronto, Ontario, Canada. 11USDA-ARS
Mycology and Nematology Genetic Diversity and Biology Laboratory, Beltsville Agricultural Research Center, Beltsville, MD, USA. 12Department of Biological Sciences, Host Parasite Interactions
Program, Faculty of Science, University of Calgary, Calgary, Alberta, Canada. 13Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada. 14Mediterranean Institute for Life
Sciences, Split, Croatia. 15Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada. ✉e-mail: andy.burns@utoronto.ca; peter.roy@utoronto.ca

102 | Nature | Vol 618 | 1 June 2023


a b Free-living nematodes Parasitic nematodes

R2
Ce Cb Pp KR Co Mi Mc Mh
Sel-A 0 1
Selectivin R1
N
R3 Sel-B Relative
core structure N S Sel-C viability

0
1.6
3.1
6.3
12.5
25
50
100
0
1.6
3.1
6.3
12.5
25
50
100
0
1.6
3.1
6.3
12.5
25
50
100
0
1.6
3.1
6.3
12.5
25
50
100
0
1.6
3.1
6.3
12.5
25
50
100
0
5
15
45
0
5
15
45
0
5
15
45
N
Sel-A Cl
Concentration (μM)
N S
c
Fungi Fish Human cells Insect
N
Sel-B F Sc Ca Dr HEK HepaRG Dm (ad) Dm (lv)
N S
Sel-A 0 1
N Sel-B Relative
Sel-C Br Sel-C viability, growth
N S
or motility

12.5

12.5

12.5

12.5

12.5
0
1.6
3.1
6.3

25
50
100
0
1.6
3.1
6.3

25
50
100
0
1.6
3.1
6.3

25
50
100
0
1.6
3.1
6.3

25
50
100
0
1.6
3.1
6.3

25
50
100
0
5
15
45
0
5
15
45
Concentration (μM)

Fig. 1 | The selectivins. a, The selectivin core scaffold and structures of (Dm (lv)). Viability was assessed for D. rerio and D. melanogaster larvae. Cell
selectivin-A (Sel-A), selectivin-B (Sel-B) and selectivin-C (Sel-C). b, Selectivin growth was assessed for S. cerevisiae, C. albicans, HEK293 cells and HepaRG cells.
dose–response for the following eight nematode species: C. elegans (clade V) Motility was assessed for D. melanogaster adults. The colour-coded scale denotes
(Ce); Caenorhabditis briggsae (clade V) (Cb); Pristionchus pacificus (clade V) viability, growth or motility relative to untreated control, as appropriate for
(Pp); Rhabditophanes spp. KR3021 (clade IV) (KR); Cooperia oncophora the system tested. In b,c, dose–response data are an average across n biological
(clade V) (Co); M. incognita (clade IV) (Mi); Meloidogyne chitwoodi (clade IV) replicates (BRs) or technical replicates (TRs), with the following n values for each
(Mc); and Meloidogyne hapla (clade IV) (Mh). The colour-coded scale denotes system: Ce (n = 10 BRs for sel-A, 9 BRs for sel-B, 13 BRs for sel-C), Cb (n = 3 BRs),
viability relative to untreated control. c, Selectivin dose–response for the Pp (n = 3 BRs), KR (n = 3 BRs), Co (n = 3 BRs), Mi (n = 6 BRs), Mc (n = 6 BRs), Mh
following six phylogenetically distinct non-nematode model systems: Sc, (n = 6 BRs), Sc (n = 3 BRs), Ca (n = 3 TRs), Dr (n = 3 BRs), HEK (n = 3 TRs), HepaRG
S. cerevisiae (Sc); Candida albicans (Ca); D. rerio (Dr); HEK293 cells (HEK); (n = 3 TRs), Dm (n = 3 BRs).
HepaRG cells; D. melanogaster adults (Dm (ad)); and D. melanogaster larvae

exhibited little-to-no activity against fungi, insects, fish or human cells CYP enzymes by compromising EMB-8 activity during larval develop-
at concentrations that readily killed nematodes (Fig. 1c). Furthermore, ment, bypassing its essential role during embryogenesis27. EMB-8 is
mice given a daily oral dose of 50 mg kg–1 selectivin-A for 5 days showed the C. elegans P450 oxidoreductase (POR) enzyme that is a necessary
no obvious pathologies relative to untreated mice. To our knowledge, redox partner of all microsomal CYP enzymes27,28. Consistent with the
broad-spectrum nematicidal activity has not been previously described notion that selectivins are bioactivated nematicides, emb-8 knockdown
for any compound containing the 6-phenylimidazo[2,1-b]thiazole scaf- rendered C. elegans resistant to selectivin-induced lethality (Fig. 2a). By
fold of selectivins. Thus, the selectivins represent a new structural class contrast, the compound wact-11, which does not require bioactivation
for the development of selective and effective PPN control. for activity14, killed worms in an EMB-8-independent manner.
To identify selectivin metabolites, we incubated both young
adult and first larval stage (L1) worms in the presence or absence of
Selectivins have a new mode of action 100 µM selectivin-A for 6 h and then processed whole worm lysates
As a first step towards understanding the mode of action of selectivins, by reversed-phase high-performance liquid chromatography (HPLC)
we tested selectivin-A and selectivin-C against eight mutant strains coupled with a diode-array absorbance detector (HPLC-DAD) (Fig. 2b).
of C. elegans that are each specifically resistant to one of the major We identified five distinct peaks in the selectivin-A-treated lysates
anthelmintic or nematicide classes14–22 (Extended Data Table 1). None that were absent from the untreated controls. The peak that eluted
of the mutants were resistant to selectivins, which suggests that selec- at 7 min had the same retention time and absorbance spectrum as
tivins have a distinct mode of action compared with the commercial the selectivin-A standard that was directly injected onto the column.
agents examined. This peak is probably unmodified selectivin-A (parent molecule) that
To further characterize the mode of action of the selectivins, we persists in worm tissue following incubation. The remaining four peaks
performed a forward genetic screen of C. elegans mutants (generated eluted earlier in the HPLC run relative to selectivin-A, which suggested
through random mutagenesis) that resist selectivin-induced lethality. that they are relatively less lipophilic. These peaks were absent from
This approach has previously produced large numbers of mutants the selectivin-A standard, and they were not found in the lysates of
that specifically resist many of the major classes of anthelmintics and dead worms incubated in selectivin-A. This result supports the idea
nematicides that are commercially used14,15,17,19,20,22,23. Despite screening that they are bona fide selectivin-A metabolites and not accumulated
more than 10 million mutagenized genomes, selectivin-resistant worms contaminants or spontaneous oxidation products (Fig. 2b,c). We named
could not be generated (Extended Data Table 2). This result reinforces these presumptive selectivin-A metabolites M1, M2, M3 and M4.
the conclusion that selectivins kill nematodes using a mechanism that Consistent with our hypothesis that selectivin-A is bioactivated, the
is distinct from traditional nematode control agents. abundance of all four metabolites in worm tissue was substantially
reduced following emb-8 knockdown, whereas the abundance of the
unmodified selectivin-A parent increased by almost twofold (Fig. 2b,d).
Selectivins are bioactivated nematicides Unmodified selectivin-A is unlikely to be the active agent in vivo because
Previous work using C. elegans has shown that metabolites of com- worms in which emb-8 was knocked down survived treatment with
pounds that are lethal to nematodes often accumulate in worm tissue24. 100 µM selectivin-A even though the internal concentration was nearly
It is therefore possible that a nematicide could be transformed from an twice as high as that found in wild-type worms (Fig. 2a,d). Thus, selec-
innocuous parent molecule into a metabolic product that is lethal to tivins are bioactivated pro-nematicides.
worms. Microsomal cytochrome P450 (CYP) enzymes are commonly
involved in the oxidative biotransformation of inert pro-drugs into
bioactive metabolites25. The C. elegans genome encodes 76 microso- Bioactivation to a toxic electrophile
mal CYP enzymes26, many of which are known to metabolize drugs27. To identify the toxic selectivin metabolite (or metabolites), we
To test whether selectivins are bioactivated by C. elegans, we capital- collected HPLC fractions containing the four selectivin-A metabo-
ized on previous work that achieved broad disruption of microsomal lites and analysed their structures by electrospray ionization mass

Nature | Vol 618 | 1 June 2023 | 103


Article
a Wild-type emb-8 KD b mAU per mg dry weight
Sel-A
Sel-B 0 50 100 150 200
Sel-C 340
Sel-A 320
Wact-11

standard
300

Sel-A
0
1.6
3.1
6.3
12.5
25
50
100

0
1.6
3.1
6.3
12.5
25
50
100
280
Concentration (μM)
260
0 1
Relative viability
M4 340
M1 M2 M3 Sel-A
320
c P = 0.0019

+ sel-A
WT L1
300
20,000 Live worms 280
Dead worms
10,000 260

3,000 340

Wavelength (nm)
Sel-A
AUC

320

P = 0.0015

P = 0.0004

P = 0.0265
P = 0.0027

Dead WT L1
2,000

+ sel-A
300
1,000 280
260
0
Sel-A M1 M2 M3 M4
Analyte M4 340
M1 M2 M3 Sel-A 320

WT adults
+ sel-A
300
d 2,000 WT 280
emb-8 KD
P = 0.0415 260
1,500
P = 3.543 × 10–6
10–5
10–5

P = 2.539 × 10–5

340
emb-8 KD adults
AUC

1,000 Sel-A
P = 2.146 ×
P = 2.545 ×

320
+ sel-A

300
500 280
260
0
Sel-A M1 M2 M3 M4
3
4
5

9
0
9
0
1
0
1
2
2.
2.
2.

4.
5.
6.
7.
7.
2.
2.
2.

Analyte
Retention time (min)

Fig. 2 | Selectivins are bioactivated nematicides. a, Selectivin and wact-11 is the sel-A standard injected directly onto the column and is not biomass-
dose–response for wild-type (WT) and emb-8 knockdown (KD) adult worms. normalized or background-corrected. Unmodified sel-A and metabolites M1–M4
WT dose–response data are an average across 7 BRs for sel-A and 4 BRs for the are indicated. c, HPLC-DAD quantification of unmodified sel-A and M1–M4 in the
other three compounds. emb-8 KD dose–response data are an average across 3 lysates of live and dead L1 worms (n = 3 BRs with 200,000 worms per replicate).
BRs. b, HPLC chromatograms of lysates from live and dead L1 worms and from d, HPLC-DAD quantification of sel-A and M1–M4 in the lysates of WT and emb-8
WT and emb-8 KD adults treated with 100 µM sel-A. The chromatograms shown KD adults (n = 3 BRs with 1,250 worms per replicate). In both c and d, the AUC is
are the product of dry weight normalization of raw chromatograms followed the area under the curve for the given analyte peak, calculated at 260 nm for
by subtraction of the absorbance intensity from paired untreated controls. M1–M3 and at 300 nm for M4. Data are presented as the mean ± s.e.m. For each
Absorbance intensity is in milliabsorbance units (mAU) per mg of dry weight, analyte, P values were obtained from unpaired one-tailed Student’s t-tests
and the absorbance wavelength (y axis) is in nanometres. The top chromatogram comparing the means of live and dead worms (c) or WT and emb-8 KD worms (d).

spectrometry (ESI-MS) followed by MS/MS fragmentation. Based on our and Extended Data Fig. 2). The 1H-13C heteronuclear single quantum
analysis of the M4 fraction, we speculated that M4 is a disulfide of two coherence spectrum indicated a carbon (C13) at 64.1 ppm containing
4-(4-chlorophenyl)-1H-imidazole-2-thiol monomers (Extended Data two diastereotopic protons at 3.82 and 4.67 ppm, and a carbon (C14) at
Fig. 1a,b, Extended Data Table 3 and Supplementary Table 1). Alkylating 60.9 ppm containing a proton at 6.00 ppm. From the 1H-1H correlation
lysis resulted in the presumptive alkylated monomer, but not the spectroscopy spectrum, a correlation between the protons on C13 and
disulfide, which indicated that the monomer forms in vivo, whereas C14 was observed, which suggested that these carbons are directly con-
the disulfide forms after lysis (Extended Data Fig. 1c–e and Supplemen- nected. Connectivity of the glutathione molecule through sulfur to C14
tary Table 1). Dose–response analysis with the imidazole-thiol mono- was confirmed by a heteronuclear multiple bond correlation between the
mer, which is commercially available, revealed that it was unable to kill proton on C14 and C17 and vice versa. Therefore, we identified metabolite
worms up to a concentration of 100 µM, despite readily accumulating M2 as a glutathione conjugate of an electrophilic sulfoxide metabolite of
in worm tissue (Extended Data Fig. 1f–h). Therefore, the imidazole-thiol selectivin-A. As is typical for reactive electrophiles, it was not possible
is not responsible for the lethality of selectivins. to obtain a pure isolate of the unconjugated metabolite. We therefore
Abundant masses in the M1, M2 and M3 fractions, and respective inferred its structure from the glutathione conjugate, which is a com-
MS/MS fragmentations, were consistent with γ-glutamylcysteine, glu- mon practice in drug bioactivation studies29,30. We speculated that M1
tathione and cysteine conjugates of an electrophilic sulfoxide metabo- and M3 are γ-glutamylcysteine and cysteine conjugates of the same
lite of selectivin-A, respectively (Fig. 3a–c, Extended Data Table 3 and sulfoxide metabolite, respectively. Alkylating lysis did not lower the
Supplementary Table 1). NMR characterization of a HPLC-purified abundance of the sulfoxide conjugates in worm lysates, which indicated
M2 fraction was consistent with the glutathione sulfur reacting at the that they are produced in vivo (Extended Data Fig. 1c,e). Thus, our data
electrophilic β-carbon of the proposed sulfoxide metabolite (Fig. 3d suggest that worms have a canonical detoxification pathway in which

104 | Nature | Vol 618 | 1 June 2023


a c
M1 Control 501.07 MS/MS 148.04 M3 Control 372.02 MS/MS
H NH2
HOOC N HOOC NH2
Relative abundance

Relative abundance
501.07 148.04 COOH 372.02 148.04 224.99
$
1.0 $ $ 1.0
$ $ $
O
277.09 224.99 S γ-Glu-Cys 224.99 S
Cys
N N
224.99 Cl Cl
N S 374.02
!"# !"# N S
!"#
0.5 !"#
503.06 !"# 277.09 !"#
0.5
O 501.07 O 148.04
Metabolite M1 Metabolite M3
[M1+H]+ = 501.07 [M3+H]+ = 372.02
372.02
!0 ! ! !0 ! !

0!

&5#
&5#

!0

!0

!0

z
0

5
0

5
0

44%

&0!
&0!
/z!

0!

(&m/
%37

%37
%37

%37
$1%4

$1&9

24

'&29

34
50

50
50

50
14

19

24

29

34

39

'%

(%
!

!
d O NH2
b M2 Control 558.09 MS/MS 205.06 H
O H NH2 HO C33 C19 N C22 C25 OH
558.09 205.06 N C34 N C18 C21 C24 C26
Relative abundance

HOOC N COOH
$
1.0 $ $ H
H
O
224.99 S GSH O C17 O O
334.11 S
N
Cl
!"#
0.5 560.09
!"# !"# 224.99 N S 129.7 127.5 115.8
334.11 C1 C6 60.9 COSY
O 558.09 C11 C14
Metabolite M2 132.8
N
[M2+H]+ = 558.09 Cl C2 C5 C7150.0 C13 HMBC
134.5 C9 64.1
!0 ! ! C3 C4 N
153.2
S
129.7 127.5

! /z
%7

'2
%7

'2
!! 0

#!0

!0

!0

!0

!0
##55

#&56
##55

#&56
%20

%25

&3! 0

&3#5

'!!40

'#45

#!50

##m
O
f

(relative to untreated) (%)


– NACET
+ NACET
e Sulfoxide
Electrophilic
Glutathione γ-Glu-Cys Cys 150
carbon
Sel-A CYP metabolite

Viability
M2 S M1 S M3 S
100
enzymes GST PCS GGT N
N N N N
Cl Cl Cl Cl Cl
N S N N S N S
50
CYP- S N S
35C1 O GSH O O O 0
Sel-A Sel-B Sel-C
Treatment
g h 1 j 1

Relative viability

Relative growth
WT cyp-35c1Δ EV CYP-35C1
50 l
Sel-A Sel-A
Sel-B Sel-B No selectivin
CYP-35C1
Moving animals (%)

40 Sel-C Sel-C bioactivation

0
25
50

25
50
100

100
12.5

12.5
0

0
25
50

25
50
6.3

6.3
100

100
12.5

12.5

0 0 EV
30
Concentration (μM) Concentration (μM)
20 i k
WT Selectivin
EV P = 1.492 × 10–6
10 Z-score > 3 P = 0.0033 Selectivin sulfoxide
cyp-35c1Δ + CYP-35C1 CYP-35C1
0
0 2,000 4,000 6,000 8,000 0 20,000 40,000 60,000
13A01
13A02
13A03
13A04
13A05
13A06
13A07
13A11
13B01
13B02
14A01
14A04
14A05
23A01
25A01
25A02
25A03
25A04
25A05
25A06
29A02
31A02
32A01
33B01

33E01
33E02
33E03
34A01
34A02
34A03
34A04
34A05
34A06
34A07
34A08
34A09
34A10
35A02
35A03
35A04
35A05
35B01
35B03

36A01
37A01
37B01
43A01
33C01
33C02
33C03
33C04
33C06
33C07
33C08
33C09
33C11
33D01
33D03

35C01
35D01

Individual C. elegans CYP RNAi KDs AUC (M1–M3) AUC (M1–M3)

