ArticlePDF Available

A common muscarinic pathway for diapause recovery the distantly related nematode species Caenorhabditis elegans and Ancylostoma caninum

Authors:

Abstract and Figures

Converging TGF-beta and insulin-like neuroendocrine signaling pathways regulate whether Caenorhabditis elegans develops reproductively or arrests at the dauer larval stage. We examined whether neurotransmitters act in the dauer entry or recovery pathways. Muscarinic agonists promote recovery from dauer arrest induced by pheromone as well as by mutations in the TGF-beta pathway. Dauer recovery in these animals is inhibited by the muscarinic antagonist atropine. Muscarinic agonists do not induce dauer recovery of either daf-2 or age-1 mutant animals, which have defects in the insulin-like signaling pathway. These data suggest that a metabotropic acetylcholine signaling pathway activates an insulin-like signal during C. elegans dauer recovery. Analogous and perhaps homologous cholinergic regulation of mammalian insulin release by the autonomic nervous system has been noted. In the parasitic nematode Ancylostoma caninum, the dauer larval stage is the infective stage, and recovery to the reproductive stage normally is induced by host factors. Muscarinic agonists also induce and atropine potently inhibits in vitro recovery of A. caninum dauer arrest. We suggest that host or parasite insulin-like signals may regulate recovery of A. caninum and could be potential targets for antihelminthic drugs.
The effect of muscarinic agonists and an antagonist on dauer recovery in C. elegans and A. caninum. (A) Oxotremorine, a synthetic muscarinic agonist, promotes dauer recovery in both C. elegans and A. caninum. daf2(e1370) fails to recover at all drug concentrations. The scale for the drug concentration for A. caninum is 10. All points are the average of two experiments, where at least two plates were scored with the exception of 100 mM daf-7, N2, daf-2, where only one experiment was done. (B) Atropine can specifically inhibit the muscarinic agonist-induced response. In C. elegans, at 1 mM oxotremorine, as the concentration of atropine, a muscarinic antagonist, is increased, dauer recovery is completely inhibited. In A. caninum L3 incubated with serum and GSM plus atropine (5 mM), dauer recovery was inhibited by 99.5% (from 86.7% to 0.5%). Similarly, in A. caninum larvae, 0.5 mM arecoline and increasing amounts of atropine cause dauer recovery to be completely inhibited. Concentrations of 1-5 mM of a drug are used routinely in drug assays in C. elegans (13, 14, 24). The unusually high doses may be due to a cuticle permeability barrier. Drugs in the following classes were tested on daf-7(e1372) and daf-2(e1370) mutant strains for dauer recovery and on wild type and the daf-22(m130) mutant for dauer induction and had no reproducible effect on dauer recovery or formation. (a) Noradrenalineadrenaline: cocaine*, imipramine*, reserpine*, tetrabenazine*, clonidine, isoproteronol, epinephrine, propanolol, and metopolol; (b) serotonin: 5-hydroxytryptamine, p-chlorophenylalanine, and spiperone; (c) opioids: hydromorphone, meperidine, nalorphine, PCP*, and methaqualone; (d) dopamine: dopamine and metoclopramide; (e)-aminobutyric acid: muscimol, diazepam, pentobarbitol, bicuculline, and picrotoxin; (f) glutamate: NMDA, kainate,-amino
… 
No caption available
… 
Content may be subject to copyright.
A common muscarinic pathway for diapause recovery
in the distantly related nematode species
Caenorhabditis elegans
and
Ancylostoma caninum
Heidi A. Tissenbaum*
, John Hawdon
, Melissa Perregaux
, Peter Hotez
, Leonard Guarente*, and Gary Ruvkun
†§
*Department of Biology, Massachusetts Institute of Technology, Building 68-289, 77 Massachusetts Avenue, Cambridge, MA 02139; Medical Helminthology
Laboratory, Yale University, 501 LEPH, 60 College Street, New Haven, CT 06520; and Department of Molecular Biology, Massachusetts General Hospital and
Department of Genetics, Harvard Medical School, 50 Blossom Street, Boston, MA, 02144
Edited by William B. Wood, University of Colorado, Boulder, CO, and approved November 9, 1999 (received for review October 29, 1997)
Converging TGF-
b
and insulin-like neuroendocrine signaling path-
ways regulate whether Caenorhabditis elegans develops repro-
ductively or arrests at the dauer larval stage. We examined
whether neurotransmitters act in the dauer entry or recovery
pathways. Muscarinic agonists promote recovery from dauer arrest
induced by pheromone as well as by mutations in the TGF-
b
pathway. Dauer recovery in these animals is inhibited by the
muscarinic antagonist atropine. Muscarinic agonists do not induce
dauer recovery of either daf-2 or age-1 mutant animals, which
have defects in the insulin-like signaling pathway. These data
suggest that a metabotropic acetylcholine signaling pathway ac-
tivates an insulin-like signal during C. elegans dauer recovery.
Analogous and perhaps homologous cholinergic regulation of
mammalian insulin release by the autonomic nervous system has
been noted. In the parasitic nematode Ancylostoma caninum, the
dauer larval stage is the infective stage, and recovery to the
reproductive stage normally is induced by host factors. Muscarinic
agonists also induce and atropine potently inhibits in vitro recov-
ery of A. caninum dauer arrest. We suggest that host or parasite
insulin-like signals may regulate recovery of A. caninum and could
be potential targets for antihelminthic drugs.
Animals have developed many adaptations that allow them to
survive sporadic or seasonal declines in growth conditions.
Diapause, an alternative developmental state of low metabolic
activity, is an example of such an adaptation that is found in a
wide range of species (1). In the nematode, Caenorhabditis
elegans, the dauer diapause stage is induced by unfavorable
environmental conditions (2). Under normal growth conditions,
C. elegans develops through four larval stages (L1–L4) to the
reproductive adult stage. However, in response to a secreted
pheromone, low food levels, and high temperature (i.e., unfa-
vorable growth conditions) during the L1 lar val stage, animals
enter an alternative developmental pathway and arrest at the
dauer larval L3 stage. The C. elegans dauer larva has morpho-
logical, physiological, and anatomical changes that differ from a
reproductive L3 stage animal including: altered neuroanatomy,
radial shrinkage of a thick cuticle, sealing of mouth and anal
openings, suspension of feeding, and changes in metabolism (2).
Animals can remain arrested as dauer larvae for many months.
When pheromone levels and temperature decline, and food
levels increase, the animals molt and reenter the life cycle as an
L4 larva that is indistinguishable from animals that have not
arrested at the dauer stage (2). In parasitic nematodes, such as
the hookworm, Ancylostoma caninum, diapause arrest is an
obligatory part of the life history. The dauer stage in parasitic
nematodes is the infective stage that is morphologically, behav-
iorally, and functionally analogous to the C. elegans dauer (3–5).
Recovery to the reproductive stages in the parasitic nematodes
is induced upon host infection by unknown host factors.
Mutants that affect C. elegans dauer formation (daf) fall into
two categories: dauer constitutive and dauer defective. Dauer
constitutive mutations cause animals to enter the dauer stage
even under favorable growth conditions (low pheromone, high
food, and low temperature). Conversely, mutants that are dauer
defective do not arrest at the dauer stage even under conditions
of high pheromone (2). The mutations affecting dauer arrest
have been ordered into a genetic epistasis pathway (2). The
pathway represents the steps in a neuroendocrine signaling
system from sensory detection of food, pheromone, and tem-
perature to overall remodeling of the animal to form a full dauer.
The initiation and recovery of dauer arrest is regulated by
exposed sensory neurons. First, several mutants that do not
arrest as dauer larvae in response to the dauer pheromone have
structural abnormalities in the sensory amphid sensillum (2).
Second, killing particular sensory neurons with a laser mi-
crobeam induces dauer arrest, suggesting these neurons nor-
mally signal to induce reproductive development (2).
Molecular genetic analysis has identified a TGF-
b
pathway (2,
6) and an insulin-like signaling pathway (7) thought to couple the
sensory neural inputs to other secretory cell or to the target
tissues that are remodeled and metabolically shifted in the dauer
larvae (2, 7). DAF-7, the TGF-
b
ligand, is expressed in a pair of
exposed sensory neurons in reproductively growing animals but
not expressed in animals under conditions of high pheromone
(2). DAF-4, one of the TGF-
b
receptors, and DAF-3, a Smad
protein implicated in coupling TGF-
b
signals to the nucleus, are
expressed broadly in secretory as well as target tissues (6).
Predicted insulin-like ligands of the DAF-2 insulin receptor have
been identified in the genome sequence but not yet shown to
engage DAF-2 or regulate dauer arrest (8).
Temperature also modulates dauer arrest because even null
mutations in the TGF-
b
pathway genes are temperature-
sensitive and most daf-2 alleles are temperature-sensitive (2, 9,
10). The temperature input to this neuroendocrine pathway is
mediated at least partially by the thermoregulatory AIY and
AIZ interneurons (11, 12), but how the activity of these neurons
is coupled to neuroendocrine outputs is unknown.
We reasoned that the neural pathways from the dauer regu-
latory sensory neurons and interneurons to neurosecretory cells
that signal target tissues are likely to utilize known neurotrans-
mitters. Glutamate, acetylcholine,
g
-aminobutyric acid, seroto-
nin, FMRFamide, and dopamine have been implicated in par-
ticular C. elegans behaviors (13, 14), and drugs that interact with
the mammalian receptors of these neurotransmitters affect
particular C. elegans behaviors (13, 14). Moreover, the C. elegans
genome sequence has identified members of the receptor su-
perfamilies for most of these neurotransmitters (15).
This paper was submitted directly (Track II) to the PNAS office.
Abbreviation: GSM, S-methyl-glutathione.
§To whom reprint requests should be addressed. E-mail: Ruvkun@molbiol.mgh.harvard.
edu.
The publication costs of this article were defrayed in part by page charge payment. This
article must therefore be hereby marked advertisement in accordance with 18 U.S.C.