Fig. 3 | Selectivins are bioactivated to a toxic electrophile. a–c, MS data identifies CYP-35C1. Z-scores were calculated using mean and s.d. of 50 mock
for metabolite M1 (a) M2 (b) and M3 (b) HPLC fractions and untreated control experiments. Raw data are provided in Supplementary Table 2. h, Selectivin
fractions, together with MS/MS fragmentation data for selected masses. dose–response for C. elegans WT and cyp-35c1 deletion (cyp-35c1Δ) mutants.
Structures are shown for γ-glutamylcysteine (γ-Glu-Cys), glutathione (GSH) WT dose–response data are an average across 7 BRs for sel-A and 4 BRs for the
and cysteine conjugates of a sel-A sulfoxide metabolite, along with fragment other two compounds. cyp-35c1Δ mutant dose–response data are an average
structures. d, 1H-NMR and 13C-NMR characterization of HPLC-purified M2. 1H-1H across 3 BRs. i, HPLC-DAD quantification of total M1–M3 in the lysates of
correlation spectroscopy (COSY) and 1H-13C heteronuclear multiple bond C. elegans WT and cyp-35C1Δ mutants (n = 3 BRs). j, Selectivin dose–response
correlation (HMBC) values are indicated. Carbon chemical shifts are in green. for CYP-35C1-expressing yeast and empty vector (EV) control. Dose–response
NMR spectra are provided in Extended Data Fig. 2. e, A proposed pathway for data are an average across 3 BRs. k, HPLC-DAD quantification of total M1–M3 in
sel-A sulfoxidation and low-molecular-weight thiol conjugation and CYP the lysates of CYP-35C1-expressing yeast and EV control (n = 4 BRs). l, Selectivin
enzymes that probably mediate S-oxidation. GSH reacts with the electrophilic bioactivation occurs in CYP-35C1-expressing yeast not in EV control. In i and k,
sulfoxide metabolite spontaneously or through glutathione-S-transferase AUC is area under the curve at 260 nm for M1–M3 combined. Data are presented
(GST), and phytochelatin synthase (PCS) and γ-glutamyl transferase (GGT) as the mean ± s.e.m. P values are from unpaired one-tailed Student’s t-tests
convert it to γ-Glu-Cys and cysteine. f, Viability of selectivin-treated L1 worms comparing the means of WT and cyp-35C1Δ worms (i) or EV control and CYP-35C1-
with or without 2.5 mM NACET (n = 5 BRs). Data are presented as the mean ± s.d. expressing yeast (k).
g, A C. elegans CYP RNAi screen for suppression of sel-A-induced lethality

glutathione is enzymatically conjugated to the potentially toxic electro- multiple oxidation schemes (Methods). Instead, we tested whether
philic sulfoxide metabolite, thereby sequestering it from cellular nucleo- the exogenous addition of N-acetylcysteine ethyl ester (NACET),
philes. The conjugated glutathione is then enzymatically converted which is enzymatically converted into cysteine for use in the synthe-
to γ-glutamylcysteine and cysteine, which results in metabolites that sis of new glutathione35, could suppress selectivin-induced lethality.
are readily excretable31–34 (Fig. 3e). Alternatively, but not in a mutually NACET has been used to block the damage caused by acetaminophen
exclusive manner, the sulfoxide metabolite could spontaneously react overdose, which results from the build-up of a reactive electrophilic
with glutathione and the other low-molecular-weight thiols without the metabolite that depletes glutathione and reacts with proteins to cause
aid of enzyme catalysts. Regardless of the mechanism, the accumula- hepatotoxicity35. We reasoned that the sulfoxide metabolite may kill
tion of the sulfoxide conjugates in worm tissue suggests that sulfoxide nematodes through a similar mechanism. Administration of 2.5 mM
metabolite production outpaces the capacity of worms to detoxify them. NACET suppressed selectivin-induced lethality, which indicated
We could not directly test whether the sulfoxide metabolite that the sulfoxide metabolite is the active nematicidal agent in vivo
induces lethality because it could not be synthesized despite trying (Fig. 3f).

Nature | Vol 618 | 1 June 2023 | 105


Article
To identify the CYP enzyme (or enzymes) responsible for selectivin levels is phylogenetically restricted to nematodes. Future work will be
bioactivation, we individually knocked down 60 out of 76 C. elegans required to determine the long-term effects of the relatively low levels
microsomal CYP enzymes by RNA interference (RNAi) and assayed for of metabolite production observed in some non-nematode species.
selectivin-A resistance. cyp-35c1 knockdown conferred resistance to
selectivin-A, which suggested that it has a role in bioactivation (Fig. 3g
and Supplementary Table 2). We validated this result with a cyp-35c1 Lead nematicides for PPN control
deletion mutant, which demonstrated resistance to all three selectivin We were encouraged by the selectivity, broad-spectrum activity and new
analogues (Fig. 3h). Furthermore, abundance of the metabolites M1, M2 mechanism of action of the selectivins. Therefore we performed an ana-
and M3 were reduced by half in the tissues of cyp-35c1 deletion mutants logue series screen to identify lead nematicides capable of effectively con-
compared with wild-type worms (Fig. 3i), which provides support that trolling root infection by the root-knot nematode M. incognita, one of the
the reactive sulfoxide metabolite is produced by CYP-35C1. Metabolite most destructive crop pathogens in the world36. Eleven analogues were
production was not completely eliminated in cyp-35c1 deletion mutants, included in the series, along with all four of the marketed next-generation
which suggested that additional CYP enzymes act redundantly to pro- soil-applied synthetic nematicides for which powder is commercially
duce the sulfoxide metabolite. This redundancy may explain why we did available: fluensulfone, fluopyram, iprodione and tioxazafen. Selectivin
not identify cyp-35c1 in our genetic screen, which was carried out using L1 analogues were either purchased or synthesized (Methods). Tests were
worms that are twice as sensitive to selectivin-A compared with the young performed against M. incognita in its soil environment with a tomato host
adults used for the RNAi screen and follow-up dose–response analyses plant (Fig. 4a). Soil-based assays are vital for establishing the real-world
(Figs. 1b and 3h). To demonstrate that CYP-35C1 is able to produce the utility of candidate nematicides, as many compounds that are effective
reactive metabolite, we heterologously expressed it in budding yeast in vitro lose activity in the soil because of poor mobility and/or degrada-
and assayed for both the production of the reactive metabolite and tion37. We assayed compounds by adding 20 ml of 45 µM compound in
for selectivin-induced growth inhibition. Unlike wild-type yeast, yeast water and between 1,000 and 2,500 infective second-stage juvenile ( J2)
expressing nematode CYP-35C1 readily produced the sulfoxide conju- nematode larvae to 90 g of potted soil. The final soil concentration was
gates of selectivin-A and were sensitive to selectivin-induced cytotoxic- approximately 2.5 ppm or 4.5 kg per hectare (kg ha–1), which is compa-
ity (Fig. 3j–l, Extended Data Table 3 and Supplementary Table 1). These rable to the application rates of commercial nematicides37,38. After 24 h,
results demonstrate that CYP-35C1 is both necessary and sufficient for we planted 2–3-week-old tomato seedlings in the soil. Eight weeks later,
the production of the selectivin sulfoxide metabolite and further support the per cent effectiveness of the compounds at reducing the number of
the conclusion that selectivins are bioactivated to a toxic electrophile. eggs in the roots was calculated relative to untreated controls (Methods).
Of the 15 compounds tested, selectivin analogues A, C and E, and the
four commercial nematicides, had per cent effectiveness values that dif-
Bioactivation is specific to nematodes fered significantly from zero effect (one-sample t-test P < 0.01) (Fig. 4a).
Given their selective nematicidal activity, we proposed that the bio- Fluensulfone and fluopyram demonstrated nearly 100% effectiveness
activation of selectivins may be phylogenetically restricted to nema- and were the best-performing nematicides. Consistent with previous
todes. To test the phylogenetic specificity of selectivin bioactivation, reports, iprodione was the least effective commercial nematicide (38%
we analysed selectivin-A metabolites in both the lysates and incubation effective)13. Selectivin-A and selectivin-E were the most effective selec-
buffers of five distinct nematode species spanning four genera and two tivin analogues (43% and 56% effective, respectively), and their efficacies
evolutionary clades, and in three non-nematode species from diverse were not significantly different from that of tioxazafen (one-tailed t-test
phyla (Extended Data Fig. 3a,b). The non-nematode species tested were P > 0.05) (Fig. 4a). Of note, we challenged the plants with a nematode
the yeast Saccharomyces cerevisiae, the fly Drosophila melanogaster density that was at least twofold greater than what is considered high
and the fish Danio rerio. Low-molecular-weight thiol conjugates of the density in fields39,40, which may account for why some of the nematicides
sulfoxide metabolite were abundantly produced by all five of the nema- tested had incomplete efficacies in this assay. Treatment with the major-
tode species, including the PPN M. incognita (Extended Data Fig. 3a–c, ity of selectivins, including selectivin-A and selectivin-E, increased the
Extended Data Table 3 and Supplementary Table 1). By contrast, the three root weight relative to untreated control (Extended Data Fig. 4), which
non-nematode species produced significantly less of the conjugates is probably due to the reduced nematode burden.
relative to nematodes (Extended Data Fig. 3a–c). Notably, the conjugates To assess the selectivity of selectivin-A and selectivin-E for PPNs, we
were undetectable by HPLC-DAD and MS in yeast lysates and buffers, tested them against a panel of phylogenetically diverse and experi-
and they are undetectable by HPLC-DAD in the lysates and buffers from mentally tractable non-target systems, including rhizobacteria, fungi,
flies (Extended Data Fig. 3a–c and Extended Data Table 3). Only the plants, free-living nematodes (including the soil-beneficial nematode
cysteine conjugate was detectable by MS at very low levels in fly lysates Phasmarhabditis hermaphrodita), insects, fish and two different human
(Extended Data Table 3). Fish produced more than fourfold less of the cell types (Fig. 4b,c). One of the human cell types is HepaRG, which is a
conjugates than C. elegans, and more than twofold less than the other liver progenitor cell type that is a well-established model for assessing
nematode species tested (Extended Data Fig. 3a–c). Furthermore, the drug metabolism and bioactivation in human liver41. We also included
glutathione conjugate (M2) readily accumulated in the tissues of all five in our tests all of the newly developed synthetic nematicides that are
of the nematode species tested over the course of the incubation period, purchasable, the natural product nematicide abamectin, the organo-
but not in the tissues of the non-nematode species. This result suggests phosphate nematicide fenamiphos and the carbamate nematicide
that abundant sulfoxide metabolite production and oversaturation of oxamyl, which is still in use. Selectivin-E had the best selectivity profile
the detoxification pathway is common to nematodes but not to distinct compared with all of the nematicides tested. Selectivin-E was inactive
phyla (Extended Data Fig. 3a,d). Notably, tissue accumulation of the against all of the non-target systems up to 100 µM, which was the high-
selectivin-A parent compound in the non-nematode species was com- est concentration assayed. By contrast, every commercial nematicide
parable to, or greater than, accumulation in C. elegans tissue (Extended tested had unwanted toxicity in at least one of the non-target assays.
Data Fig. 3a, e). This result suggests that poor bioavailability is unlikely to Notably, selectivin-E was non-lethal to the three free-living nematodes
account for the lower levels of metabolite production by non-nematode included in our analyses, which suggested that it may be innocuous to
species and provides further evidence that the selectivins require non-target soil-beneficial nematodes. Notably, the free-living nema-
bioactivation to induce lethality. Taken together, these data indicate todes did not produce a reactive sulfoxide metabolite of selectivin-E.
that distinct nematode species, including PPNs, are capable of strongly Instead, it is likely that they detoxify selectivin-E through hydroxyla-
bioactivating the selectivins, and that bioactivation to acutely toxic tion and subsequent phosphoglucosidation (Extended Data Fig. 5).

106 | Nature | Vol 618 | 1 June 2023


a [Aqueous] [Soil] b
Sel-E AB FE FS FP
Treatment (μM) (ppm) kg ha–1 R1 R2 R3 n

Mock
Fluensulfone 38 2.5 4.5 – – – 6 7.76 × 10–15
Fluopyram 28 2.5 4.5 – – – 6 1.44 × 10–15

Tioxazafen 45 2.3 4.1 – – – 6 0.004

15 μM
Sel-E 45 2.5 4.5 Cl H Me 6 0.001

Sel-A 45 2.4 4.3 Cl H H 6 0.009

45 μM
Sel-C 45 2.8 5.0 Br H H 6 0.007
Iprodione 34 2.5 4.5 – – – 6 0.008

Sel-K 45 3.4 6.1 I H Me 6 0.025


Sel-G 45 2.3 4.1 F H Me 4 0.167 Sel-A IP OX SP TX
Sel-B 45 2.2 4.0 F H H 6 0.066

Mock
Sel-H 45 2.9 5.2 Br Me H 6 0.194
Sel-J 45 3.3 5.9 I H H 4 0.386
Sel-F 45 2.3 4.1 F Me H 6 0.411

15 μM
Sel-D 45 2.5 4.5 Cl Me H 4 0.274
Sel-I 45 2.9 5.2 Br H Me 6 0.796

45 μM
–50 0 50 100
Treatment effectiveness (%)

c Rhizobacteria Fungus Non-target nematodes Insect Fish Human cells

Nematicide P. simiae C. albicans C. elegans P. pacificus P. herm. D. melanogaster D. rerio HEK293 HepaRG

Sel-E
Sel-A
Abamectin
Fenamiphos
Fluensulfone
Fluopyram
Iprodione
Oxamyl
Spirotetramat
Tioxazafen
0
1.56
3.13
6.25
12.5
25
50
100
0
1.56
3.13
6.25
12.5
25
50
100
0
1.56
3.13
6.25
12.5
25
50
100
0
1.56
3.13
6.25
12.5
25
50
100
0
5
15
45

0
5
15
45
0
5
15
45

0
1.56
3.13
6.25
12.5
25
50
100
0
1.56
3.13
6.25
12.5
25
50
100
0
1.56
3.13
6.25
12.5
25
50
100
Concentration (μM)

0 1 0 1 0 1 0 1 0 1 0 1 0 1 0 1 0 1 0 1
Relative Relative Relative Relative Relative Relative Relative Relative Relative Relative
growth growth viability viability motility motility viability viability growth growth
(adult) (larval)

Fig. 4 | Selectivin-A and selectivin-E are lead nematicides for plant-parasitic (SP) and tioxazafen (TX). A representative image taken from one of three
nematode control. a, Per cent effectiveness of 11 selectivin analogues and 4 independent trials is shown. c, Dose–response of phylogenetically diverse
commercial nematicides at preventing tomato plant root infection by non-target systems treated with sel-E, sel-A and eight commercial nematicides.
M. incognita (Methods). For each analogue, aqueous molar concentration, parts The phenotypic outcome of each treatment, relative to untreated control is
per million soil concentration and kg ha–1 values are indicated. The R-groups for denoted by the colour-coded scales. The following systems were treated:
each analogue are also indicated (see Fig. 1a for the core structure and R-group P. simiae, C. albicans, C. elegans, P. pacificus, P. hermaphrodita (P. herm.),
positions of selectivins). P values at the far right of the graph were obtained D. melanogaster; D. rerio, human HEK293 cells and human HepaRG cells. Dose–
from two-tailed one-sample t-tests comparing the mean per cent effectiveness response data are an average across: P. simiae (n = 3 TRs), C. albicans (n = 3 TRs),
of each treatment with zero effect. Error bars are s.e.m. b, Arabidopsis thaliana C. elegans (n = 10 BRs for sel-A, 4 BRs for sel-E, 6 BRs for all other compounds),
seedling establishment and greening in response to sel-E, sel-A and the P. pacificus (n = 3 BRs for sel-A, 4 BRs for sel-E, 6 BRs for all other compounds),
following eight commercial nematicides: abamectin (AB), fenamiphos (FE), P. herm. (n = 3 BRs), D. melanogaster (n = 3 BRs), D. rerio (n = 3 BRs), HEK293 (n = 3
fluensulfone (FS), fluopyram (FP), iprodione (IP), oxamyl (OX), spirotetramat TRs for sel-A, 3 TRs for FE, 4 TRs for all other compounds), HepaRG (n = 3 TRs).

Selectivin-A was active against free-living nematodes, as were several enzyme (Fig. 3j–l). Using this system, we individually expressed 19 dis-
marketed next-generation nematicides, but it was more selective for tinct M. incognita CYP enzymes in separate yeast strains and measured
nematodes than the majority of the commercial nematicides tested their growth in the presence or absence of either 50 µM selectivin-A
(Fig. 4b,c). Selectivin-A and selectivin-E have lipophilic properties or selectivin-E. These 19 CYP enzymes are a representative subset of a
(logP value of around 4) and are only moderately water soluble. These larger set of 31 CYP enzymes identified through bioinformatics assays
results indicate that they will not readily leach into ground water and from the most recent iteration of the M. incognita genome (Methods
are unlikely to be taken up systemically by plants42, thereby limiting resi- and Supplementary Tables 3,4). Selectivin-A-treated yeast cells express-
dues in fruits and other plant products ingested by consumers. Taken ing M. incognita CYP4731A3 grew slower than untreated cells (Fig. 5a,b
together, these data suggest that our best-performing lead nematicides and Supplementary Table 5). By contrast, yeast carrying the empty
can effectively and selectively control nematode pests. Going forwards, vector were relatively insensitive to selectivin-A treatment (Fig 5a,b and
it will be important to test the selectivins against additional organisms Supplementary Table 5). Yeast expressing CYP4731A3 produced sulfox-
that affect soil health and plants of commercial interest. ide conjugates of selectivin-A, whereas the empty vector control strain
did not (Fig. 5c,d, Extended Data Table 3 and Supplementary Table 1).
These data demonstrate that CYP4731A3 can bioactivate selectivin-A.
CYP4731A3 bioactivates selectivin-A Notably, CYP4731A3 has a high level of basal expression in M. incognita
To identify the M. incognita CYP enzyme (or enzymes) that bioactivate tissue, falling within the top 3.5% of all expressed genes, and it is the
selectivins, we exploited the yeast-based CYP expression system that third most highly expressed CYP of the 31 CYP enzymes we identified
we used to validate C. elegans CYP-35C1 as a selectivin-bioactivating in the genome43 (Supplementary Table 6). Furthermore, CYP4731A3

Nature | Vol 618 | 1 June 2023 | 107


Article
a c mAU per mg dry weight against non-nematode phyla, this new class of nematicides has the
3
potential to selectively control a wide variety of PPNs. Unlike several
CYP4731A3 0 250 500
new nematicides on the market, which are repurposed fungicides or
Sel-A 320
P < 0.01 M1 M2 M3
insecticides9,13, the selectivins are ‘true nematicides’ that selectively
2 300
–log10(P value)

+ sel-A
target nematodes.