§1734 solely to indicate this fact.
460–465
u
PNAS
u
January 4, 2000
u
vol. 97
u
no. 1
To identify neurotransmitter inputs to the dauer neuroendo-
crine pathways, we tested a variety of neurotransmitter agonists
and antagonists for induction of dauer arrest or recover y. We
find that muscarinic agonists specifically promote dauer recov-
ery in pheromone-induced dauer larvae as well as particular
classes of dauer constitutive mutants. The muscarinic agonists do
not induce recovery of either daf-2-orage-1-induced dauer
larvae, which have defective insulin-like signaling. We show that
this muscarinic pathway also regulates A. caninum recovery from
dauer arrest. In mammals, muscarinic agonists promote insulin
release both in vivo and in vitro (17, 18). We suggest that
insulin-like secretory cells in the nematodes are regulated by
cholinergic inputs in a metabolic control pathway homologous to
mammalian autonomic input to pancreatic beta cell activity.
Materials and Methods
Strains and Growth Conditions. Animals were grown on standard
NG agar plates. In this study, the mutations in C. elegans used
were: LGI, daf-8(e1393); LGII, daf-22(m130),unc-4(e120),sqt-
1(sc13),age-1(m333 and mg44); LGIII, daf-7(e1372),daf-
2(e1370, e1391), daf-4(m63); LGIV, daf-1(m40),daf-10(e1387);
LGX, daf-12(m20).age-1 animals were maintained as marked
heterozygous strains [sqt-1(sc13) age-1(mg44)ymnC1 or unc-
4(e120) age-1(m333)ymnC1]. A. caninum were maintained as
described previously (18).
Dauer Recovery Assay. Minimal media plates were used for the
drug assays: 3.0 g NaCl, 20 g agarose (Sigma Type II A6877),
0.025 M KPO
4
(pH 6.0), 1 mM CaCl
2
, 1 mM MgSO
4
, and 5
m
gyml
cholesterol were added. In some assays, Escherichia coli (DH5})
bacteria arrested with streptomycin was added to each plate. All
of the drugs used were from Sigma. Dauer-stage animals were
transferred to drug plates at 25° without the addition of food. For
experiments presented in Figs. 1 and 2 and Table 1, about 10,000
L1s were placed in 10 ml of S medium (19) containing 1–2 ml of
a 0.4% (wtyvol) solution of E. coli DH5}bacteria in M9 solution
(19) arrested with streptomycin in a 25-ml flask on a rotating,
heated water bath at 25°C. For wild-type dauer lar vae, 600
m
lof
the 0.4% (wtyvol) bacterial solution and 20–50
m
l of pheromone
also were added to the flask as described in ref. 20. After 72 hr,
the cultures were centrifuged and the supernatant was removed.
Animals were resuspended in a preheated 25° solution of 1%
SDS and placed on a rocker at 25° for 30 min, and the SDS was
removed. Animals were washed with either M9 or S medium (19)
four to six times. After a final spin, 100–200 dauer larvae were
placed onto the drug plates without food and scored 24 and 48
hr later for dauer larvae and non-dauer adults. Because muta-
tions in age-1 are maternally rescued, age-1 dauer larvae were
isolated either by plating synchronized L1s or picking maternally
rescued unc-4 age-1 or sqt-1 age-1 non-dauer animals to plates at
25°C and, after 3 days, picking dauer larvae to the drug and
control plates. Consequently, the plates with age-1 had some
bacteria on them.
Drug Assay in
A. caninum
.Hookworm-infective L3 animals were
collected from 1- to 4-week-old coproculture by the Baermann
technique and decontaminated with 1% HCl in BU buffer (50
mM Na
2
PO
4
y22 mM KH
2
PO
4
y70 mM NaCl, pH 6.8) (5, 21) for
30 min at 22°C. Approximately 250 L3 animals were incubated
in each individual wells of a 96-well plate containing 0.1 ml of
RPMI 1640 tissue culture medium, supplemented with 0.25 mM
Hepes, pH 7.2y100 units/ml penicilliny100
m
g/ml streptomyciny
100
m
g/ml gentamyciny2.5
m
g/ml amphotericin B. The L3 ani-
mals were activated to resume development and feeding by
including 10% (volyvol) canine serum and 25 mM S-methyl-
glutathione (GSM; ref. 22) in the medium. Nonactivated L3
animals were incubated in RPMI 1640 alone. The agonists were
tested for activation by incubation with the L3 animals in the
absence of the normal stimulus (i.e., serum 1GSM), whereas
atropine was tested in the presence of either the normal stimulus
or the agonists. Animals were incubated at 37°C in 5% CO
2
for
24 hr. The percentage of feeding was determined by incubating
the animals with 2.5 mgyml FITC-BSA for 2–3 hr and counting
the number of L3 animals that ingested the labeled BSA by
microscopic examination under epif luorescent illumination (23).
Each treatment was done in triplicate, and each experiment was
repeated at least once.