EV
280

Wavelength (nm)
260 Nematicides are deployed in metric tonne quantities to protect
crops, therefore they must have scalable synthesis routes and be inex-
1
M2 M3 Sel-A 320 pensive to produce. The synthesis scheme we chose to produce selec-

CYP4731A3
300
tivin analogues satisfies many of the criteria for a large-scale industrial

+ sel-A
280
0 260 process. Selectivin synthesis using this method requires only two steps,
–0.5 0 0.5 two easily accessible solvents, moderate reaction temperatures and
log2(fold difference in growth) pressures, and no transition metal catalysts. Furthermore, the synthesis

0
1
2
3
4
5
9
0
1
2.
2.
2.
2.
2.
2.
6.
7.
7.
relative to EV control
Retention time (min) of selectivin-A and selectivin-E uses inexpensive, easily accessible and
b d relatively non-toxic starting reagents. The yields we report here are
Relative growth
0 1 EV
P = 1.821 × 10–9 two-step yields; intermediates were not purified or isolated between
steps, which is advantageous for large-scale synthesis.
EV In recent years, it has become more common for soil-beneficial
CYP-4731A3
+ CYP4731A3 microbes such as entomopathogenic bacteria, fungi and nematodes, as
well as plant-growth-promoting rhizobacteria (PGPR) and mycorrhizal
0

12.5

25

50

100

0 5,000 10,000 15,000


[Sel-A] (μM) AUC (M2 + M3) fungi, to be applied to soil or seeds as part of an integrated pest manage-
ment strategy44–46. Ideally, new nematicides will complement, rather
Fig. 5 | M. incognita CYP4731A3 bioactivates selectivin-A. a, Volcano plot of
than antagonize, these organisms. It is notable that our lead nematicide
the yeast-based M. incognita CYP expression screen data. The x axis shows the
selectivin-E had no effect on either the PGPR Pseudomonas simiae or
log 2-transformed fold difference in growth between the EV control and 19
M. incognita CYP-expressing yeast strains resulting from treatment with 50 µM
the soil-beneficial gastropod-killing nematode P. hermaphrodita, which
sel-A. The y axis shows log10 -transformed P values obtained from unpaired suggests that it could be used to control PPNs in combination with
two-tailed Student’s t-tests comparing mean relative growth values between soil-beneficial microbes without compromising their utility. Our other
CYP-expressing strains and EV control. Each data point corresponds to an lead nematicide, selectivin-A, was also innocuous against P. simiae and
individual yeast strain expressing a specific M. incognita CYP enzyme. The data two distinct fungal species. However, despite demonstrating greater
point above the orange dotted line has a P value of 0.0034 and corresponds to nematode selectivity than the majority of the commercial nemati-
the strain expressing M. incognita CYP4731A3. Raw screening data can be found cides examined, selectivin-A was lethal to P. hermaphrodita and other
in Supplementary Table 5. n = 6 BRs. b, Sel-A dose–response for yeast expressing free-living nematodes. This property of selectivin-A does not preclude
CYP4731A3 and yeast carrying an EV that does not express CYP4731A3. it from commercial development given that several next-generation
Dose–response data are an average across 13 and 11 BRs for EV and CYP4731A3- commercial nematicides are also lethal to free-living nematodes
expressing yeast, respectively. c, Biomass-normalized and background- (for example, abamectin, fluopyram and tioxazafen). However, its
corrected HPLC chromatograms of lysates from yeast expressing CYP4731A3
potentially adverse effects on beneficial soil-dwelling nematode com-
or EV control yeast treated with 100 µM sel-A. Unmodified sel-A and metabolites
munities should be minimized. In principle, one way to mitigate the
M2 and M3 are indicated. MS data for HPLC-purified M1–M3 fractions can be
negative effects of selectivin-A on beneficial nematodes is to formulate
found in Extended Data Table 3 and Supplementary Table 1. d, HPLC-DAD
quantification of total M2 and M3 in the lysates of yeast expressing M. incognita
it as a seed treatment, thereby confining its activity to a concentrated
CYP4731A3 and yeast carrying EV that does not express CYP4731A3 (n = 4 BRs). zone around the seed and developing seedling. Indeed, both abamectin
AUC is area under the curve for M2 and M3 combined, calculated at 260 nm. and tioxazafen are formulated as seed treatments47,48.
Data are presented as the mean ± s.e.m. The P value was obtained from an Our work demonstrates that worm-bioactivated compounds can be
unpaired one-tailed Student’s t-test comparing the means of EV control and broadly effective against disparate worm species. This result is perhaps
CYP4731A3-expressing yeast. M1 was not detectable. unexpected because broad-spectrum activity of a worm-activated
compound will depend on low interspecies and intraspecies variability
in enzyme expression and activity. Indeed, drug metabolism commonly
expression is induced when nematodes were treated with commercial varies among individuals of the same species49. Thus, it would not be
nematicides43. Taken together, these results suggest that CYP4731A3 surprising if a pro-nematicide approach was overlooked by industrial
may bioactivate selectivin-A in vivo. None of the 19 CYP-expressing discovery programmes. Although we focused on PPN control, this
yeast strains treated with selectivin-E had perturbed growth (Sup- approach could also be applied to anthelmintics for the treatment of
plementary Table 5). We speculate that selectivin-E may be bioacti- worms that parasitize humans, livestock and companion animals50.
vated either by an untested CYP enzyme or by a combination of CYP
enzymes in a step-wise fashion. Alternatively, selectivin-E may have
a CYP-independent mechanism of action. Regardless, we identified Online content
a target CYP enzyme for the lead nematicide selectivin-A, which will Any methods, additional references, Nature Portfolio reporting summa-
facilitate lead optimization, and we established CYP-mediated bioac- ries, source data, extended data, supplementary information, acknowl-
tivation as a new approach to achieve selective killing of nematodes. edgements, peer review information; details of author contributions
and competing interests; and statements of data and code availability
are available at https://doi.org/10.1038/s41586-023-06105-5.
Discussion
Here we reported our discovery of the nematicidal utility and mode
1. Tilman, D., Balzer, C., Hill, J. & Befort, B. L. Global food demand and the sustainable
of action of the selectivins, which is a nematicide class that can kill intensification of agriculture. Proc. Natl Acad. Sci. USA 108, 20260–20264 (2011).
through metabolic bioactivation in nematodes. From a small set of ana- 2. Hunter, M. C., Smith, R. G., Schipanski, M. E., Atwood, L. W. & Mortensen, D. A. Agriculture
logues, we identified promising lead compounds that can effectively in 2050: recalibrating targets for sustainable intensification. Bioscience 67, 386–391
(2017).
control the PPN M. incognita. Given the broad-spectrum activity of 3. Cunningham, S. A. et al. To close the yield-gap while saving biodiversity will require
the selectivins against diverse nematode species, and their innocuity multiple locally relevant strategies. Agric. Ecosyst. Environ. 173, 20–27 (2013).

108 | Nature | Vol 618 | 1 June 2023


4. Popp, J., Petö, K. & Nagy, J. Pesticide productivity and food security. A review. Agron. 30. Ryan, E. et al. Evidence for the in vitro bioactivation of aminopyrazole derivatives: trapping
Sustain. Dev. 33, 243–255 (2013). reactive aminopyrazole intermediates using glutathione ethyl ester in human liver
5. Oerke, E.-C. Crop losses to pests. J. Agric. Sci. 144, 31–43 (2006). microsomes. Chem. Res. Toxicol. 28, 1747–1752 (2015).
6. Singh, S. K., Hodda, M. & Ash, G. J. Plant-parasitic nematodes of potential phytosanitary 31. Cooper, A. J. L. & Hanigan, M. H. Metabolism of glutathione S-conjugates: multiple
importance, their main hosts and reported yield losses. EPPO Bull. 43, 334–374 (2013). pathways. Compr. Toxicol. https://doi.org/10.1016/B978-0-12-801238-3.01973-5 (2018).
7. Abad, P. et al. Genome sequence of the metazoan plant-parasitic nematode Meloidogyne 32. Ferguson, G. D. & Bridge, W. J. The glutathione system and the related thiol network in
incognita. Nat. Biotechnol. 26, 909–915 (2008). Caenorhabditis elegans. Redox Biol. 24, 101171 (2019).
8. Jones, R. K. in Nematology in South Africa: A View from the 21st Century (eds Fourie, H. et al.) 33. Blum, R., Meyer, K. C., Wünschmann, J., Lendzian, K. J. & Grill, E. Cytosolic action of
129–150 (Springer, 2017). phytochelatin synthase. Plant Physiol. 153, 159–169 (2010).
9. Desaeger, J., Wram, C. & Zasada, I. New reduced-risk agricultural nematicides—rationale 34. Essig, Y. J., Webb, S. M. & Stürzenbaum, S. R. Deletion of phytochelatin synthase modulates
and review. J. Nematol. 52, e2020-911 (2020). the metal accumulation pattern of cadmium exposed C. elegans. Int. J. Mol. Sci. 17, 257
10. EU Pesticides Database, https://ec.europa.eu/food/plant/pesticides/eu-pesticides-database/ (2016).
start/screen/active-substances (European Commission). 35. Giustarini, D., Milzani, A., Dalle-Donne, I., Tsikas, D. & Rossi, R. N-Acetylcysteine ethyl
11. NPIC Product Research Online (NPRO), http://npic.orst.edu/NPRO/# (National Pesticide ester (NACET): a novel lipophilic cell-permeable cysteine derivative with an unusual
Information Center, accessed 25 July 2022). pharmacokinetic feature and remarkable antioxidant potential. Biochem. Pharmacol. 84,
12. Jordan, S., Nischwitz, C., Ramirez, R. & Gordillo, L. F. Managing the spread of alfalfa stem 1522–1533 (2012).
nematodes (Ditylenchus dipsaci): the relationship between crop rotation periods and 36. Trudgill, D. L. & Blok, V. C. Apomictic, polyphagus root-knot nematodes: exceptionally
pest reemergence. Nat. Resour. Model. 30, e12083 (2017). successful and damaging biotrophic root pathogens. Annu. Rev. Phytopathol. 39, 53–77
13. d’Errico, G., Giacometti, R., Roversi, P. F., d’Errico, F. P. & Woo, S. L. Mode of action and (2001).
efficacy of iprodione against the root-knot nematode Meloidogyne incognita. Ann. Appl. 37. Oka, Y. From old-generation to next-generation nematicides. Agronomy 10, 1387 (2020).
Biol. 171, 506–510 (2017). 38. Devguard 500SC Label,https://za.uplonline.com/download_links/
14. Burns, A. R. et al. Caenorhabditis elegans is a useful model for anthelmintic discovery. awyXPTiWs8OT3dGCErYdw3nnA3nLnwxpCpUn7jzb.pdf (deVGen/UPL).
Nat. Commun. 6, 7485 (2015). 39. Asif, M., Rehman, B., Parihar, K., Ganai, M. A. & Siddiqui, M. A. Effect of various physico-
15. Culetto, E. et al. The Caenorhabditis elegans unc-63 gene encodes a levamisole- chemical factors on the incidence of root knot nematode Meloidogyne spp. infesting
sensitive nicotinic acetylcholine receptor α subunit. J. Biol. Chem. 279, 42476–42483 tomato in District Aligarh (Uttar Pradesh) India. J. Plant Sci. 10, 234–243 (2015).
(2004). 40. Barker, K. R., Schmitt, D. P. & Imbriani, J. L. in An Advanced Treatise on Meloidogyne
16. Hu, Y., Xiao, S. H. & Aroian, R. V. The new anthelmintic tribendimidine is an l-type Volume II: Methodology (eds Barker, K. R. et al.) 135–148 (North Carolina State University
(levamisole and pyrantel) nicotinic acetylcholine receptor agonist. PLoS Negl. Trop. Dis. Department of Plant Pathology and the United States Agency for International
3, e499 (2009). Development, 1985).
17. Driscoll, M., Dean, E., Reilly, E., Bergholz, E. & Chalfie, M. Genetic and molecular analysis 41. Marion, M. J., Hantz, O. & Durantel, D. in Hepatocytes: Methods and Protocols (ed. Maurel, P.)
of a Caenorhabditis elegans β-tubulin that conveys benzimidazole sensitivity. J. Cell Biol. 261–272 (Humana Press, 2010).
109, 2993–3003 (1989). 42. Rocher, F. et al. Salicylic acid transport in Ricinus communis involves a pH-dependent
18. Dent, J. A., Smith, M. M., Vassilatis, D. K. & Avery, L. The genetics of ivermectin resistance carrier system in addition to diffusion. Plant Physiol. 150, 2081–2091 (2009).
in Caenorhabditis elegans. Proc. Natl Acad. Sci. USA 97, 2674–2679 (2000). 43. Wram, C. L., Hesse, C. N. & Zasada, I. A. Transcriptional response of Meloidogyne incognita
19. Kaminsky, R. et al. A new class of anthelmintics effective against drug-resistant nematodes. to non-fumigant nematicides. Sci Rep. 12, 9814 (2022).
Nature 452, 176–180 (2008). 44. Dara, S. K. The new integrated pest management paradigm for the modern age. J. Integr.
20. Guest, M. et al. The calcium-activated potassium channel, SLO-1, is required for the action Pest. Manag. 10, 12 (2019).
of the novel cyclo-octadepsipeptide anthelmintic, emodepside, in Caenorhabditis elegans. 45. Cardarelli, M., Woo, S. L., Rouphael, Y. & Colla, G. Seed treatments with microorganisms
Int. J. Parasitol. 37, 1577–1588 (2007). can have a biostimulant effect by influencing germination and seedling growth of crops.
21. Kawasaki, I., Jeong, M. H., Oh, B. K. & Shim, Y. H. Apigenin inhibits larval growth of Plants 11, 259 (2022).
Caenorhabditis elegans through DAF-16 activation. FEBS Lett. 584, 3587–3591 (2010). 46. Migunova, V. D. & Sasanelli, N. Bacteria as biocontrol tool against phytoparasitic nematodes.
22. Rand, J. B. & Russell, R. L. Choline acetyltransferase-deficient mutants of the nematode Plants 10, 389 (2021).
Caenorhabditis elegans. Genetics 106, 227–248 (1984). 47. Slomczynska, U. et al. in Discovery and Synthesis of Crop Protection Products (eds.
23. Hu, Y., Platzer, E. G., Bellier, A. & Aroian, R. V. Discovery of a highly synergistic anthelmintic Maienfisch, P. & Stevenson, T.) 129–147 (American Chemical Society, 2015).
combination that shows mutual hypersusceptibility. Proc. Natl. Acad. Sci. USA 107, 48. Jensen, J. P., Kalwa, U., Pandey, S. & Tylka, G. L. Avicta and clariva affect the biology of
5955–5960 (2010). the soybean cyst nematode, Heterodera glycines. Plant Dis. 102, 2480–2486 (2018).
24. Burns, A. R. et al. A predictive model for drug bioaccumulation and bioactivity in 49. Ahmed, S., Zhou, Z., Zhou, J. & Chen, S.-Q. Pharmacogenomics of drug metabolizing
Caenorhabditis elegans. Nat. Chem. Biol. 6, 549–557 (2010). enzymes and transporters: relevance to precision medicine. Genomics Proteomics
25. Ortiz de Montellano, P. R. Cytochrome P450-activated prodrugs. Future Med. Chem. 5, Bioinformatics 14, 298–313 (2016).
213–228 (2013). 50. Nixon, S. A. et al. Where are all the anthelmintics? Challenges and opportunities on the
26. Leung, M. C. K., Goldstone, J. V., Boyd, W. A., Freedman, J. H. & Meyer, J. N. Caenorhabditis path to new anthelmintics. Int. J. Parasitol. Drugs Drug Resist. 14, 8–16 (2020).
elegans generates biologically relevant levels of genotoxic metabolites from aflatoxin
B1 but not benzo[a]pyrene in vivo. Toxicol. Sci. 118, 444–453 (2010). Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in
27. Harlow, P. H., Perry, S. J., Stevens, A. J. & Flemming, A. J. Comparative metabolism of published maps and institutional affiliations.
xenobiotic chemicals by cytochrome P450s in the nematode Caenorhabditis elegans.
Sci Rep. 8, 13333 (2018). Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this
28. Porter, T. D. New insights into the role of cytochrome P450 reductase (POR) in microsomal article under a publishing agreement with the author(s) or other rightsholder(s); author
redox biology. Acta Pharm. Sin. B 2, 102–106 (2012). self-archiving of the accepted manuscript version of this article is solely governed by the
29. Kalgutkar, A. S. et al. Reactive metabolite trapping studies on imidazo- and terms of such publishing agreement and applicable law.
2-methylimidazo[2,1-b]thiazole-based inverse agonists of the ghrelin receptor. Drug Metab.
Dispos. 7, 1375–1388 (2013). © The Author(s), under exclusive licence to Springer Nature Limited 2023