Dauer Arrest Assay. Animals were grown at 15° and placed in a
bleach solution to isolate eggs. One hundred to 200 eggs or
synchronized L1 larvae then were added to plates containing a
Fig. 1. The effect of muscarinic agonists and an antagonist on dauer recovery
in C. elegans and A. caninum.(A) Oxotremorine, a synthetic muscarinic
agonist, promotes dauer recovery in both C. elegans and A. caninum. daf-
2(e1370) fails to recover at all drug concentrations. The scale for the drug
concentration for A. caninum is 103. All points are the average of two
experiments, where at least two plates were scored with the exception of 100
mM daf-7,N2,daf-2, where only one experiment was done. (B) Atropine can
specifically inhibit the muscarinic agonist-induced response. In C. elegans,at
1 mM oxotremorine, as the concentration of atropine, a muscarinic antago-
nist, is increased, dauer recovery is completely inhibited. In A. caninum L3
incubated with serum and GSM plus atropine (5 mM), dauer recovery was
inhibited by 99.5% (from 86.7% to 0.5%). Similarly, in A. caninum larvae, 0.5
mM arecoline and increasing amounts of atropine cause dauer recovery to be
completely inhibited. Concentrations of 1–5 mM of a drug are used routinely
in drug assays in C. elegans (13, 14, 24). The unusually high doses may be due
to a cuticle permeability barrier. Drugs in the following classes were tested on
daf-7(e1372) and daf-2(e1370) mutant strains for dauer recovery and on wild
type and the daf-22(m130) mutant for dauer induction and had no reproduc-
ible effect on dauer recovery or formation. (a) Noradrenalineyadrenaline:
cocaine*, imipramine*, reserpine*, tetrabenazine*, clonidine, isoproteronol,
epinephrine, propanolol, and metopolol; (b) serotonin: 5-hydroxytryptamine,
p-chlorophenylalanine, and spiperone; (c) opioids: hydromorphone, meperi-
dine, nalorphine, PCP*, and methaqualone; (d) dopamine: dopamine and
metoclopramide; (e)
g
-aminobutyric acid: muscimol, diazepam, pentobarbi-
tol, bicuculline, and picrotoxin; (f) glutamate: NMDA, kainate,
a
-amino-
adipate, glutamate diethyl ester, and quisqualate. *, Affects multiple signal-
ing pathways.
Tissenbaum et al. PNAS
u
January 4, 2000
u
vol. 97
u
no. 1
u
461
NEUROBIOLOGY
drug and food at 20°C. When the non-dauer larvae had reached
the gravid adult hermaphrodite stage and were beginning to lay
eggs, each plate was examined visually for the presence of dauer
larvae and non-dauer larvae. After this, animals were rinsed off
the plate into a plastic dish containing 1% SDS (dauer larvae are
the only larval stage resistant to this treatment). After 30 min,
dishes were examined for the presence of dauer larvae and
non-dauer larvae.
Results
Cholinergic Input to Dauer Recovery. We tested drugs that affect the
following mammalian neuronal pathways for effects on C.
elegans dauer induction and dauer recovery: adrenergicy
noradrenergic, serotonergic, cholinergic, glutaminergic, dopa-
minergic, GABAergic, and opioid. Most drugs tested did not
affect induction or recover y from dauer arrest (Fig. 1). However,
multiple, unrelated muscarinic agonists promote dauer recovery.
All four muscarinic agonists tested, carbachol, oxotremorine,
pilocarpine, and arecoline, promote recovery of dauer larvae
induced by mutation as well as by pheromone although ox-
otremorine was the most potent (Fig. 1 and Table 1). Muscarinic
agonists induce recovery of dauer larvae induced by defective
TGF-
b
signaling in the daf-7(e1372) mutant, with a defect in the
TGF-
b
ligand, as well as daf-8 and daf-14 mutants, which encode
members of the Smad family of signal transduction transcription
factors that act downstream of the daf-7 ligand (A. Estevez, M.
Sundermeyer, M. Estevez, K. V. King, and D. L. Riddle, personal
communication; T. Inoue, and J. H. Thomas, personal commu-
nication) (Table 1). The muscarinic agonists do not induce
recovery of daf-2 mutants, which have defects in the C. elegans
homologue of the mammalian insulin receptor gene, or in age-1
mutants, which have a defect in a PI-3-kinase that acts down-
stream of the daf-2 receptor (refs. 2 and 9; Fig. 1 and Table 1).
Thus, the muscarinic recovery pathway depends on insulin-like
signaling.
The infective ‘‘dauer’’ L3 of the hookworm A. caninum can be
stimulated to resume feeding and development in vitro by
incubation with canine serum and GSM, but not by tissue culture
medium alone (18). When A. caninum L3 were incubated with
either oxotremorine or arecoline without canine serum or GSM
in the tissue culture medium, 60–80% of the animals recovered,
as indicated by the resumption of feeding (Fig. 1A). Therefore,
muscarinic agonists mimicked the recovery induced by serum
and GSM.