Nature | Vol 618 | 1 June 2023 | 109


Article
Methods in an identical manner; however, WT worms fed HT115(DE3) E. coli
carrying the empty RNAi vector L4440 were used. Dose–response
Free-living nematode strains and culture methods assays with the CYP-35C1 deletion mutant were carried out the same
The C. elegans WT N2 strain, the MJ69 strain harbouring the temperature- way as for the WT control assays.
sensitive emb-8(hc69) allele and the RB1993 strain harbouring the The dose–response experiments for the anthelmintic-resistant and
cyp-35c1(ok2628) deletion allele were obtained from the C. elegans nematicide-resistant mutants were carried out as described for the L1
Genetics Center (CGC, University of Minnesota). C. briggsae strain dose–response assays. One notable exception is the aldicarb-resistant
AF16 and P. pacificus strain PS312 were both obtained from the CGC. strain PR1152. This strain grows slowly, and so the viability counts were
Rhabditophanes spp. KR3021 was obtained from Marie-Anne Félix (Insti- performed 5 days after addition of the chemical as opposed to 3 days
tute of Biology of the Ecole Normale Supérieure). P. hermaphrodita to allow the DMSO control worms to reach adulthood.
strain B178 was obtained from R. Rae (Liverpool John Moores University). At least three biological replicates were performed for each dose–
The following anthelmintic-resistant or nematicide-resistant C. elegans response assay. For each biological replicate, two technical replicates
strains were used for experiments: VC731 (unc-63(ok1075)), PR1152 were performed and the numbers of viable animals for each technical
(cha-1(p1152)), RP2674 (sdhc-1(tr393)), CB3474 (ben-1(e1880)), RB2119 replicate were combined (that is, about 50 worms assayed per con-
(acr-23(ok2804)), NM1968 (slo-1(js379)), CF1038 (daf-16(mu86)), and centration). The number of viable worms at each concentration was
DA1316 (avr-14(ad1302);avr-15(vu227);glc-1(pk54)). With the exception divided by the corresponding DMSO control value to give the rela-
of RP2674, which was obtained from a genetic screen performed in our tive viability for each concentration. The relative viability values were
laboratory, all of these strains were obtained from the CGC. With the then averaged across the biological replicates. Where appropriate, the
exception of C. elegans strain MJ69, all nematode species and strains lethal concentration 50 (LC50) values were calculated using GraphPad
were cultured and propagated at 20 °C using standard techniques51. Prism (v.6.0). The concentration values were log-transformed and a
C. elegans strain MJ69 was propagated at the permissible temperature four-parameter logistic curve was fitted to the dose–response data
of 15 °C. We found that using solid NGM agar plates with thin bacte- by nonlinear regression, from which the LC50 values were extracted.
rial lawns and high nematode densities promoted growth of Rhabdi-
tophanes spp. KR3021. C. briggsae, P. pacificus and Rhabditophanes spp. KR3021 dose–
response experiments
Commercial chemical sources Dose–response assays and quantification for C. briggsae, P. pacificus
Selectivin-A, selectivin-B, selectivin-G, selectivin-J, selectivin-K and and Rhabditophanes spp. KR3021 were carried out as described for the
tioxazafen were purchased from ChemBridge. Selectivin-C was pur- C. elegans L1 dose–response assays; however, the selectivin-induced
chased from Vitas-M and MilliporeSigma. Selectivin-D was purchased phenotypes in P. pacificus and Rhabditophanes, even at the highest
from MolPort. 4-(4-Chlorophenyl)-1H-imidazole-2-thiol was purchased concentrations, were a combination of lethality and larval arrest with
from Life Chemicals. Abamectin, fenamiphos, fluopyram, iprodione, severe sickness. Therefore, for these dose–response assays, the num-
oxamyl, spirotetramat and iodoacetamide (IAA) were purchased from ber of animals that reached the L2 stage or older was quantified as
MilliporeSigma. Fluensulfone was purchased from Cayman Chemical. opposed to the number of viable worms. The arrested animals appeared
extremely sick and would probably die before reaching reproductive
C. elegans dose–response experiments adulthood. Therefore, this arrested phenotype was considered to be
A HB101 bacterial suspension in liquid NGM was prepared by concen- practically analogous to death.
trating a saturated overnight HB101 culture twofold with liquid NGM
(see ref. 14 for the NGM recipe). Forty microlitres of this bacterial sus- P. hermaphrodita dose–response experiments
pension was added to each well of a 96-well flat-bottom culture plate, In brief, 24-well solid-medium chemical assay plates were prepared
after which approximately 25 synchronized L1 worms, in 10 µl of M9 as previously described52. Each well of the assay plates contained 1 ml
buffer (see ref. 52 for the recipe), were added to each well. The synchro- of solid MYOB medium with dissolved chemical at the appropriate
nized L1 worms were obtained from an embryo preparation performed concentration or DMSO alone for untreated solvent control. The final
the previous day (see ref. 52 for the protocol). For the L1 assays, 0.5 µl DMSO concentration was 0.5% in every well. After thoroughly drying
of chemical solution (or DMSO alone) was immediately added to the the plates in a sterile flow hood, the wells were seeded with 25 µl of an
wells using a multichannel pipette; the final DMSO concentration was overnight culture of OP50 E. coli and allowed to sit at room temperature
1% (v/v). The culture plate was sealed with Parafilm and placed in a box for 24 h. Using glass pipettes, P. hermaphrodita B178 nematodes (mixed
with several wet paper towels to prevent evaporation. The worms were stage) were washed off of several starved 10-cm culture plates with M9
incubated for 3 days at 20 °C with shaking at 200 r.p.m., and the num- buffer (see ref. 52 for the recipe) and collected in clean glass culture
ber of viable animals was counted. A dead worm was considered any tubes. Larger worms were allowed to settle to the bottom of the tubes
worm that failed to move after vigorous agitation of the plate and that by gravity and young larvae (mostly L1 to L2 stage) were collected into
appeared morphologically ‘dead’, that is, clear appearance and unre- separate clean glass culture tubes. The nematode concentration was
solved internal structures. Although the counts were performed after adjusted to about 15 worms in 20 µl of M9 buffer. Next 20 µl of worm
3 days of incubation in the chemical, it was noted that the L1 worms suspension was added to each well of the 24-well chemical assay plates.
were dead within 24 h of the addition of the chemicals. The M9 buffer was allowed to soak into the agar substrate and then the
For the emb-8 knockdown dose–response assays, the MJ69 strain was plates were sealed with Parafilm and stored upside down in a Tupper-
used and worms were fed HT115(DE3) Escherichia coli harbouring a RNAi ware box at 20 °C for 3 days. After 3 days, the number of moving worms
feeding vector expressing dsRNA targeting the emb-8 gene. The HT115 in each well was counted. The number of moving worms at each chemi-
suspension was made by concentrating an overnight bacterial culture cal concentration was divided by the corresponding DMSO control
grown at 20 °C, with an OD600 of 0.8, 5-fold with liquid NGM containing value to give the relative motility for each concentration. The relative
1 mM IPTG and 100 µg ml–1 carbenicillin. The HT115 cells were induced motility values were then averaged across three biological replicates.
with 1 mM IPTG for 1 h at 20 °C before concentrating with NGM. Syn-
chronized L1 worms were grown for 2 days at the non-permissive tem- C. oncophora dose–response experiments
perature of 25 °C until they reached young adulthood, at which point the Fresh cattle faeces containing eggs of an ivermectin-resistant strain
chemical was added to the wells using a multichannel pipette. Viability of C. oncophora were supplied by D. Colwell and D. Gray (Lethbridge
was scored 1 day later. The WT control experiments were performed Research Station, Agriculture and Agri-Food Canada). Established
methods were used to carry out the experimental cattle infections53, absorbance value at the first time point from all absorbance values
and these methods were performed in accordance with the Canadian of the growth curve. Using GraphPad Prism (v.6.0), four-parameter
Council of Animal Care regulations (licence numbers AC13-0157 (Uni- logistic curves were fitted to the growth data by nonlinear regression.
versity of Calgary) and 1730 (Agriculture AgriFood Canada Research The time point at which absorbance was equal to 95% of the absorbance
Station, Lethbridge)). Cattle faeces containing C. oncophora eggs were maximum of the curve was determined for each untreated control, and
anaerobically stored at room temperature for a maximum of 6 days the AUC was calculated up to that time point for both the untreated
before use. Eggs were isolated from faeces using a standard saturated control and the corresponding chemical-treated samples. The AUC
salt flotation method54 immediately before the egg hatch assay. Next for each treatment concentration was divided by the AUC for the cor-
80 µl of distilled and deionized water was added to each well of a 96-well responding untreated control, resulting in a relative growth value for
culture plate, and then 1 µl of chemical at the appropriate concentration each concentration tested. At least three biological replicates were
in DMSO was added to each well using a multichannel pipette. Approxi- performed for each dose–response experiment, and the relative growth
mately 50 eggs were added per well in 20 µl of water for a final volume values were averaged across the replicates.
of 100 µl in each well; the final DMSO concentration was 1% (v/v). The
eggs were incubated in the chemicals for 2 days at room temperature, Bioinformatics identification of M. incognita CYP proteins
after which hatching was stopped by the addition of 1 µl iodine tincture WormBase ParaSite60 (v.WBPS16) BioMart was used to query the
to each well. The number of hatched larvae was counted at each con- M. incognita v3 genome (NCBI BioProject identifier PREJEB8714)61
centration, and eggs that failed to hatch were scored as dead. Relative and to extract all protein sequences containing a CYP superfamily
viability values were calculated by dividing the fraction of eggs that domain (InterPro identifier IPR036396). Incomplete sequences and
hatched at each concentration by the fraction of eggs that hatched those shorter than 450 amino acids in length were removed. This
in the corresponding DMSO control well. Three biological replicates resulted in a set of 31 proteins, all of which contained a CYP, E-class,
were performed for each dose–response experiment, and the relative group I domain (InterPro identifier IPR002401). These sequences were
viability values were averaged across the biological replicates. The aligned using Clustal Omega62 and organized into homologous group-
average hatch rate for the DMSO control wells was greater than 93% ings based on protein sequence identity (Supplementary Table 3). In
for both biological replicates. total, 19 M. incognita CYP enzymes that were representative of the
diversity of sequences within the family were selected for cDNA syn-
Meloidogyne spp. in vitro dose–response experiments thesis and further analysis. All 19 CYP sequences were codon-optimized
M. incognita (Kofoid & White) Chitwood Race 1 (originally isolated in for yeast expression before synthesis (see Supplementary Table 4 for
Maryland) maintained on pepper (Capsicum annuum L.) cv. PA-136 the sequences).
and M. chitwoodi (originally isolated from Washington) maintained
on tomato (Solanum lycopersicum) were grown in a greenhouse CYP-expressing yeast strain construction
as previously described55,56. M. hapla (originally isolated from a muck The C. elegans cyp-35c1 and emb-8 cDNA sequences were obtained from
soil sample in Ste-Clotilde, Quebec, Canada) were maintained on WormBase63 and were codon-optimized for yeast expression (see Sup-
tomato (S. lycopersicum ‘Rutgers’). Infective J2 larvae were collected plementary Table 4 for the sequences). The codon-optimized cDNAs
as described in ref. 57. The microwell dose–response experiments were were synthesized by Integrated DNA Technologies. Using common
conducted in 96-well polystyrene plates, similar to previously described molecular biology techniques, cyp-35c1 and emb-8 cDNA were cloned
protocols55,58. In brief, each well received approximately 35–50 J2 larvae into the American Type Culture Collection (ATCC) p426 GAL1 expres-
in 10 µl of sterile distilled water (SDW), followed by the addition of 190 µl sion vector (URA3(-) rescuing marker; 2µ origin of replication) and
SDW or of 190 µl of SDW containing dissolved chemical or DMSO alone. the ATCC p425 GAL1 expression vector (LEU2(-) rescuing marker; 2µ
The chemicals were tested at 5, 15 and 45 µM concentrations, and the origin of replication), respectively. S. cerevisiae BY4741 (MATa his3Δ1
final concentration of DMSO in each well was 0.5% (v/v). The wells were leu2Δ0 met15Δ0 ura3Δ0) was co-transformed with the cyp-35c1 and
covered with a plastic adhesive strip, and the lids of the plates were emb-8 expression vectors and co-transformants were selected for on
sealed with Parafilm. The nematodes were incubated in chemicals for URA(-) and LEU(-) SD medium59.
2 days at 26 °C, at which point the J2 larvae were rinsed twice with SDW The 19 M. incognita CYP-expression vectors were constructed by
and incubated in the second SDW rinse for an additional day. After Twist Bioscience using the ATCC p416 GAL1 expression vector (URA3(-)
incubation, the fraction of viable nematodes at each concentration rescuing marker; CEN origin of replication). The 19 CYP expression
was calculated by dividing the number of motile nematodes by the vectors were individually transformed into S. cerevisiae BY4741 (MATa
total number of nematodes in the well. Relative viability was calculated his3Δ1 leu2Δ0 met15Δ0 ura3Δ0) and transformants were selected for
by dividing the fraction of viable nematodes at a given treatment con- on URA(-) SD medium59.
centration by the fraction of viable nematodes in the untreated DMSO
control sample. The average relative viability was calculated across six Dose–response experiments with CYP-expressing yeast
biological replicates. Yeast SD medium supplemented with the appropriate amino acids or
nucleobases was used for yeast growth and dose–response assays59.
WT S. cerevisiae (budding yeast) dose–response experiments The CYP-35C1 and EMB-8 co-expressing strain, as well as a strain car-
A saturated culture of the yeast strain RY0568 was diluted to an OD600 rying both of the empty p425 GAL1 and p426 GAL1 expression vectors,
of 0.015 with fresh YPD medium (see ref. 59 for the recipe). Next 100 µl were grown in URA(-) and LEU(-) selection medium. The M. incognita
of this dilute yeast suspension was added to each well of a 96-well plate. CYP4731A3-expressing strain, as well as a strain carrying an empty p416
The yeast was grown for 4 h at 30 °C with shaking at 140 r.p.m. Using a GAL1 expression vector, were grown in URA(-) selection medium. The
multichannel pipette, 1 µl of chemical solution was added to each well following methods apply to the CYP-35C1 and EMB-8 co-expressing
to achieve the desired final concentrations. The final DMSO concentra- strain, the M. incognita CYP4731A3-expressing strain and to both empty
tion was 1% (v/v). The microwell plate was sealed with a clear plastic vector (EV) strains. In brief, 5 ml of an overnight saturated culture was
adhesive strip and then loaded into a TECAN plate reader set at 30 °C. grown in appropriate selection medium containing 2% raffinose at 30 °C
National Instruments LabVIEW 2013 software (v.13.0) was used for data with shaking. The culture was diluted to an OD600 of 0.03 in appropri-
acquisition. The OD600 of each well was measured every 15 min over a ate selection medium containing 2% raffinose. Yeast were grown for
48-h period, and the plate was shaken intermittently throughout the 4 h at 30 °C with shaking. To induce expression, galactose was added
run. The growth data were background-corrected by subtracting the to a final concentration of 2%. Next 100 µl of yeast suspension was
Article
added to each well of a 96-well culture plate. Yeast were grown for 4 h at values from the 0-h time point. The relative growth for each chemical
30 °C with shaking at 140 r.p.m. A multichannel pipette was used to add treatment was defined by normalizing the corrected OD600 value to
1 µl of chemical dissolved in DMSO at the appropriate concentration that of the corresponding untreated DMSO control. All dose–response
to each well (1% DMSO v/v). DMSO alone was added to the untreated assays were performed in technical triplicate and relative growth levels
control wells. The plate was sealed with a clear plastic adhesive strip were averaged across replicates.
and loaded into a TECAN plate reader set at 30 °C. The OD600 of each
well was measured every 15 min over a 48-h period, and the plate was D. rerio (zebrafish) dose–response experiments
shaken intermittently throughout the run. Dose–response assays were Fish were maintained at 28.5 °C on a 14–10-h light–dark cycle and staged
repeated at least three times (biological replicates). Data analysis was according to hours post fertilization (h.p.f.). For each biological rep-
performed in an identical manner to that described for the WT yeast licate, eggs from AB WT fish were collected at 4 h.p.f. At 3 days post
dose–response experiments (see above). fertilization (d.p.f.), both male and female embryos were arrayed in
24-well culture plates at 10 per well, which is a commonly used sample
Yeast-based screen for M. incognita CYP enzymes that size for this type of dose–response experiment. Next 5 µl of the chemi-
bioactivate selectivin-A cal dissolved in DMSO at the appropriate concentration was added
Selectivin-A and selectivin-E sensitivity assays with the 19 M. incognita to 1 ml of E3 medium and then intensively vortexed to mix. Water was
CYP-expressing yeast strains were carried out as described for the removed from the embryos in the wells and 1 ml of chemical-treated
dose–response experiments with CYP-expressing yeast (see above), water was transferred to each of the wells. The DMSO control wells
with the following two exceptions: (1) overnight cultures were diluted contained DMSO alone. The final DMSO concentration in every well
to 0.05 instead of 0.03 and grown for 2 h instead of 4 h before adding was 0.5% (v/v). The culture plates were sealed with Parafilm, wrapped
galactose; (2) after the addition of galactose, yeast cells were grown in aluminium foil and incubated at 28.5 °C for 72 h. After incubation,
for 2 h instead of 4 h before addition of chemical. A strain carrying the viability was assessed. Using a stereomicroscope, lethality was scored
empty p416 GAL1 expression vector was included as the EV control. Each either by appearance of whole-body necrosis or the absence of both a
strain was treated with 50 µM selectivin-A, 50 µM selectivin-E or DMSO heartbeat and touch-evoked response. Relative viability was calculated
alone as an untreated control (1% v/v). All experiments were repeated six by dividing the number of viable embryos in the treatment wells by the
times (biological replicates). Data analysis was performed in an identi- number of viable embryos in the DMSO control well. Three biological
cal manner to that described for the WT yeast dose–response experi- replicates were performed for each dose–response experiment, and
ments, and relative growth values for each strain and each condition the relative viability values were averaged across the three replicates.
were calculated the same way. Average relative growth values for each The zebrafish ethics protocol is 100005273, approved by the Animal
strain were normalized to the EV control, and then log2-transformed to Care Committee at The Hospital for Sick Children, Canada. Fish were
give log2 fold difference in relative growth. For each strain, a P value was chosen at random for inclusion in the dose–response assays. The assays
obtained from an unpaired two-tailed Student’s t-test comparing the were blinded such that the experimenter did not have any knowledge
mean relative growth values between the CYP-expressing strain and the of the compounds being tested.
EV control. The P values were log10-transformed. The log-transformed
data were plotted as a volcano plot. A P value less than 0.01 indicates HEK293 cell dose–response experiments
a significant difference. HEK293 cells (Thermo Fisher, R71007) were seeded into 96-well plates
at 5,000 cells per well in 100 µl total volumes of DMEM, 10% FBS and
C. albicans dose–response experiments 1% penicillin–streptomycin medium and grown overnight at 37 °C in
Compound potency against C. albicans (SN95) was assessed using two- the presence of 5% CO2. Compounds (0.5-µl volumes from appropriate
fold dose–response assays following a standard protocol. In brief, YPD source plates) were then added to cells, and growth was continued for
medium was inoculated with about 1 × 103 cells per ml from saturated an additional 48 h. Following growth, 10 µl of CellTiter-Blue viability
overnight cultures. Assays were performed in 384-well, flat-bottom reagent (Promega) was added to each well, and plates were incubated
microtitre plates (Corning) in a final volume of 40 µl per well. Each drug for an additional 4 h at 37 °C in the presence of 5% CO2. Fluorescence
was added to wells in a twofold concentration gradient from 100 µM to measurements (560 nm excitation/590 nm emission) were then per-
0 µM. Plates were then incubated in the dark at 30 °C under static condi- formed using a CLARIOstar plate reader (BMG Labtech) to quantify
tions for 48 h. End point growth was quantified by measuring the OD600 reagent reduction and to estimate cell viability. Fluorescence measure-
using a Molecular Devices SpectraMax Plus 384 Microplate Reader with ments were corrected for background from medium. Relative growth
SoftMax Pro 7 software, and the results were corrected for background was calculated by dividing the corrected fluorescence in the treatment
medium. Relative fungal growth for each compound treatment was wells by that measured in the corresponding DMSO control well. The
defined by normalizing to the levels observed in untreated controls. final values are an average of at least three technical replicates. The
All dose–response assays were performed in technical triplicate and HEK293 cells were authenticated by STR analysis at the Toronto Hospital
relative growth levels were averaged across replicates. for Sick Children authentication facility, and recent testing confirmed
that the cell line was negative for mycoplasma contamination.
P. simiae dose–response experiments
P. simiae WSC417 was used for all experiments. Liquid LB medium was HepaRG cell dose–response experiments
inoculated with around 1 × 105 cells per ml from saturated overnight HepaRG cells (Thermo Fisher, Gibco, cryopreserved HPRGC10) were
cultures. Assays were performed in 96-well flat-bottom microtitre seeded in a 96-well plate at 20,000 cells per well in 100 µl of DMEM-F12
plates in a final volume of 100 µl per well. Using a multichannel pipette, (Gibco) with 10% FBS (Invitrogen), 1× penicillin–streptomycin (Sigma),
the chemical was added to each well at the appropriate concentration 2 mM l-glutamine (Sigma), 1× ITS-A (Gibco) and 40 ng ml–1 dexametha-
in 0.5 µl of DMSO for a final DMSO concentration of 0.5% v/v. DMSO sone (BioShop). Subsequently, the medium was refreshed, and a dilu-
alone was added to the untreated control wells. The OD600 of each well tion series of the test compound was added to each well with a final
was measured using a BMG Labtech POLARStar Omega spectropho- total volume of 100 µl. After 48 h, CellTiter-Blue viability dye (Promega)
tometer with Omega software (v.1.20), and the plates were then incu- was added for 3 h, and viability was quantified using plate reader as
bated at 28 °C with shaking at 200 r.p.m. for 16 h. End point growth per the manufacturer’s protocol (Promega). Fluorescence was meas-
was quantified by measuring OD600 after the 16 h of incubation and ured at 560 nm excitation/590 nm emission and corrected for back-
then background-corrected by subtracting the corresponding OD600 ground from the medium. Relative growth was calculated by dividing
the corrected fluorescence in the treatment wells by that measured in study was blinded. One experimenter was responsible for the drug
the corresponding DMSO control well. The final values are an average treatments and a second experimenter was responsible for assessing
of at least three technical replicates. The HepaRG cells were authenti- any pathologies without knowing which mice were in the treatment
cated by SNP genotyping and confirmed to be negative for mycoplasma or control group.
contamination by the commercial supplier. The HepaRG cells were
used directly from the supplier. Incubations for HPLC analysis
For the C. elegans and C. briggsae L1 incubations, synchronized hatch-
D. melanogaster (fruit fly) dose–response experiments lings were obtained from an embryo preparation of gravid adults52.
Fly food in agar substrate was prepared by mixing together 100 ml of In total, 200,000 hatchlings in 500 μl of M9 buffer (see ref. 52 for
unsulfured molasses, 100 ml of cornmeal, 41.2 g of baker’s yeast and the M9 buffer recipe) were treated with 100 μM selectivin-A, 100 µM
14.8 g of agar into 1,400 ml of distilled deionized water and boiling 4-(4-chlorophenyl)-1H-imidazole-2-thiol (that is, imidazole-thiol metab-
for 30 min. The medium was allowed to cool to 56 °C, at which point olite (ITM)), 100 µM selectivin-E or DMSO alone for control purposes.
5 ml was added by syringe to plastic cylindrical fly vials. Next 10 µl of The final concentration of DMSO in all samples was 1% (v/v). Before
chemical, or DMSO alone, was added to the medium in each vial. The the incubations, the hatchlings used for the dead worm controls were
chemicals were mixed into the medium by mechanical mixing using heat-killed at 95 °C for 45 min. The incubations were carried out in
a pipette. The final DMSO concentration was 0.2% (v/v). The medium standard 1.5 ml microcentrifuge tubes on a nutating shaker at 20 °C for
was allowed to solidify at room temperature (about 22 °C) overnight. 6 h. After 6 h of incubation, the worms were transferred to the wells of
The following day (day 0), 8 pairs of male and female w1118 flies were a Pall AcroPrep 96-well filter plate (0.45 μm wwPTFE membrane, 1 ml
added to each vial so that there were 16 flies in total per vial. The vials well volume), the buffer was drained from the wells by vacuum and
were stored at room temperature for 7 days, at which point the number the worms were subsequently washed once with 600 μl of M9 buffer.
of motile flies was counted. Fly motility was scored as any observable After washing, the worms were re-suspended in 50 μl of M9 buffer
movement after the vial had been vigorously jostled. Relative motility using low-binding tips, transferred to a new 1.5-ml microcentrifuge
was calculated by dividing the number of motile flies in the treatment tube and stored frozen at –80 °C. In some cases, the incubation buffer
vials by the average number of motile flies in two DMSO control vials. was also collected and frozen at –80 °C. For the samples alkylated
On day 8, the 16 parental flies were removed from the vials and the with IAA, worms were resuspended in 50 µl of M9 buffer containing
progeny larvae were allowed to continue to grow and hatch into adult 50 mM IAA and allowed to incubate for 40 min at room temperature
flies. To assess larval viability, hatched flies were counted and discarded before freezing at –80 °C. The P. pacificus L1 incubations were carried
on days 10, 12, 14, 16, 18 and 20. The counts were summed. Relative out using the same methods as above; however, 100,000 hatchlings
viability was calculated by dividing the number of hatched flies in the were used. The Rhabditophanes incubations were carried out in a
treatment vials by the average number of hatched flies in the two DMSO similar way; however, a mixed population of 50,000 L1 and L2 larvae
control vials. The final relative motility and relative viability values are obtained through gravity separation of adults and larvae were used for
an average across three biological replicates. incubations. The M. incognita incubations were carried out the same
way but with three differences: (1) 25,000 J2 larvae were used; (2) incu-
A. thaliana greening experiments bations and washes were performed using distilled water instead of M9
Greening experiments were performed with A. thaliana seeds of WT buffer; (3) after incubation and washing, worms were resuspended in
Col-0; seeds were surface-sterilized in bleach and plated onto 0.5× MS, 25 µl of M9 buffer. M. incognita culture and J2 collection were done as
0.5% sucrose agar medium supplemented with compounds of interest previously described64.
at 5, 15 and 45 µM concentrations (0.2% DMSO (v/v/)). After 4 days of For the WT and cyp-35c1Δ mutant C. elegans young adult incuba-
stratification at 4 °C, plates were transferred to a growth chamber tions, a culture of HT115 bacteria harbouring the empty L4440 RNAi
(16 h–8 h, 150 µE m–2) and greening was recorded after 4 days. Pictures feeding vector was grown to an OD600 of 0.6 in LB medium supple-
were recorded by camera (SONY a7s) with a FE1.8/55 lens (FE 55 mm mented with 100 µg per ml ampicillin, and then induced with 1 mM
F1.8 ZA; SEL55F18Z). Three biological replicates were performed for IPTG for 1 h at 20 °C until the culture reached an OD600 of about 0.8.
each treatment. After induction, the culture was concentrated 5-fold with liquid NGM
supplemented with 1 mM IPTG and 100 μg ml–1 carbenicillin (see ref. 14
Selectivin-A mouse studies for the NGM recipe). Next 2 ml of this bacterial suspension was added
Female C57Bl/6 mice aged 6–8 weeks (bred and maintained at the to a 15-ml conical tube. For the treated samples, selectivin-A was added
animal care facility, Department of Biological Sciences, University of to the suspension to a final concentration of 100 µM and 1% DMSO
Calgary, Canada) were used. All animal experiments were approved (v/v). For the untreated control samples, DMSO was added alone to a
by the University of Calgary’s Life and Environmental Sciences Ani- 1% (v/v) final concentration. Next 1,250 L1 hatchlings, obtained from
mal Care Committee (protocol AC17-0082). All protocols for animal an embryo preparation of gravid adults52, were added to each tube in
use and euthanasia were in accordance with the Canadian Council for 0.5 ml of M9 buffer. The worms were incubated on a nutating shaker
Animal Care (Canada). Infected mice were orally gavaged with 200 for 2 days at 25 °C until they reached the young adult stage, at which
third stage Heligmosomoides polygyrus larvae (maintained in-house. point selectivin-A was added to a final concentration of 100 µM and
Original stock was a gift from A. Shostak, University of Alberta, Canada) 1% DMSO (v/v). DMSO alone was added to the untreated control sam-
and euthanized on day 22 after infection. Each group (treated versus ples. The worms were incubated for 6 h at 25 °C. After incubation, the
non-treated) had a minimum of 7 mice. Experiments of this type worms were pelleted by centrifugation and washed three times with
commonly use five mice per group, and we chose a seven-per-group M9 buffer to remove the bacteria. After the final wash, the buffer was
minimum to buffer against variability in the effects of drug treatment. aspirated down to 0.5 ml, and using low-binding tips, the worms were
Mice were littermates. Mice were treated orally with 5 daily doses of transferred to the wells of a Pall AcroPrep 96-well filter plate (0.45 μm
selectivin-A (50 mg kg–1 resuspended in DMSO). Control mice were wwPTFE membrane, 1 ml well volume). The buffer was drained from the
given DMSO only as a control. Mice were chosen at random for inclu- wells by vacuum. The worms were re-suspended in 50 μl of M9 buffer
sion in either the treatment or control groups. To reduce human error using low-binding tips, transferred to a new 1.5-ml microcentrifuge
with drug delivery, all mice in one cage (from two to five) were on the tube and stored frozen at –80 °C. The emb-8 knockdown incubations
same treatment. There were two cages per treatment group. Mice were were performed in an identical manner to the WT young adult dose–
monitored daily for visible signs of treatment-induced pathology. The response experiments, except the MJ69 strain was used in place of the
Article
WT strain, and worms were fed HT115(DE3) E. coli harbouring an RNAi A 100 µM concentration was chosen for the incubations because
feeding vector expressing dsRNA targeting the emb-8 gene. it is the highest concentration that remains soluble in the buffer and
For the WT S. cerevisiae incubations, an overnight culture of the yeast therefore maximizes the signal-to-noise ration. A 6-h incubation period
strain RY0568 was diluted to an OD600 of 0.1 in 5 ml of YPD medium and was chosen because C. elegans nematodes are obviously affected by
grown at 30 °C for 3 h until the OD600 reached 0.3 (see ref. 59 for YPD selectivin-A at this time point but they are not yet dead (by 12 h they are
recipe). This culture was diluted to an OD600 of 0.2 in 2.5 ml of YPD and dead). The incubations were performed in at least biological triplicates.
selectivin-A was added at a concentration of 100 µM (1% DMSO (v/v)).
DMSO was added alone to the untreated control samples. The culture Incubation buffer de-salting and drying
was incubated for 6 h at 30 °C until the OD600 reached 3.2. The cells were Worm, fish and fly incubation buffers were de-salted using a 96-well
transferred to a 15-ml conical tube and pelleted by centrifugation at HyperSep C8 solid phase extraction (SPE) plate with 1 ml well volume
1,000g for 5 min. Next 2 ml of the medium was collected and frozen at and 100 mg bed weight (Thermo Scientific). The SPE plates were cou-
–80 °C, leaving the cells in 0.5 ml of medium. The cells were washed pled to a vacuum manifold and vacuum pump, and a flow rate of around
twice with distilled and deionized water. After the second wash and 1 ml min–1 was used. The columns were activated with 3 ml of 100%
spin, all of the water was removed and the cells were re-suspended acetonitrile and then washed with 3 ml of distilled deionized water.
in 0.5 ml of M9 buffer. The cells were pelleted again, and all of the M9 Incubation buffers were added to each well and passed through the
buffer was removed by aspiration. The cell pellet was frozen at –80 °C. columns by vacuum. The columns were washed once with 1 ml of water.
For the CYP-expressing (and EV control) yeast strains, saturated cul- Three sequential elutions were done using 250 µl of 20% acetonitrile,
tures were grown in the appropriate SD selection medium containing 2% 250 µl of 50% acetonitrile and 250 µl of 100% acetonitrile. The eluates
raffinose. The saturated cultures were diluted to an OD600 of 0.3 in the were dried in an Eppendorf Vacufuge concentrator and then stored
appropriate SD selection medium containing 2% raffinose to achieve a frozen at –80 °C for downstream HPLC analysis.
4.95 ml final volume. The yeast was grown in glass test tubes for 4 h at Yeast incubation buffers were de-salted using Sep-Pak light C8 car-
30 °C on a rotating wheel. Next 550 µl of 20% galactose was added to tridges (Waters). The columns were activated, loaded, washed and
the culture to induce protein expression, and cells were grown for 4 h eluted in the same manner described above for the SPE plates; however,
at 30 °C on a rotating wheel. After that, 55 µl of 10 mM selectivin-A in solvent and sample was passed through the cartridges using a syringe
DMSO, or DMSO alone, was added to the cultures. The cultures were and manual force. A flow rate of about 1 ml min–1 was used. The eluates
gently vortexed and then incubated at 30 °C for 8 h on a rotating wheel. were dried and stored frozen at –80 °C for downstream HPLC analysis.
The cells were transferred to 15-ml conical tubes and pelleted at 1,000g
for 5 min. Then 5 ml of incubation buffer was removed and the cells were Sample lysis for HPLC analysis
transferred to 1.5-ml microcentrifuge tubes. The tubes were centrifuged Free-living worm and fish lysis. Frozen worm and fish samples were
at 1,000g for 2 min, and all of the medium was aspirated. The cell pellet thawed, and 50 µl of 2× lysis buffer (20 mM Tris-HCl pH 8.3, 0.2% SDS
was resuspended in 0.5 ml of ddH2O. The cells were pelleted again at and 240 µg ml–1 proteinase K) was added to the tubes. The tubes were
1,000g for 2 min, the medium was aspirated and the cells were resus- incubated for 80 min in a 56 °C water bath with vigorous vortexing
pended in 0.5 ml of M9 buffer. The cells were pelleted again, all of the every 15 min. The samples were then bath sonicated for 20 min at room
medium was removed and the cell pellet was stored frozen at –80 °C. temperature using a Branson 1510 bath sonicator. The lysates were
For the zebrafish incubations, 50 AB WT zebrafish aged 1 d.p.f. were stored frozen at –80 °C until processing by HPLC.
transferred into 0.5 ml of E3 medium in 24-well culture plates and
allowed to grow to 5 d.p.f. At 5 d.p.f., they were transferred into fresh M. incognita J2 lysis. J2 larvae were thawed, and 25 µl of 2× lysis buffer
0.5 ml E3 medium in 1.5-ml microcentrifuge tubes and either 100 µM (20 mM Tris-HCl pH 8.3, 0.2% SDS and 1,600 µg ml–1 proteinase K) was
selectivin-A or DMSO alone was added (1% DMSO (v/v)), as well as 0.05% added to the tubes. The tubes were incubated for 3 h in a 65 °C water
gelatin to break the surface tension of the water. The fish were incu- bath with vigorous vortexing every 20 min. The samples were then
bated for 6 h at 20 °C on a nutating shaker. After 6 h of incubation, the bath sonicated for 30 min using a Branson 1510 bath sonicator with
fish were transferred to the wells of a Pall AcroPrep 96-well filter plate the heater turned on. The lysates were stored frozen at –80 °C until
(0.45 μm wwPTFE membrane, 1 ml well volume) using low-binding processing by HPLC.
tips with the ends cut to allow the fish to pass through the opening.
The buffer was drained from the wells by vacuum, and the fish were WT yeast lysis. Frozen yeast pellets were thawed and re-suspended in
subsequently washed once with 600 μl of M9 buffer. After washing, 40 µl of M9 buffer containing 1 M sorbitol and 300 U ml–1 zymolase. The
the fish were re-suspended in 100 μl of M9 buffer using low-binding samples were incubated at 37 °C for 60 min with vortexing every 10 min,
tips with the ends cut to allow the fish to pass through the opening, after which 50 µl of 2× yeast lysis buffer (20 mM Tris-HCl pH 8.3, 0.2%
and transferred to a new 1.5-ml microcentrifuge tube. The fish were SDS and 720 µg ml–1 proteinase K) was added to the tubes. The tubes
quickly pelleted by centrifugation and 50 µl of M9 buffer was removed. were incubated for 2 h at 56 °C with vigorous vortexing every 10 min.
The fish were then frozen at –80 °C. The incubation buffer was also The samples were then bath sonicated for 20 min at room temperature
collected and frozen at –80 °C. using a Branson 1510 bath sonicator. The lysates were stored frozen at
For the fly larvae incubations, three third instar w1118 fly larvae were –80 °C until processing by HPLC.
transferred into 0.5 ml of PBS buffer in 1.5 ml microcentrifuge tubes.
Either 100 µM selectivin-A or DMSO alone was added (1% DMSO (v/v)). CYP-expressing yeast lysis. Yeast cell pellets were thawed. With the
The fly larvae were incubated for 6 h at 20 °C on a nutating shaker. exception of CYP-35C1-expressing yeast incubated in selectivin-A,
After 6 h of incubation, the larvae were transferred to the wells of a Pall cells were resuspended in 70 µl of M9 buffer containing 1 M sorbitol