Fig. 1Ashows the oxotremorine dose-response curve for
wild-type pheromone-induced dauer lar vae, daf-7(e1372), daf-
2(e1370), and A. caninum dauer larvae. Oxotremorine and
pilocarpine induce maximum recover y of daf-7(e1372) dauer
larvae at 5 mM concentration, whereas pheromone-induced
dauer larvae reach maximum recovery at 1 mM (Fig. 1Aand
data not shown). A. caninum L3 dauer larvae also reach maxi-
mum recovery at 5 mM oxotremorine (Fig. 1A), but fail to
recover when incubated with pilocarpine (data not shown).
Pilocarpine had the least effect on all the strains tested (Table
1). Additionally, it had no effect on A. caninum (data not shown).
The maximal response for arecoline is 10-fold lower than for the
other agonists in both C. elegans and A. caninum (Table 1 and
data not shown).
Atropine Specifically Inhibits Dauer Recovery. To determine the
specificity of the muscarinic response, we added both oxotremo-
rine and atropine, a muscarinic antagonist, varying the concen-
tration of antagonist (Fig. 1B). In 1 mM oxotremorine, 40% of
the daf-7(e1372) dauer larvae recover at 25°C, the nonpermissive
temperature for daf-7(e1372ts). However, in combination with 1
mM atropine, 1 mM oxotremorine induced only 5% recovery; at
5 mM atropine, the 1 mM oxotremorine response is completely
abolished. For wild-type pheromone-induced dauer lar vae, the
results are almost identical (Fig. 1B). This suggests that the
drug-induced recovery is a specific muscarinic response, because
in mammals, atropine affects muscarinic and not nicotinic
receptors (25, 26).
Atropine can partially inhibit C. elegans dauer recovery in-
duced by food signals. When bacteria are added to pheromone-
induced dauer larvae at 25°C, 91% of the animals recover,
compared with 9% recovery without food (Fig. 2A). Atropine
partially inhibits the food-induced recovery of dauer larvae,
from 91% without drug to 75% with drug. However, pheromone
reduced this response much more severely (from 91% to 20%).
Thus, food induces more than the muscarinic pathway to trigger
dauer recovery.
In A. caninum, recovery induced by serum and GSM was
inhibited nearly completely by atropine (5 mM) (Fig. 2 A; from
86.7% to 0.5%). Moreover, A. caninum L3 incubated with 0.5
mM arecoline and 1.0 mM atropine failed to recover. These data
indicate that the muscarinic pathway is a major recovery signal
from arrest in hookworm.
Temperature downshifts in the presence of food induce dauer
recovery in animals bearing mutations in the TGF-
b
- or insulin-
like-signaling pathways (2, 6, 9). Temperature downshift in the
absence of food partially induces dauer recovery in daf-2 mutants
and does not induce dauer recovery in daf-7 mutants (Fig. 2B).
Similarly, bacterial food at 25°C does not allow reproductive
development of either daf-2 or daf-7 mutants (ref. 2; data not
shown). However, temperature downshift in the presence of food
induces more than 40% recovery of both mutants (Fig. 2B). This
temperature shift plus food recovery in both daf-7 and daf-2
mutants is potently inhibited by atropine (Fig. 2B). Similar
inhibition of dauer recovery was observed with another daf-2
allele, daf-2(e1391) (Fig. 2B).
Table 1. Induction of dauer recovery by muscarinic agonists
Drug daf-7(e1372) daf-8(e1377) daf-14(m77) daf-2(e1370) age-1(m333) age-1(mg44)
No drugyno food 1.3 61.3 4.5 60.1 0 0 0 0
Pilocarpine 8.8 66.0 17.7 60.4 2.5 60.1 0 ND ND
Arecoline 34.3 624.4 34.0 60.7 0.2 6,0.1 0 0 0
Oxotremorine 41.2 624.5 36.2 60.3 14.3 6,0.1 0 0 0
Oxotremorine atropine 5.2 62.7 4.8 60.01 ,0.1 6,0.1 0 ND ND
Experiments were performed as in Materials and Methods. Animals were scored 24 –48 h after being placed as dauer larvae on plates containing drugs. Values
are mean 6SE. Numbers shown indicate the percent dauer recovery from two trials, where each plate was at least in duplicate for daf-7(e1372), daf-2(e1370),
and age-1(m333). The remaining strains show values from one experiment in which each drug was tested on at least two plates, although similar trends have
been observed previously. For each drug tested the number of animals are in the following order: no drug no food; 1 mM pilocarpine; 1 mM arecoline; 1 mM
oxotremorine; and 1 mM oxotremorine with 1 mM atropine. Number of animals tested are: daf-7(e1372), 2345, 692, 756, 1363, 1227; daf-8(e1377), 1582, 354,
652, 753, 925; daf-14(m77), 1300, 676, 698, 902, 1101; daf-2(e1370), 872, 222, 258, 207, 460; age-1(m333), 113, not done (ND), 74, 74, ND; age-1(mg44), 71, ND,
57, 52, ND.
462
u
www.pnas.org Tissenbaum et al.
Neurotransmitter Regulation of Dauer Arrest. We examined
whether exogenous application of neurotransmitters could
mimic the dauer pheromone to induce dauer arrest. Drugs that
affect mammalian neuronal pathways including adrenergicy
noradrenergic, serotonergic, cholinergic, glutaminergic, dopa-
minergic, GABAergic, and opioid were examined for effects on
C. elegans dauer induction (Table 1). We tested these drugs for
induction of dauer arrest in wild-type and daf-22 mutants, a
mutant that does not secrete pheromone, but arrests at the dauer
stage when exposed to exogenous pheromone (2). None of the
drugs tested induced dauer arrest under favorable growth con-
ditions. The drugs were active because several of the drugs
caused either paralysis, death, or egg-laying defects.
Discussion
Arrest at the dauer stage is a nematode survival strategy that is
a specific example of the phyletically general diapause arrest
(1–3). In C. elegans, dauer arrest occurs under harsh environ-
mental conditions whereas in the hookworm, A. caninum,a
parasitic nematode, diapause is a nonconditional stage in the life
cycle (3, 4). Dauer recovery is regulated by levels of pheromone,
food, and temperature in C. elegans, whereas in A caninum,
unknown host factors induce dauer recovery upon infection
(3–5).
We have shown that muscarinic agonists cause dauer recovery
in both C. elegans and A. caninum and that this recovery is
inhibited specifically by the muscarinic antagonist atropine (Fig.
1). The endogenous neurotransmitter at muscarinic receptors is
acetylcholine, which, in vertebrates, functions at cholinergic
synapses in both the peripheral and central nervous system (26).
Acetylcholine has a wide variety of functions in vertebrate
signaling including sympathetic and parasympathetic ganglion
cells as well as the adrenal medulla, synapses within the central
nervous system, and motor end plates on skeletal muscle inner-
vated by somatic motoneurons (26). Muscarinic receptors are
found in muscle, the autonomic ganglia, the central nervous
system, and secretory glands. These receptors couple to G
proteins and signal on longer time scales than nicotinic recep-
tors. Both muscarinic and nicotinic receptors also have been
found in invertebrates such as Drosophila and C. elegans (13, 14,
26–28).
The nicotinic receptor has been the primary focus of the
studies on cholinergic signaling in the worm. The drug levami-
sole, a nicotinic agonist, is toxic to C. elegans, causing muscle
hypercontraction (13). Mutants that are resistant to this drug
have revealed components of a nicotinic-signaling cascade (13).
Levamisole has no effect on dauer recovery (data not shown),
suggesting that the nicotinic receptor pathway does not regulate
dauer arrest.
Fewer studies have been done on muscarinic signaling in C.
elegans. Binding studies on crude homogenates of C. elegans have
shown that they contain muscarinic receptors that have the
potential to bind to the muscarinic ligands [
3
H]QNB (29) and
[
3
H]N-methylscopalamine (30) with high affinity (13). These
receptors were found in both C. elegans adults and L1 and L2
larvae (29). Several muscarinic receptor homologues have been
identified in the C. elegans genome sequence database (ref. 28;
H.A.T., G. Sandoval, and G.R., unpublished observation).
Both arecoline and pilocarpine are naturally occurring drugs
from the betel nut seed and the Pilocarpus leaf, respectively,
whereas oxotremorine and carbachol are synthetic drugs (26).
Arecoline, pilocarpine, and oxotremorine have the same sites of
action on mammalian metabotropic cholinergic receptors, but
arecoline also acts on nicotinic receptors (26). Atropine, a
natural product, specifically inhibits mammalian muscarinic
responses (26). Because all of the drug-induced dauer recovery
was inhibited by atropine, and this drug does not inhibit nicotinic
signaling in C. elegans, we conclude that dauer recovery is
mediated by muscarinic signaling.
Muscarinic agonists potently induced recovery of dauer larvae
induced by pheromone or mutations in the daf-7 TGF-
b
group
of genes, but did not induce recovery of the daf-2 and age-1
Fig. 2. Atropine specifically inhibits dauer recovery in C. elegans and A.
caninum.(A) Wild-type pheromone-induced dauer larvae placed on plates
containing bacterial food, no bacteria and no pheromone, bacteria and 1 mM
atropine, and pheromone at 25°C and scored 2442 hr later for dauer larvae
and reproductive L4yadults. Experiments were performed at least twice. With
no food or pheromone, 91% of the animals remain arrested (n51,141). Dauer
larvae placed on plates with food recovered efficiently, with less than 10%
remaining arrested at the dauer stage (n52,596). The addition of 1 mM
atropine in the presence of food partially inhibited dauer recovery: 25%
remained arrested at the dauer stage (n51,311). Eighty-nine percent of the
animals maintained on plates with pheromone but no food (n51,027)
remained arrested at the dauer stage. The pheromone preparation contained
bacterial contaminants that may have been used as a food source. In A.
caninum incubated with 10% serum and 25 mM GSM, 9% of the infective
larvae remained as dauer larvae and did not resume feeding. The addition of
atropine (0.5 mM) to the serum and GSM completely inhibited recovery of A.
caninum L3, and no worms resumed feeding (data not shown). (B)daf-
7(e1372) (horizontally striped bars) and daf-2(e1370) (diagonally striped bars)
dauer larvae from 25°C liquid cultures were placed onto plates at 15°C.