AcroPrep 96-well filter plate (0.45 µm wwPTFE membrane, 1 ml well and 300 U ml–1 zymolase. CYP-35C1-expressing cells incubated in
volume) using low-binding tips with the ends cut to allow the larvae selectivin-A were resuspended in 42 µl of the M9, sorbitol and zymo-
to pass through the opening. The buffer was drained from the wells by lase solution (the lower volume was to account for the lower biomass
vacuum, and the larvae were subsequently washed once with 600 μl resulting from growth inhibition). The cells were then incubated at 37 °C
of M9 buffer. After washing, the larvae were transferred into 100 µl of for 1 h with vortexing every 10 min. Next 2× lysis buffer (20 mM Tris-HCl
M9 buffer using a fine hair brush. The volume of M9 was adjusted to pH 8.3, 0.2% SDS and 720 µg ml–1 proteinase K) was added to the tubes
50 µl, and the larvae were frozen at –80 °C. The incubation buffer was (50 µl for CYP-351-expressing cells incubated in selectivin-A and 100 µl
also collected and frozen at –80 °C. for all other cells), and the cells were incubated for 2 h with vortexing
every 10 min. The samples were then bath sonicated for 20 min at room for the incubation buffer samples, with two minor exceptions for the
temperature using a Branson 1510 bath sonicator. The lysates were yeast incubation buffers. (1) A YPD blank sample prepared in the same
stored frozen at –80 °C until processing by HPLC. way as the experimental yeast buffers was processed by HPLC, and the
absorbance intensity values were subtracted from the experimental
Fly larvae lysis. Frozen larvae were thawed, and 50 µl of fly lysis buffer intensities to account for absorbing material from the YPD medium.
(20 mM Tris-HCl pH 8.3, 0.2% SDS and 720 µg ml–1 proteinase K) was This was done before dry weight normalization and subtraction of
added to the tubes. The larvae were incubated for 30 min at 56 °C and the paired DMSO control absorbance intensities. (2) As only 2 out of
were then crushed with a pestle to promote lysis (the exoskeleton 2.5 ml (4/5) of yeast incubation buffer was processed, the dry weight
remained intact). The samples were incubated at 56 °C for an additional values used to normalize the yeast buffer data were multiplied by 4/5.
1.5 h with vigorous vortexing every 10 min. The samples were then bath Average AUC values were calculated across at least three experimental
sonicated for 20 min at room temperature using a Branson 1510 bath replicates.
sonicator. The lysates were stored frozen at –80 °C until processing
by HPLC. Metabolite MS
HPLC fractions containing either the selectivin-A metabolites M1, M2,
Dried incubation buffer preparation. Dried incubation buffers were M3 and M4 and ITM-alk, or the selectivin-E metabolites E-M1, E-M2 and
lysed in the same way as the free-living worm pellets; however, 50 µl of E-M3 were collected from three separate lysates, combined and dried
M9 buffer was added to the dried buffer before proceeding through using a SpeedVac concentrator. The identical fractions from DMSO
the lysis steps. control lysates were also collected and dried. The dried fractions were
re-suspended in a minimal volume of 1:1 (v/v) methanol:0.1% aqueous
HPLC method formic acid. Electrospray ionization MS (ESI-MS) analyses were carried
The frozen lysates were thawed, and 50 µl of acidified acetonitrile solu- out using a 6538 UHD model quadrupole time-of-flight mass analyser
tion (50% acetonitrile, 0.2% acetic acid) was added to the thawed lysates. equipped with an atmospheric pressure ESI source and a 1260 Infinity
For the fly samples, three separate lysates were pooled for each repli- model HPLC system (Agilent Technologies). Samples were analysed by
cate to minimize variability associated with having only three larvae loop injection with a mobile phase composed of 1:1 (v/v) methanol:0.1%
per sample. The samples were mixed by vortexing for approximately aqueous formic acid and a flow rate of 0.25 ml min–1. Mass spectra were
10 s and then centrifuged at 17,949g for 1 min. After centrifugation, recorded in the 2 GHz mode, and the high-resolution MS analyses for
50 μl of lysate was injected onto a 4.6 × 150 mm Zorbax SB-C8 column molecular formula determinations were obtained using external cali-
(5 µm particle size) and eluted over 8.65 min with the solvent and flow bration. Tandem MS/MS analyses were obtained by collision-induced
rate gradients shown in Extended Data Table 4. UV-vis absorbance was dissociation using the targeted MSn function of the acquisition soft-
measured every 2 nm between 190 and 602 nm. HPLC was performed ware. MS/MS spectra were recorded sequentially at three different
using a HP 1050 system equipped with HP Chemstation for LC 3D soft- fragmentation voltages (10, 20 and 30 V), and the resulting spectrum
ware (v.A.10.02), an autosampler, a vacuum degasser and a variable was composed of the average of those three collision energies. Agilent
wavelength DAD. The column was maintained at around 22 °C. MassHunter Data Acquisition software (v.B.05.01) was used for MS data
acquisition. Primary data analysis and export were done using Agilent
HPLC-DAD quantification MassHunter Qualitative Analysis software (v.B.06.00 SP1). Downstream
Raw absorbance data were exported to .csv files using HP Chemsta- analysis and MS plot generation (using centroid data) were done using
tion for LC 3D (v.A.10.02), and Microsoft Excel for Mac (v.16.16.27) was Microsoft Excel for Mac (v.16.16.27).
used for all chromatogram normalizations, background corrections
and AUC determinations. The dry weight of biomaterial in each of the Metabolite M2 purification and characterization by NMR
yeast, worm and fish samples was determined by calculating the AUC HB101 E. coli cells from an 8-litre saturated culture were pelleted by
at 300 nm for the major peak of endogenous content, which eluted centrifugation and resuspended in 400 ml of liquid S-medium (see
between 1 and 1.5 min, and then deriving the dry weight in mg using ref. 51 for the recipe). The bacterial suspension was transferred to a
standard curves of AUC300 versus dry weight generated using known 1-litre glass Erlenmeyer flask. Next, 6 million synchronized C. elegans L1
dry weights for each of the test organisms. The dry weight of fly larvae larvae were added to the bacterial suspension in 12 ml of M9 buffer (see
biomaterial was derived in the same way; however, AUC220 was used ref. 52 for the recipe). The synchronized L1 larvae were obtained from
instead of AUC300 as it scaled more linearly. To control for differences in a large-scale embryo preparation of gravid adults performed the day
biomass, the absorbance intensities of the raw chromatograms, in mil- before (see ref. 52 for the protocol). Worms were incubated at 20 °C on
liabsorbance units (mAU), were divided by the dry weight of the sample an orbital shaker at 200 r.p.m. for 3.5 days until they reached adulthood.
resulting in mAU per mg-of-dry-weight values. Every selectivin-A and Worms were then washed 7 times with M9 buffer, resuspended in 300 ml
ITM-treated sample was paired with an untreated DMSO control sample of M9 buffer and transferred to a 500 ml glass Erlenmeyer flask. The
prepared and processed at the same time and in the same way. To correct worms were allowed to settle to the bottom of the flask (about 20 min
for background absorbance from endogenous material, the untreated at room temperature), and 3 ml of 10 mM selectivin-A was added to the
DMSO control mAU per mg-of-dry-weight values were subtracted from worm suspension (note that the selectivin-A used for this incubation
the corresponding treated sample mAU per mg-of-dry-weight values was synthesized in-house). The flask was shaken moderately by hand
to give background-corrected chromatograms. The heat-mapped for 1 min to fully dissolve selectivin-A. The worms were then incubated
chromatograms shown in the main figures are these normalized and at 20 °C with shaking at 200 r.p.m. for 6.5 h. The worms were allowed
corrected chromatograms. The heat-mapped chromatograms were to settle on ice for 30 min. Using a glass pipette, 250 ml of incubation
generated using custom Python scripts. The AUC for the analyte peaks buffer was collected and distributed to 6 chilled 50-ml plastic conical
was computed using the normalized and corrected chromatograms, tubes (note that there were still many worms in the incubation buffer
and an average across at least three biological replicates was calcu- at this stage). The tubes were centrifuged for 2 min at 800g and then
lated. A peak was considered not detectable if an absorbance intensity placed on ice for 30 min to allow the worms to completely settle to the
maximum could not be found at the expected retention time. If a peak bottom of the tubes. Using a glass pipette, 40 ml of incubation buffer
was not detectable, it was given an AUC value of zero. AUCs for M1, M2, was then collected from each tube, being careful not to aspirate any
M3 and ITM-alk were calculated at 260 nm, whereas the AUC for M4 worms, and combined into a clean 1-litre glass bottle. The incubation
was calculated at 300 nm. The same quantification scheme was used buffer was frozen at −80 °C.
Article
Sep-Pak C8 35CC cartridges with 10 g sorbent (Waters) were used worms was summed across the two replicates. The number of viable
to de-salt and concentrate the incubation buffer. Five separate car- worms for each condition was divided by the corresponding DMSO
tridges were each activated with 50 ml 100% acetonitrile (flow rate of control value to give the relative viability for each condition. This
about 2 ml min–1). The cartridges were then washed with 50 ml ddH2O. was repeated five separate times to give five independent biological
Next, 48 ml of thawed incubation buffer was loaded onto each col- replicates.
umn. Columns were washed with 50 ml ddH2O. Columns were eluted
sequentially with 10 ml 20% acetonitrile, 10 ml 50% acetonitrile and C. elegans cyp RNAi screen
10 ml 100% acetonitrile. The eluates from each column were collected RNAi-mediated knockdown of 60 out of 76 C. elegans microsomal CYP
in 5 × 100-ml glass bottles. The 150 ml of eluate was distributed evenly enzymes was performed in 96-well culture plates using methods identi-
across 24 × 20-ml scintillation vials and stored at 4 °C overnight. The cal to those described for the emb-8 RNAi dose–response assays, but
next day, the eluates were dried using a Genevac centrifugal evaporator with the following three modifications: (1) WT worms were used instead
(about 3 h at 55 °C). The dried eluate from the 24 vials was dissolved in of the MJ69 strain; (2) synchronized L1 larvae were grown for 2.5 days at
2.5 ml of a 5% acetonitrile and 0.1% acetic acid solution (about 1.5 ml of 20 °C instead of 2 days at 25 °C; (3) a single selectivin-A concentration
dissolved eluate remained after dissolution and transfers). The com- of 25 µM was used for the screen. Each well had bacteria expressing
ponents of the eluate solution were separated using a HP 1050 HPLC a single dsRNA targeting a single cyp, and around 25 L1 larvae were
system equipped with an autosampler, vacuum degasser and variable added to each well. After the worms had fed on RNAi-expressing bac-
wavelength DAD, and the fraction containing metabolite M2 was col- teria for 2.5 days and had reached adulthood, 25 µM selectivin-A (1%
lected. For each HPLC run, 50 μl of the eluate solution was injected onto DMSO (v/v)) was added to each well. After 24 h of chemical exposure,
a 4.6 × 150 mm Zorbax SB-C8 column (5 µm particle size) and eluted the percentage of worms in the well that were moving was quantified.
over 30 min with the solvent and flow rate gradients shown in Extended Each cyp RNAi knockdown was tested in technical duplicate and an
Data Table 4. UV-vis absorbance was measured every 2 nm between 190 average per cent-moving value was calculated. Fifty control repli-
and 602 nm. The column was maintained at about 22 °C. Thirty separate cates using bacteria carrying the empty L4440 RNAi vector were also
HPLC runs were required to process all of the eluate solution. The M2 included for control purposes. Using the mean and standard deviation
metabolite eluted between 20.3 and 21 min. M2 metabolite fractions from the control replicates, Z-scores were calculated for the average
were collected and split into two pre-weighed 20 ml glass scintillation per cent-moving values for each cyp knockdown. A Z-score greater than
vials. The combined fractions were dried using a Genevac centrifu- three was considered to be significant. The raw data for the C. elegans
gal evaporator (about 4 h at 40 °C). Before Genevac drying, a small cyp RNAi knockdown screen can be found in Supplementary Table 2.
amount of the M2 fraction was taken for purity analysis by HPLC-DAD.
Purity was estimated by dividing area under the M2 curve at 250 nm Attempted oxidation of selectivin-A
by the total peak area across the run. The purity of M2 was 95.4%. The In an attempt to generate the sulfoxide metabolite of selectivin-A in the
yield of M2 was estimated to be 2 mg by subtracting the weight of the laboratory, we tried three different oxidation methods: (1) hydrogen
empty vials from the weight of the vials containing dried metabolite peroxide as oxidant: H2O2 (1 equiv.), acetone/H2O (5:1) 0 °C to room
and multiplying by 0.954. temperature; (2) oxone as oxidant: oxone (5 equiv.), H2O/methanol,
NMR data for purified M2 was obtained at 293 K on a Varian Mer- room temperature; (3) m-CPBA as oxidant: m-CPBA (2.5 equiv.), dichlo-
cury 300 MHz, Varian Mercury 400 MHz, Bruker Advance III 400 MHz, romethane, room temperature. Using hydrogen peroxide as the oxidant
Agilent DD2 500 MHz equipped with a 5 mm Xses cold probe, Agilent resulted in the recovery of starting material alone (that is, selectivin-A),
DD2 600 MHz or Agilent DD2 700 MHz. 1H spectra were referenced to and cleavage of the imidazole ring occurred when oxone was used as
the residual solvent signal (CDCl3 = 7.26 ppm, DMSO-d6 = 2.50 ppm, the oxidizing agent. When m-CPBA was used as the oxidant, a com-
CD3OD = 3.31 ppm). 13C{1H} spectra were referenced to the residual sol- plex mixture was observed and purification only resulted in further
vent signal (CDCl3 = 77.16 ppm, DMSO-d6 = 39.52 ppm, CD3OD = 49.00). decomposition.
Data for 1H-NMR are reported as follows: chemical shift (δ ppm), mul-
tiplicity (s = singlet, d = doublet, t = triplet, q = quartet, m = multiplet) Synthesis of selectivin-A, selectivin-E, selectivin-F, selectivin-H
and coupling constant (Hz), integration. Agilent VnmrJ software (v.4.2) and selectivin-I
was used for data acquisition, and MestreNova software (v.14.3.1) was For each selectivin analogue, the appropriate 2-bromo-acetophenone
used for data processing. All NMR spectra were Fourier transformed was synthesized from the corresponding commercially available aceto-
with exponential line broadening, phased and baseline corrected. phenone according to procedures in the literature65. The imidazo[2,1-b]
thiazoles were prepared according to a previously published pro-
NACET experiments cedure66, but with modifications. To a 2 dram vial, the appropriate
Saturated HB101 bacterial cultures were concentrated twofold with α-bromoketone (1 mmol, 1 equiv.), the appropriate 2-aminothiazole
either regular liquid NGM or liquid NGM containing 2.5 mM NACET. In (1.3 mmol, 1.3 equiv.) and ethanol (1.6 ml) were added, and the reaction
the wells of a 96-well culture plate, 25 synchronized L1 worms in 10 µl mixture was stirred at reflux until disappearance of the α-bromoketone
of M9 buffer were added to 40 µl of the bacterial suspensions. The was evident by thin-layer chromatography. The mixture was concen-
synchronized L1 worms were obtained from an embryo preparation trated then purified by column chromatography using the given eluent
performed the previous day (see ref. 52 for the protocol). The culture to provide the imidazo[2,1-b]thiazole.
plate was sealed with Parafilm and placed in a box with several wet
paper towels. The worms were incubated for 4 h at 20 °C, after which 6-(4-Chlorophenyl)imidazo[2,1-b]thiazole (selectivin-A). At 16 mmol
100 µM of nematicide, or DMSO alone, was added independently to scale. Purified using pentanes–ethyl acetate (13:7 to 10:10 (v/v)), fol-
NACET-containing or NACET-free wells. The plates were re-sealed with lowed by trituration with methanol. White solid (1.0 g, 28%). 1H-NMR
Parafilm, replaced in the box with wet paper towels and allowed to incu- (CDCl3, 400 MHz) δ 7.72 (d, J = 8.6 Hz, 2H), 7.67 (s, 1H), 7.38 (d, J = 4.5 Hz,
bate for an additional 24 h at 20 °C before assessing lethality. Lethality 1H), 7.34 (d, J = 8.6 Hz, 2H), 6.80 (d, J = 4.5 Hz, 1H). 13C-NMR (CDCl3,
was assessed as described above for the C. elegans L1 dose–response 100 MHz) δ 150.4, 146.7, 133.1, 132.6, 128.9, 126.5, 118.6, 112.8 and 108.2.
assays. A concentration of 2.5 mM NACET was chosen because it is
the highest concentration of NACET that can be used without sub- 6-(4-Chlorophenyl)-2-methylimidazo[2,1-b]thiazole (selectivin-E).
stantially slowing worm growth or killing the worms. Two technical At 10 mmol (2.3 g) scale. Purified using pentanes–ethyl acetate (15:5 to
replicates were performed for each condition, and the number of living 10:10 (v/v)). Pale orange solid (30%). 1H-NMR (CDCl3, 500 MHz): 7.73
(d, J = 8.5 Hz, 2H), 7.59 (s, 1H), 7.34 (d, J = 8.5 Hz, 2H), 7.13 (q, J = 1.4 Hz, garden packs. The soil was drenched with 18 ml of deionized water
1H), 2.42 (d, J = 1.4 Hz, 3H). 13C{1H}-NMR (CDCl3, 125 MHz): 149.9, 145.5, containing dissolved chemical or DMSO alone. Depending on the
132.9, 132.8, 128.9, 127.0, 126.4, 115.2, 107.9 and 14.2. batch, either 1,000 or 2,500 infective J2 larvae were then added to
the soil in 2 ml of water for a total water volume of 20 ml. All of the tests
6-(4-Fluorophenyl)-3-methylimidazo[2,1-b]thiazole (selectivin-F). done in the same batch and on the same day had the same number of
Purified using pentanes–ethyl acetate (15:5 (v/v)). Brown solid (38%, J2 larvae added to the soil so that the DMSO control and treatment
MP = 109–114 °C). 1H-NMR (CDCl3, 500 MHz): 7.83–7.78 (m, 2H), 7.57 samples had the same number of J2 larvae added. The final aqueous
(s, 1H), 7.12–7.05 (m, 2H), 6.42 (q, J = 1.3 Hz, 1H), 2.43 (d, J = 1.3 Hz, 3H). concentration of the selectivin analogues was 45 µM. This concentra-
13
C{1H}-NMR (CDCl3, 125 MHz): 162.4 (d, J = 246.2 Hz), 149.9, 146.9, 130.4, tion was chosen because the analogues identified from our primary
127.9 and 127.0 (d, J = 8.0 Hz), 115.7 (d, J = 21.6 Hz), 107.0, 105.8 and 13.5. screen are lethal to root-knot nematodes at this concentration in vitro
19 1
F{ H}-NMR (CDCl3, 375 MHz): –115.0. Infrared (neat): 3,134, 2,965, 2,926, (Fig. 1b). This concentration corresponds to around 4.5 kg of active
2,883, 1,750, 1,475, 1,375, 1,155, 1,092, 1,009, 831, 755 and 692. Mass: ingredient per ha of soil. Despite the label rates being lower than
DART+, calculated for C12H10N2FS 233.05432 [M+H]+, found 233.05424. 4.5 kg ha–1 for a subset of the commercial nematicides, we used the
same 4.5 kg of active ingredient per ha application rate for all of the
6-(4-Bromophenyl)-3-methylimidazo[2,1-b]thiazole (selectivin-H). commercial nematicides tested so that the results would be compara-
Purified using pentanes–ethyl acetate (16:4 to 15:5 (v/v)). Orange solid ble among the commercial controls and the selectivin analogues. The
(33%). The spectral data were in accordance with literature62. 1H-NMR final aqueous DMSO concentration varied from 0.05% to 0.4% (v/v)
(CDCl3, 500 MHz): 7.73–7.69 (m, 2H), 7.61 (s, 1H), 7.53–7.49 (m, 2H), depending on the stock concentration of the chemical and the final
6.42 (q, J = 1.3 Hz, 1H), 2.42 (d, J = 1.3 Hz, 3H). 13C{1H}-NMR (CDCl3, 125 concentration of the chemical tested. The highest DMSO concentra-
MHz): 150.1, 146.8, 133.4, 131.9, 127.8, 126.8, 121.2, 107.1, 106.3 and 13.5. tion of 0.4% (v/v) was used for all DMSO controls. Importantly, we
observed that 0.4% DMSO did not inhibit root infection compared
6-(4-Bromophenyl)-2-methylimidazo[2,1-b]thiazole (selectivin-I). with water control. There were four DMSO controls in each batch. The
Purified using pentanes–ethyl acetate (16:4 to 8:12 (v/v)). White solid J2 larvae were incubated in the soil and chemical for 24 h, after which
(32%, MP = 235–240 °C). 1H-NMR (CDCl3, 500 MHz): 7.69–7.64 (m, 2H), 2–3-week-old tomato seedlings were transplanted into the soil (one
7.61 (s, 1H), 7.52–7.47 (m, 2H), 7.13 (q, J = 1.4 Hz, 1H), 2.42 (d, J = 1.5 Hz, 3H). plant per compartment). Inoculated plants were grown for 8 weeks in a
13
C{1H}-NMR (CDCl3, 125 MHz): 150.0, 145.5, 133.2, 131.9, 127.0, 126.7, greenhouse as previously described56 under long-day conditions (16 h
121.0, 115.2, 108.0 and 14.2. Infrared (neat): 3,134, 2,965, 2,926, 2,883, photoperiod) with 26/18 °C day/night temperatures. After 8 weeks,
1,750, 1,475, 1,375, 1,155, 1,092, 1,009, 831, 755 and 692. Mass: DART+, the plants were destructively collected. The tops were removed and
calculated for C12H10N2SBr 292.97426 [M+H]+, found 292.97416. discarded, and roots were gently washed with water to remove adher-
Work-up and isolation of compounds were performed using stand- ing soil. Eggs were extracted by placing rinsed roots in 0.6% sodium
ard benchtop techniques. All commercial reagents were purchased hypochlorite and agitating at 300 r.p.m. for 3 min. Roots were then
from chemical suppliers (Sigma-Aldrich, Combi-Blocks, Alfa Aesar rinsed over nested 250 and 25.4 μm sieves, with eggs collected from
or Strem Chemicals) and used without further purification. Dry sol- the latter and suspended in water. Roots were dried in a 65 °C oven for
vents were obtained using standard procedures (THF was distilled over at least 72 h, after which dry roots were weighed. The number of eggs
sodium–benzophenone, dichloromethane was distilled over calcium from each plant root was counted on a dissection microscope using a
hydride). Reactions were monitored using thin-layer chromatogra- haemocytometer, and the number of eggs per milligram of root was
phy on EMD Silica Gel 60 F254 plates. Visualization was performed calculated by dividing the total egg number by the mass of the dried
under UV light (254 nm) or using potassium permanganate (KMnO4) root material. Quantifying the number of eggs per unit of root weight
or I2 stain. Flash column chromatography was performed on Siliaflash is a common method for assessing plant susceptibility to parasitic
P60 40–63 µm silica gel purchased from Silicycle. NMR characteriza- nematodes67–71. Furthermore, this normalization step controls for
tion data were obtained at 293 K on a Varian Mercury 300 MHz, Varian lower egg numbers in the untreated DMSO control samples that result
Mercury 400 MHz, Bruker Advance III 400 MHz, Agilent DD2 500 MHz from stunted root growth owing to nematode infection. The eggs per
equipped with a 5 mm Xses cold probe or Agilent DD2 600 MHz. 1H spec- milligram of root value for each treatment replicate was normalized to
tra were referenced to the residual solvent signal (CDCl3 = 7.26 ppm, the average of the four DMSO controls for a given batch. To calculate
DMSO-d6 = 2.50 ppm). 13C{1H} spectra were referenced to the residual per cent effectiveness, the normalized values were subtracted from 1
solvent signal (CDCl3 = 77.16 ppm, DMSO-d6 = 39.52 ppm). Data for and then multiplied by 100. An average per cent effectiveness value
1
H-NMR are reported as follows: chemical shift (δ ppm), multiplicity was then calculated across at least four replicates (zero eggs = 100%
(s = singlet, d = doublet, t = triplet, q = quartet, m = multiplet), cou- effectiveness; same number of eggs relative to control = 0% effective-
pling constant (Hz) and integration. NMR spectra were recorded at the ness; more eggs relative to control = less than 0% effectiveness). Impor-
University of Toronto Department of Chemistry NMR facility. Infrared tantly, although treatment with our two lead nematicides selectivin-A
spectra were recorded on a Perkin-Elmer Spectrum 100 instrument and selectivin-E increased root weight relative to the no-compound
equipped with a single-bounce diamond/ZnSe ATR accessory in the controls (as expected for effective nematicides), the root weight
solid state and are reported in wavenumber (cm–1) units. Melting point values were not among the highest measured across the 15 different
ranges were done on a Fisher-Johns melting point apparatus and are treatments (Extended Data Fig. 4). Therefore, it is unlikely that the
reported uncorrected. High resolution mass spectra were recorded at relatively high per cent effectiveness values for these compounds
the Advanced Instrumentation for Molecular Structure in the Depart- are simply due to higher root weight values. Parts-per-million soil
ment of Chemistry at the University of Toronto. concentrations were calculated as µg active ingredient per g of soil.
Kilograms per hectare values were calculated assuming a soil depth
M. incognita tomato plant root infection experiments of 15 cm and a soil density of 1,200 kg m–3.
A M. incognita population originally collected from grape (Vitis vinif-
era) in Parlier, California, was used in all experiments, and they were Forward genetic screens for selectivin-resistant mutants
maintained on tomato plants (S. lycopersicum ‘Rutgers’) as previously Forward genetic screens were carried out as previously described14.
described56. Infective M. incognita J2 larvae were collected as previously In brief, WT parental (P0) worms were mutagenized in 50 mM ethyl
described56. For the infection assays, 90 g of soil (1:1 sand:loam mix) methanesulfonate for 4 h. Synchronized L1 larvae from either the F1
was added to each compartment of several 6-compartment plastic (progeny) or F2 (grand progeny) generations were dispensed onto
Article
10 cm MYOB agar plates (see ref. 52 for how to prepare MYOB agar
medium) containing a 100% penetrant lethal dose of the nematicide. Reporting summary
Worms were plated at a density of about 20,000 L1 larvae per plate. The Further information on research design is available in the Nature Port-
plates were scanned by eye for viable worms. In total, 5 million F1 larvae folio Reporting Summary linked to this article.
(that is, 10 million independently mutagenized haploid genomes)
and 50,000 F2 larvae (that is, 100,000 independently mutagenized
haploid genomes) were screened for selectivin-A resistance. Next, Data availability
75,000 F1 larvae (that is, 150,000 independently mutagenized haploid Data are available for Figs. 1– 5 and Extended Data Figs. 1, 3 and 4. Any
genomes) and 25,000 F2 larvae (that is, 50,000 independently muta- other data that support the conclusions of this manuscript are available
genized haploid genomes) were screened for selectivin-C-resistant upon request. Source data are provided with this paper.
mutants. Therefore, a total of 10.3 million mutagenized genomes
were screened.
Code availability
Statistical information The Python and R code used to generate the dose–response heatmaps
HPLC analyses of unmodified selectivin parent and metabolites were and the HPLC chromatogram heatmaps have been published72.
performed at least three times for each strain or organism, and AUC
values for each analyte were calculated and averaged across the repli- 51. Lewis, J. A. & Fleming, J. T. in Caenorhabditis elegans: Modern Biological Analysis of an
Organism (eds Epstein, H. F. & Shakes, D. C.) 3–29 (Academic Press, 1995).
cates. Data are presented as the mean ± s.e.m. P values were calculated 52. Burns, A. R. et al. High-throughput screening of small molecules for bioactivity and target
using unpaired one-tailed Student’s t-tests assuming equal standard identification in Caenorhabditis elegans. Nat. Protoc. 1, 1906–1914 (2006).
of deviation. In Extended Data Fig. 3c, the pairwise P values are as 53. Demeler, J., Kuttler, U. & von Samson-Himmelstjerna, G. Veterinary parasitology
adaptation and evaluation of three different in vitro tests for the detection of resistance to
follows: Ce/Cb = 0.0028, Ce/Pp = 0.0011, Ce/KR = 0.0028, Ce/Mi = 0.0023, anthelmintics in gastro intestinal nematodes of cattle. Vet. Parasitol. 170, 61–70 (2010).
Ce/Sc = 0.0001, Ce/Dm = 0.0001, Ce/Dr = 0.0001, Cb/Pp = 0.4160, 54. Bartley, D. J. et al. A survey of anthelmintic resistant nematode parasites in Scottish sheep
flocks. Vet. Parasitol. 117, 61–71 (2003).
Cb/KR = 0.4680, Cb/Mi = 0.3311, Cb/Sc = 0.0008, Cb/Dm = 0.0009,
55. Jindapunnapat, K., Reetz, N. D., Macdonald, M. H., Bhagavathy, G. & Meyer, S. L. F. Activity
Cb/Dr = 0.0069, Pp/KR = 0.1971, Pp/Mi = 0.0787, Pp/Sc = 2.315 × 10–6, of vetiver extracts and essential oil against Meloidogyne incognita. J. Nematol. 50,
Pp/Dm = 2.124 × 10–6, Pp/Dr = 0.0004, KR/Mi = 0.1555, KR/Sc = 3.380 × 10–7, 147–162 (2018).
56. Wram, C. L. & Zasada, I. A. Short-term effects of sublethal doses of nematicides on
KR/Dm = 8.059 × 10–8, KR/Dr = 0.0017, Mi/Sc = 4.109 × 10–5, Mi/Dm =
Meloidogyne incognita. Phytopathology 109, 1605–1613 (2019).
3.816 × 10–5, Mi/Dr = 0.0041, Sc/Dm = 0.3169, Sc/Dr = 0.0044 and 57. Meyer, S. L. F., Chauhan, K. R. & MacDonald, M. H. Evaluation of roselle (Hibiscus
Dm/Dr = 0.0046. In Extended Data Fig. 3d, the pairwise P values sabdariffa) leaf and pomegranate (Punica granatum) fruit rind for activity against
Meloidogyne incognita. Nematropica 46, 85–96 (2016).
are as follows: Ce/Cb = 0.0004, Ce/Pp = 0.0021, Ce/KR = 0.0323,
58. Meyer, S. L. F. et al. Plantago lanceolata and Plantago rugelii extracts are toxic to
Ce/Mi = 0.1166, Ce/Sc = 0.0007, Ce/Dm = 0.0007, Ce/Dr = 0.0001, Meloidogyne incognita but not to certain microbes. J. Nematol. 38, 333–338 (2006).
Cb/Pp = 3.586 × 10 –5, Cb/KR = 1.407 × 10 –5, Cb/Mi = 4.775 × 10 –6, 59. Sherman, F. Getting started with yeast. Methods Enzymol. 350, 3–41 (2002).
60. Howe, K. L. et al. WormBase ParaSite—a comprehensive resource for helminth genomics.
Cb/Sc = 0.0022, Cb/Dm = 0.0022, Cb/Dr = 0.0006, Pp/KR = 3.505 × 10–6,
Mol. Biochem. Parasitol. 215, 2–10 (2016).
Pp/Mi = 8.507 × 10–6, Pp/Sc = 2.618 × 10–9, Pp/Dm = 2.618 × 10–9, Pp/Dr = 61. Blanc-Mathieu, R. et al. Hybridization and polyploidy enable genomic plasticity without
4.158 × 10 –11 , KR/Mi = 0.0003, KR/Sc = 4.242 × 10 –7, KR/Dm = sex in the most devastating plant-parasitic nematodes. PLoS Genet. 13, e1006777 (2017).
62. Sievers, F. et al. Fast, scalable generation of high-quality protein multiple sequence
4.242 × 10–7, KR/Dr = 1.067 × 10–8, Mi/Sc = 1.719 × 10–5, Mi/Dm = 1.719 × 10–5,
alignments using Clustal Omega. Mol. Syst. Biol. 7, 539 (2011).
Mi/Dr = 1.093 × 10–6, Sc/Dm = ND, Sc/Dr = ND, Dm/Dr = ND. For the 63. WormBase release WS283, http://www.wormbase.org (Alliance of Genome Resources,
yeast-based M. incognita CYP expression screen, assays with the accessed 16 December 2021).
64. Wram, C. L. & Zasada, I. Differential response of Meloidogyne, Pratylenchus, Globodera,
CYP-expressing strains and the EV control strain were replicated six and Xiphinema species to the nematicide fluazaindolizine. Phytopathology. 110,
times. P values were obtained from unpaired two-tailed Student’s 2003–2009 (2020).
t-test comparing the mean relative growth values between the 65. Chundawat, T. S., Kumari, P., Sharma, N. & Bhagat, S. Strategic synthesis and in vitro
antimicrobial evaluation of novel difluoromethylated 1-(1,3-diphenyl-1H-pyrazol-4-yl)-
CYP-expressing strains and the EV control. For the C. elegans cyp RNAi 3,3-difluoro-1,3-dihydro-indol-2-ones. Med. Chem. Res. 25, 2335–2348 (2016).
knockdown screen, each cyp knockdown was replicated twice, and 66. Pyl, T., Giebelmann, R. & Beyer, H. Über bicyclische heterocyclen mit gemeinsamem
no-knockdown controls (that is, mock experiments) were replicated 50 stickstoffatom, I. Zur kenntnis der imidazo[2.1-b]thiazole. Justus Liebigs Ann. Chem. 643,
145–153 (1961).
times. Using the mean per cent-moving value and s.d. from the control 67. Huang, G. et al. Engineering broad root-knot resistance in transgenic plants by RNAi
replicates, Z-scores were calculated for each cyp knockdown. A cyp silencing of a conserved and essential root-knot nematode parasitism gene. Proc. Natl
knockdown resulting in an average per cent-moving value with a Z-score Acad. Sci. USA 103, 14302–14306 (2006).
68. Mota, F. C. et al. New sources of resistance to Meloidogyne incognita race 3 in wild cotton
greater than three was considered to be significant. For the selectivin accessions and histological characterization of the defence mechanisms. Plant Pathol.
analogue series, soil-based root-infection assays were performed at 62, 1173–1183 (2013).
least four times. To determine whether chemical treatment resulted 69. Basso, M. F. et al. MiDaf16-like and MiSkn1-like gene families are reliable targets to develop
biotechnological tools for the control and management of Meloidogyne incognita. Sci
in a per cent effectiveness greater than zero, P values were calculated Rep. 10, 6991 (2020).
using two-tailed one-sample t-tests comparing the treatment mean 70. Hajihassani, A., Rutter, W. B. & Luo, X. Resistant pepper carrying N, Me1, and Me3 have
with zero. A P value < 0.01 indicates a significant difference. Per cent different effects on penetration and reproduction of four major Meloidogyne species.
J. Nematol. 51, 1–9 (2019).
effectiveness data are presented as the mean ± s.e.m. To determine 71. de Souza, J. D. A. et al. Knocking-down Meloidogyne incognita proteases by plant-
whether selectivin-E and selectivin-A mean per cent effectiveness val- delivered dsRNA has negative pleiotropic effect on nematode vigor. PLoS ONE 8, e85364
ues differ from that of tioxazafen, unpaired one-tailed Student t-tests (2013).
72. Burns, A. R. and Roy, P. J. Python and R scripts for the generation of dose–response heatmaps
were performed assuming equal s.d. P values > 0.05 indicate no sig- and heatmaps of HPLC chromatograms. Zenodo https://doi.org/10.5281/zenodo.7731172
nificant difference in effectiveness. No statistical methods were used (2023).
to predetermine sample sizes. Blinding was performed for the follow-
ing experiments: mouse studies, soil-based plant-parasitic nematode Acknowledgements Free-living nematode strains were provided by the CGC (University of
assays, A. thaliana assays, fish assays, human cell assays, C. albicans Minnesota), M.-A. Félix (Institute of Biology of the Ecole Normale Supérieure, Paris, France) and
R. Rae (Liverpool John Moores University, Liverpool, UK). M. hapla was provided by B. Mimee at
assays, in vitro M. incognita and M. chitwoodi assays, MS and NMR. All Agriculture and Agri-food Canada in Saint-Jean-sur-Richelieu, Quebec, Canada. MS analyses
other experiments were not blinded. In all experiments, test subjects were performed by staff at the Advanced Instrumentation for Molecular Structure Mass
(organisms, cells, among others) were chosen at random for experimen- Spectrometry Laboratory in the Department of Chemistry at the University of Toronto, Canada.
NMR analyses were performed by staff at the CSICOMP NMR Facility in the Department of
tation. GraphPad Prism (v.6.0) and Microsoft Excel for Mac (v.16.16.27) Chemistry at the University of Toronto, Canada. Many thanks to D. Colwell and D. Gray at the
were used for all statistical analyses. Lethbridge and Agri-Food Canada Research Centre for supplying faeces from infected calves
for C. oncophora egg collection; K. Yoshioka at the University of Toronto, Canada, for P. simiae in tomato plants in the laboratory of I.Z. A.R.B. and J.K. carried out the M. hapla assays.
WSC417; J. Goldstone from the Woods Hole Oceanographic Institution for providing the D. melanogaster experiments were carried out by C.H. and A.R.B. in the laboratories of P.J.R.
M. incognita cyp gene names; B. Ciruna for the gift of NACET; G. Brown for the culture of and H.M.K. The genetics screens for resistant mutants were performed by A.R.B. The project
S. cerevisiae; B. Derry for helpful comments on the manuscript; and N. Robbins from L. Cowen’s was conceived by A.R.B. and P.J.R. The manuscript was written by A.R.B. and P.J.R.
laboratory for comments on the work. The following grants supported this work: CIHR grants
(313296 and 173448), an NSERC i2i grant (555963-20), a David Dime Grant, and a Canada Competing interests L.E.C. is a co-founder and shareholder in Bright Angel Therapeutics,
Research Chair (Tier 1) to P.J.R.; a NSERC grant (RGPIN-2020-04168) to M.L.; a CIHR Foundation a platform company for development of new antifungal therapeutics. L.E.C. is a consultant
grant (FDN-154288) and a Canada Research Chair (Tier 1) to L.E.C.; an EvoFunPath fellowship for Boragen, a small-molecule development company focused on leveraging the specific
from the NSERC CREATE program (555337-2021) to E.P.; a CIHR project grant (303157) to I.S. chemical properties of boron chemistry for crop protection and animal health. L.E.C. is a
C.A.M.F. is supported by the Alberta Innovates Technology Fund (G2016000681). J.S.G. is Science Advisor for Kapoose Creek, a company that harnesses the therapeutic potential of
supported by a NSERC discovery grant (RGPIN-2021-02489), and by the NSERC-CREATE fungi. Mention of trade names or commercial products in this publication is solely for the
Host–Parasite Interactions Program graduate training programme at the University of Calgary. purpose of providing specific information and does not imply recommendation or endorsement
by the US Department of Agriculture. USDA is an equal opportunity provider and employer.
Author contributions Unless otherwise noted, all experiments were carried out in the laboratory The University of Toronto is pursuing patent protection related to the use of selectivions as
of P.J.R. All free-living nematode dose–response experiments were carried out by A.R.B. A.R.B. nematacides for which two national phase applications, US Patent Application 17/624,629, and
performed the HPLC-based metabolism experiments and analysed all related MS data. A.R.B. Canadian Patent Application 3,146,085, are currently under prosecution and list A.R.B., M.L.,
performed the NACET experiments. B.M.P., E.M.R. and A.R.B. performed the C. oncophora R.B. and P.J.R. as inventors. A patent application, listing A.R.B., J.K., B.C., and P.J.R. as inventors,
dose–response experiments in J.S.G.’s laboratory. S. cerevisiae work was done by A.R.B., J.K. and is under preparation for the use of yeast-based expression systems for the identification of
B.C. M. incognita bioinformatics was done by J.K. B.C. performed the yeast-based M. incognita bioactivated nematacides, insecticides, pesticides and drug leads. The patent application will
CYP expression screen, and data were analysed by A.R.B. J.S. performed the HEK293 cell dose– be soon filed listing the inventors and the University of Toronto as applicants.
response assays in I.S.’s laboratory. J.T. (in the laboratory of H.M.K.) and J.R.V. (in the laboratory
of J.J.D.) performed the zebrafish dose–response assays. S.M. and S.W.C. performed the Additional information
HepaRG cell dose–response experiments in S.A.M.’s laboratory. E.P. performed the C. albicans Supplementary information The online version contains supplementary material available at
assays in L.E.C.’s laboratory. In the laboratory of S.R.C., A.S.V. performed the Arabidopsis https://doi.org/10.1038/s41586-023-06105-5.
greening assays. A.R.B performed the P. simiae dose–response experiments. C.A.M.F. and Q.X. Correspondence and requests for materials should be addressed to Andrew R. Burns or
carried out the mouse experiments. A.R.B. performed the metabolite purification for NMR Peter J. Roy.
analyses. R.J.B. carried out the selectivin syntheses and characterization in M.L.’s laboratory, Peer review information Nature thanks Johan Desaeger, Timothy Geary, Fredd Vergara and the
and helped with metabolite NMR data analyses. A.R.B. performed the C. elegans cyp RNAi other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer
screen. M.H.M. carried out the in vitro M. incognita assays in S.L.F.M.’s laboratory. M.K. carried reports are available.
out the in vitro M. chitwoodi assays and the M. incognita soil-based reproduction assays Reprints and permissions information is available at http://www.nature.com/reprints.
Article

Extended Data Fig. 1 | The selectivin-A imidazole-thiol metabolite is not Area under the curve (AUC) for metabolites M1-M3 and ITM-alk was calculated
nematicidal. a, Mass spectrometry (MS) data for the M4 metabolite HPLC at 260 nm, and at 300 nm for M4. Data are presented as mean ± SEM. For each
fraction and untreated control fraction, as well as MS/MS data for the 418.99 analyte, unpaired one-tailed Student’s t-tests were performed comparing
mass. The proposed M4 disulfide structure is shown, along with the protonated the means of the ± IAA conditions. P-values are shown. f, Dose-response for
imidazole-thiol monomer structure resulting from fragmentation. b, A proposed C. elegans L1s treated with selectivins or commercially-sourced ITM. g, Biomass-
metabolic pathway producing the imidazole-thiol metabolite (ITM) which normalized and background-corrected HPLC chromatograms for lysates
oxidizes to form M4/ITM disulfide (ITM-DiS). Imidazothiazole ring opening is of L1 worms incubated in commercially-sourced ITM, lysed ± IAA. The top
likely CYP-dependent. c, Biomass-normalized and background-corrected chromatogram is the ITM standard injected directly onto the column, and is
HPLC chromatograms for lysates of selectivin-A-treated L1 worms lysed ± not normalized or corrected. ITM oxidizes to ITM-DiS prior to HPLC analysis.
iodoacetamide (IAA). Peaks corresponding to selectivin-A (sel-A), metabolites ITM-DiS and ITM-alk peaks are indicated. h, HPLC-DAD quantification of
M1-M4, and alkylated ITM (ITM-alk) are indicated. d, MS data for the ITM-alk M4/ITM-DiS and ITM-alk in the lysates of 100 µM selectivin-A-treated or 100 µM
HPLC fraction and untreated control fraction. e, HPLC-DAD quantification of ITM-treated L1 worms, lysed ± IAA (n = 3 biological replicates with 200,000
selectivin-A, M1-M4, and ITM-alk in the lysates of selectivin-A-treated L1 worms worms per replicate). AUC was calculated at 260 nm. Data are presented as
lysed ± IAA (n = 3 biological replicates with 200,000 worms per replicate). mean ± SEM.
Extended Data Fig. 2 | NMR spectra for HPLC-purified selectivin-A 2.2 Hz, 1H), 4.74 (dd, J = 7.6, 5.3 Hz, 1H), 4.66 (dd, J = 14.9, 8.0 Hz, 1H), 3.86 (s, 2H),
metabolite M2. a, 1H NMR spectra. The structure of M2 with all carbon and 3.81 (dd, J = 14.9, 2.2 Hz, 1H), 3.62 (t, J = 6.2 Hz, 1H), 3.23 (dd, J = 14.2, 5.3 Hz, 1H),
heteroatom assignments is given. The top number in each multiplet box 3.08 (dd, J = 14.1, 7.6 Hz, 1H), 2.54 (ddd, J = 15.9, 7.9, 6.6 Hz, 1H), 2.48 (dt, J = 15.8,
corresponds to the carbon that the proton is connected to. 1H NMR (700 MHz, 6.6 Hz, 1H), 2.16 – 2.05 (m, 2H), 1.97 (s, 3H). b, 1H-1H COSY NMR spectra. c, 1H-13C
cd3od) δ 8.02 (s, 1H), 7.82 – 7.78 (m, 2H), 7.42 – 7.37 (m, 2H), 5.98 (dd, J = 7.9, HSQC NMR spectra. d, 1H-13C HMBC NMR spectra.
Article