Animals were scored for the presence of dauer larvae and reproductive adults
after 2 days. Temperature downshift induced dauer recovery only very slightly
in daf-2(1370) animals (4%, n5320) and not in either daf-7(1372) (0.2%, n5
330) or daf-2(1391) animals, where 100% of the animals remained as dauers
(n5164). At 15°C with food, 65% of the daf-2(e1370) (n5384) and 43% of
the daf-7(e1372) dauer larvae (n5587) recovered and 76% of daf-2(e1391)
animals recovered (n5458). Atropine at 1 mM potently inhibits dauer
recovery of daf-2(e1370) to 18% (n5228), daf-7(e1372) to 6% (n5405), and
daf-2(e1391) to 23% (n5363) dauer larvae on plates at 15°C with food. For
daf-2(1370) and daf-7(e1372), each experiment was performed two or three
times, whereas for daf-2(e1391), the numbers are from only one trial. The
difference between the effects of atropine on pheromone-induced dauer
larvae and either daf-2-ordaf-7-induced dauer larvae may be because the
pheromone-induced dauer larvae had been arrested longer than the daf-2
and daf-7 dauer larvae. Older dauer larvae will recover when exogenous
pheromone is removed, even without the addition of food, but younger dauer
larvae do not (2). Alternatively, pheromone may inhibit both the TGF-
b
- and
insulin-like-signaling pathways (and perhaps other signals) whereas daf-2 or
daf-7 mutants may only decrease one endocrine signal.
Tissenbaum et al. PNAS
u
January 4, 2000
u
vol. 97
u
no. 1
u
463
NEUROBIOLOGY
insulin-signaling mutants. Thus, the cholinergic input to dauer
recovery depends on insulin-like signaling. We suggest that
muscarinic agonists induce recovery of the TGF-
b
pathway
mutant dauer larvae or pheromone-induced dauer larvae by
stimulating signaling in the daf-2 insulin-like pathway. In this
way, cholinergic stimulation can induce recovery in animals with
defective TGF-
b
pathway genes but not in animals with defective
insulin-like pathway genes.
In vertebrates, studies link the muscarinic and insulin signaling
pathways. Both adrenergic and cholinergic fibers innervate
secretory cells in the vertebrate islet of Langerhans (16, 31).
Consistent with the suggestion that muscarinic inputs increase C.
elegans insulin-like signaling, mammalian autonomic cholinergic
fibers enhance insulin secretion (29, 31). Pharmacological stim-
ulation with acetylcholine or carbachol induces insulin release
both in vivo and in vitro. This induction is completely abolished
by atropine, showing that it is mediated by activation of musca-
rinic receptors on the beta cells (16, 32, 33). In mammalian
systems, binding of acetylcholine to the beta cell muscarinic
receptor causes activation of sodium channels, which, in turn,
leads to a change in membrane potential to induce insulin release
(ref. 33; Fig. 3).
These data suggest the model shown in Fig. 3 for dauer
recovery in C. elegans. When pheromone levels decrease and
food levels increase, acetylcholine is secreted from an as yet
unidentified neuron and binds to the muscarinic receptor on an
insulin-like secreting neuron or other cell. This induces secretion
of an insulin-like signal, in turn, to induce dauer recovery.
Insulin-secreting pancreatic beta cells have many neuronal fea-
tures and are thought to be specialized ‘‘ganglia’’ related to the
enteric nervous system of lower vertebrates (34). In addition,
proteins related to insulin are produced by neurons that regulate
metabolism in Limulus (35). Distant relatives of insulin are
found in the C. elegans genome database (ref. 8; S. Pierce and
G.R., unpublished observations). We suggest that the secretory
cells expressing such an insulin-like gene will also express
muscarinic receptors and have connections to food, pheromone,
and temperature sensory neurons (Fig. 3).
Temperature acts as a modulator for dauer recovery (refs.
2 and 14; Fig. 2). The thermoregulatory circuit for temperature
sensation and output of that information to motor and endo-
crine pathways has been identified (11, 12, 36). This pathway
consists of the thermosensory neuron AFD coupled to the
interneurons AIY and AIZ (11, 12, 14, 36). ttx-3, a gene that
affects AIY function, is expressed exclusively in the AIY
interneurons (11, 12). Mutations in ttx-3 decouple this ther-
moregulatory pathway from the dauer pathway: daf-7; ttx-3
double mutant animals form dauer larvae that recover at high
temperature, unlike daf-7 single mutants (12). However, daf-2;
ttx-3 double mutant dauer larvae do not recover at high
temperature, like the daf-2 mutant alone (O. Hobert and G.R.,
unpublished observation). We suggest that thermosensor y
signals through the thermoregulatory AIY and AIZ interneu-
rons couple to insulin-like secretory neurons (Fig. 3). Given
that rates of growth and metabolism are intimately connected
to cultivation temperature in invertebrates, the coupling of
thermosensation to metabolic control is reasonable. Such a
coupling of thermosensory input to metabolic control by the
daf-2 insulin-like signaling pathway may be analogous (or even
homologous) to the hypothalamic modulation of autonomic
input to the pancreatic beta cells (31, 33, 34).