Extended Data Fig. 3 | Selectivin bioactivation is nematode-specific. species. AUC is area under the curve at 260 nm. Data are presented as mean ±
a and b, Biomass-normalized and background-corrected HPLC chromatograms SEM. For c and d, unpaired one-tailed Student’s t-tests were performed for all
of lysates (a) and incubation buffers (b) from C. elegans (Ce), C. briggsae (Cb), pairwise comparisons of means. The means sharing a letter are statistically
P. pacificus (Pp), Rhabditophanes sp. KR3021 (KR), M. incognita (Mi), indistinguishable (p > 0.01). The p-values for each comparison in c and d can be
D. melanogaster (Dm), and D. rerio (Dr) larvae, as well as S. cerevisiae (Sc) cells, found in the Statistical Information subsection of the Methods. For e, unpaired
incubated in 100 µM selectivin-A. Selectivin-A and metabolite peaks are one-tailed Student’s t-tests were performed relative to the C. elegans mean.
indicated. c, Total detectable production of M1 + M2 + M3 in lysate and buffer P-values are shown. For c-e, n = 4 biological replicates for Ce, Cb, Pp, and Dr, and
for each of the seven species tested. d, M2 tissue accumulation in each species n = 3 biological replicates for KR, Mi, Sc, and Dm.
tested. e, Unmodified selectivin-A tissue accumulation in each of the test
Extended Data Fig. 4 | Selectivin treatment generally increases the root concentration, parts-per-million soil concentration, and kilograms-per-hectare
weights of tomato plants challenged with nematode infection. The effects values are shown. The R-groups for each selectivin analog are indicated (see
of 11 selectivin analogs and 4 commercial nematicides on the root weights of Fig. 1a for the selectivin core scaffold and R-group positions). Data are presented
tomato plants challenged with M. incognita infection is shown. Root weight as mean ± SEM.
values are percent of untreated control. For each analog, aqueous molar
Article

Extended Data Fig. 5 | Free-living nematodes likely detoxify selectivin-E via in the corresponding untreated control fractions. Raw counts were taken from
hydroxylation and subsequent phosphoglucosidation. a, Structure of centroid plots of the raw mass spectra. Counts below 1 × 103 were considered
selectivin-E (sel-E) with exact mass. b-c, Raw HPLC chromatograms for lysates not detectable (nd). Based on the presumptive masses of the three metabolites
of C. elegans and P. pacificus L1 worms incubated in 100 µM selectivin-E or we predict that E-M1 is a hydroxylated metabolite of selectivin-E, E-M2 is an
DMSO alone (untreated). The top chromatogram is the selectivin-E standard O-linked glucoside of selectivin-E, and E-M3 is an O-linked phosphoglucoside.
injected directly onto the column. The unmodified selectivin-E parent Molecular formulas are indicated for the predicted metabolite structures, as
compound and selectivin-E metabolites E-M1, E-M2, and E-M3 are indicated. well as exact masses, P. pacificus experimental accurate masses, and the ppm
c, Mass spectrometry (MS) data for the E-M1, E-M2, and E-M3 HPLC fractions difference between the two. A ppm difference of less than 5 ppm suggests that
and corresponding untreated control fractions. Raw MS counts are shown for the molecular formulae are correct. Ce, C. elegans; Pp, P. pacificus.
the most abundant masses identified in the mass spectra that are not detectable
Extended Data Table 1 | Anthelmintic/nematicide-resistant mutants are sensitive to the selectivins

Selectivin-A and selectivin-C LC50 data for wild-type worms and nine distinct nematicide/anthelmintic-resistant mutant strains. LC50 is the concentration at which 50% of the nematodes are
dead. OP, organophosphate; LEV, levamisole; APA, aminophenylamidine; THP, tetrahydropyrimidine; AAD, aminoacetonitrile derivative; BZ, benzimidazole.
Article
Extended Data Table 2 | Genetic resistance to selectivins is difficult to achieve

The number of genomes screened and the number of resistant mutants obtained from genetic screens for mutants that resist selectivin-A, selectivin-C, and four additional commercial nemati-
cides/anthelmintics. Wact-11 is a structural analog of the commercial nematicide fluopyram. Wact-11 and fluopyram share the same nematicidal mode-of-action, i.e. inhibition of mitochondrial
complex II, and wact-11 resistant mutants are also resistant to fluopyram14.
Extended Data Table 3 | Summary of MS data for selectivin-A metabolites from lysates

Raw MS counts for the m/z values corresponding to the selectivin-A (sel-A) metabolites M1, M2, M3, and M4 in HPLC fractions taken from lysates of five nematode species and three non-nematode
species incubated in selectivin-A. Raw counts were taken from centroid plots of the raw mass spectra. Counts below 1 x 103 were considered not detectable (nd). Ce, C. elegans; Cb, C. briggsae; Pp,
P. pacificus; KR, Rhabditophanes sp. KR3021; Mi, M. incognita; Sc WT, S. cerevisiae wild-type strain; Sc+35C1, S. cerevisiae expressing Ce-CYP-35C1; Sc + 4731A3, S. cerevisiae expressing
Mi-CYP4731A3; Dm, D. melanogaster; Dr, D. rerio. The KR, Mi, Sc + 35C1, and Sc + 4731A3 M4 metabolite fractions were not processed by MS.
Article
Extended Data Table 4 | HPLC solvent and flow rate gradients

Solvent and flow rate gradients for the HPLC methods used in this study. Solvent A is 4.9:95:0.1 (ACN:H2O:acetic acid). Solvent B is 95:4.9:0.1 (ACN:H2O:acetic acid).
nature research | reporting summary
Corresponding author(s): Peter J. Roy and Andrew R. Burns
Last updated by author(s): March 19, 2023

Reporting Summary
Nature Research wishes to improve the reproducibility of the work that we publish. This form provides structure for consistency and transparency
in reporting. For further information on Nature Research policies, see our Editorial Policies and the Editorial Policy Checklist.

Statistics
For all statistical analyses, confirm that the following items are present in the figure legend, table legend, main text, or Methods section.
n/a Confirmed
The exact sample size (n) for each experimental group/condition, given as a discrete number and unit of measurement
A statement on whether measurements were taken from distinct samples or whether the same sample was measured repeatedly
The statistical test(s) used AND whether they are one- or two-sided
Only common tests should be described solely by name; describe more complex techniques in the Methods section.

A description of all covariates tested


A description of any assumptions or corrections, such as tests of normality and adjustment for multiple comparisons
A full description of the statistical parameters including central tendency (e.g. means) or other basic estimates (e.g. regression coefficient)
AND variation (e.g. standard deviation) or associated estimates of uncertainty (e.g. confidence intervals)

For null hypothesis testing, the test statistic (e.g. F, t, r) with confidence intervals, effect sizes, degrees of freedom and P value noted
Give P values as exact values whenever suitable.

For Bayesian analysis, information on the choice of priors and Markov chain Monte Carlo settings
For hierarchical and complex designs, identification of the appropriate level for tests and full reporting of outcomes
Estimates of effect sizes (e.g. Cohen's d, Pearson's r), indicating how they were calculated
Our web collection on statistics for biologists contains articles on many of the points above.

Software and code


Policy information about availability of computer code
Data collection HPLC-DAD data | HP Chemstation for LC 3D (v. A.10.02)
MS data | Agilent MassHunter Data Acquisition (v. B.05.01)
NMR data | Agilent VnmrJ (v. 4.2)
S. cerevisiae data | National Instruments LabVIEW 2013 (v. 13.0)
C. albicans data | SoftMax Pro 7
P. simiae data | BMG Labtech Omega (v. 1.20)
All other data were collected manually

Data analysis HPLC-DAD data | HP Chemstation for LC 3D (v. A.10.02), Microsoft Excel for Mac (v. 16.16.27), GraphPad Prism (v. 6.0), and a custom python
script for generating chromatogram heatmaps (https://doi.org/10.5281/zenodo.7731172) using Python (v. 3.9.13) and Anaconda Spyder (v.
5.3.3)
MS data | Agilent MassHunter Qualitative Analysis (v. B.06.00 SP1), Microsoft Excel for Mac (v. 16.16.27), and ChemDraw Professional (v.
18.2.0.37)
NMR data | MestreNova (v 14.3.1)
April 2020

All other data were analyzed using Microsoft Excel for Mac (v. 16.16.27) and/or GraphPad Prism (v. 6.0).
Heatmaps of dose-response data were generated using a custom R script (https://doi.org/10.5281/zenodo.7731172) using R (v. 4.2.2).
Chemical structures were drawn using ChemDraw Professional (v. 18.2.0.37).
For manuscripts utilizing custom algorithms or software that are central to the research but not yet described in published literature, software must be made available to editors and
reviewers. We strongly encourage code deposition in a community repository (e.g. GitHub). See the Nature Research guidelines for submitting code & software for further information.

1
Data

nature research | reporting summary


Policy information about availability of data
All manuscripts must include a data availability statement. This statement should provide the following information, where applicable:
- Accession codes, unique identifiers, or web links for publicly available datasets
- A list of figures that have associated raw data
- A description of any restrictions on data availability

Data and materials availability:


A data availability statement is included in the manuscript. Raw data are included in Supplementary Data Files 1-6 and in the Source Data files. Any other materials
can be made available per request through the corresponding authors ARB or PJR. There are no restrictions on data availability. A list of figures that have associated
raw data is provided in the data availability statement.

Field-specific reporting
Please select the one below that is the best fit for your research. If you are not sure, read the appropriate sections before making your selection.
Life sciences Behavioural & social sciences Ecological, evolutionary & environmental sciences
For a reference copy of the document with all sections, see nature.com/documents/nr-reporting-summary-flat.pdf

Life sciences study design


All studies must disclose on these points even when the disclosure is negative.
Sample size No statistical methods were used to predetermine sample size. Nearly throughout, at least three biological (independent) replicates were
performed for each experiment. The number of replicates for each experiment are indicated in the Methods section and in the figure legends.
Sample sizes were chosen as the largest values allowing for error calculations and significance testing while also maintaining the feasibility of
data acquisition and analysis. Overall, variability in the data was low and we were interested in large effect sizes.

Data exclusions No data were excluded from the trials.

Replication Statistical analyses were performed as described in the manuscript. At least three independent replicates were performed for nearly all
experiments and variability was relatively low between replicates. All measurements were reproducible. Covariates such as temperature,
humidity, light exposure, age, developmental stage, media/buffer composition, etc. were held constant to minimize variability.

Randomization In all experiments, test subjects (organisms, cells, etc.) were chosen at random for experimentation and there was no attempt at cherry
picking or biasing the samples.

Blinding Blinding was performed for the following: mouse studies, soil-based plant-parasitic nematode assays, arabidopsis assays, fish assays, human
cell assays, C. albicans assays, in vitro M. incognita and M. chitwoodi assays, mass spectrometry, and NMR. All other experiments were not
blinded because only one person was available for experimentation and the experiments are quantitative in nature thereby reducing the risk
of subjective bias.

Reporting for specific materials, systems and methods


We require information from authors about some types of materials, experimental systems and methods used in many studies. Here, indicate whether each material,
system or method listed is relevant to your study. If you are not sure if a list item applies to your research, read the appropriate section before selecting a response.

Materials & experimental systems Methods


n/a Involved in the study n/a Involved in the study
Antibodies ChIP-seq
Eukaryotic cell lines Flow cytometry
Palaeontology and archaeology MRI-based neuroimaging
Animals and other organisms
Human research participants
April 2020

Clinical data
Dual use research of concern

2
Eukaryotic cell lines

nature research | reporting summary


Policy information about cell lines
Cell line source(s) HEK293 cells were purchased from ThermoFisher (Cat# R71007). HepaRG cells were purchased from ThermoFisher (Gibco
Cryopreserved HPRGC10).

Authentication HEK293 cells were authenticated by STR analysis at the Toronto Hospital for Sick Children authentication facility. HepaRG
cells were used directly from ThermoFisher, and were authenticated by the commercial supplier via SNP genotyping.

Mycoplasma contamination HEK293 cells are tested regularly for mycoplasma contamination. Tests performed a short time before experimentation
confirmed that the cell line is negative for mycoplasma contamination. Mycoplasma contamination of the HepaRG cell line
was confirmed negative by the commercial supplier (ThermoFisher), and the cells were used directly from the supplier.

Commonly misidentified lines Neither the HEK293 nor the HepaRG cell line is on the list of commonly misidentified cell lines.
(See ICLAC register)

Animals and other organisms


Policy information about studies involving animals; ARRIVE guidelines recommended for reporting animal research
Laboratory animals For the mouse studies, female C57Bl/6 mice aged 6-8 weeks (bred and maintained at the animal care facility, Department of
Biological Sciences, University of Calgary, Canada) were used. For the D. rerio (zebrafish) assays, both male and female AB wild-type
fish were used at 3 dpf.

Wild animals The study did not involve wild animals.

Field-collected samples Cooperia oncophora eggs were collected from the faeces of infected calves in accordance with the Canadian Council of Animal Care
regulations; Licenses #AC13-0157 (University of Calgary) and #1730 (Agriculture AgriFood Canada Research Station, Lethbridge).

Ethics oversight For the mouse studies, all experiments were approved by the University of Calgary’s Life and Environmental Sciences Animal Care
Committee (protocol AC17-0082). All protocols for animal use and euthanasia were in accordance with the Canadian Council for
Animal Care (Canada). For the fish studies, ethics protocol 100005273 was approved by the Animal Care Committee at The Hospital
for Sick Children, Canada.
Note that full information on the approval of the study protocol must also be provided in the manuscript.

April 2020

You might also like