A muscarinic signaling pathway also induces recovery of the
hookworm infective L3 from their arrested ‘‘dauer’’ state. Re-
covery from dauer arrest in hookworm occurs in the host in
response to an undefined host-specific signal. We suggest that
up-regulation of an insulin-like molecule by a cholinergic path-
way also causes dauer recovery upon entr y into the host in A.
caninum. Known muscarinic signaling drugs may constitute
Fig. 3. A model for cholinergic input induction of dauer recovery. In dauer pheromone or in a daf-7 mutant, the DAF-7 TGF-
b
ligand is not produced by the
ASI sensoryysecretory neuron. Therefore, there is no activation of the DAF-1 and DAF-4 TGF-
b
receptors or downstream DAF-8 and DAF-14 Smad proteins, and
this results in high DAF-3 Smad activity in signaling cells or target tissues. In pheromone without muscarinic agonists, no insulin-like signal is released, causing
less insulin receptor signal transduction to the transcription factor DAF-16, which, in combination with unregulated DAF-3, induces dauer arrest. Under
pheromone-induced or daf-7 mutation-induced dauer induction conditions, muscarinic stimulation causes release of an insulin-like DAF-2 ligand, which
stimulates the DAF-2yAGE-1-signaling pathway to DAF-16 inactivation. Because daf-7 mutants can recover in muscarinic agonists, the TGF-
b
signaling pathway
is not required for dauer recovery. Under normal conditions of dauer recovery upon release from pheromone and addition of food and low temperature, we
suggest that acetylcholine released via temperature or food pathways binds to the muscarinic receptor on the insulin-like signaling cell to cause an increase in
insulin release. We suggest that temperature is coupled via the interneurons AIY and AIZ to the DAF-2 insulin-like signaling pathway rather than the TGF-
b
signaling pathway because mutations in the thermoregulatory gene ttx-3 can suppress mutations in daf-7 and not mutations in daf-2 (ref. 11; O. Hobert and
G.R., unpublished observations).
464
u
www.pnas.org Tissenbaum et al.
novel chemotherapeutic strategies to perturb the dauer mainte-
nance process in invertebrate hosts as well as the recovery
process in human hosts.
We thank Jim Thomas for providing some of the strains used in this study
and Ann Sluder for the minimal media protocol. Some of the strains were
obtained from the Caenorhabditis Genetics Center, which is supported
by the National Institutes of Health National Center for Research
Resources. We thank members of the Ruvkun and Kaplan labs and Allan
Dines for helpful discussions, suggestions, and critical reading of the
manuscript. This work was funded by a grant from Hoechst and National
Institutes of Health Grant R01AG14161 to G.R. Part of this work was
completed while H.A.T. was supported by the Helen Hay Whitney
Foundation.
1. Tauber, M. J., Tauber, C. A. & Masaki, S. (1986) in Seasonal Adaptation of
Insects (Oxford Univ. Press, New York).
2. Riddle, D. L. & Albert, P. S. (1997) in C. elegans II, eds. Riddle, D. L.,
Blumenthal, T., Meyer, B. J. & Priess, J. R. (Cold Spring Harbor Lab. Press,
Plainview, NY), pp. 739 –768.
3. Riddle, D. L. & Bird, A. F. (1985) J. Nematol. 17, 165–168.
4. Schmidt, G. D. & Roberts, L. S. (1985) in Foundations of Parasitology (Times
MirroryMosby, St. Louis).
5. Hawdon, J. M. & Schad, G. A. (1991) in Developmental Adaptations in
Nematodes., ed. Toft, C. A. (Oxford Univ. Press, Oxford), pp. 274 –298.
6. Patterson, G. I., Koweek, A., Wong, A., Liu, Y. & Ruvkun, G. (1997) Genes
Dev. 11, 2679–2690.
7. Ogg, S., Paradis, S., Gottlieb, S., Patterson, G. I., Lee, L., Tissenbaum, H. A.
& Ruvkun, G. (1997) Nature (London) 389, 994–999.
8. Duret, L., Guex, N., Peitsch, M. C. & Bairoch, A. (1998) Genome Res. 8,
348–353.
9. Kimura, K., Tissenbaum, H. A., Liu, Y. & Ruvkun, G. (1997) Science 277,
942–946.
10. Malone, E. A. & Thomas, J. H. (1994) Genetics 136, 879 886.
11. Hobert, O., D’Alberti, T., Liu, Y. & Ruvkun, G. (1998) J. Neurosci. 18,
2084–2096.
12. Hobert, O., Mori, I., Yamashita, U., Honda, H., Ohshima, Y., Liu, Y. &
Ruvkun, G. (1997) Neuron 19, 345–357.
13. Rand, J. B. & Nonet, M. L. (1997) in C. elegans II, eds. Riddle, D. L.,
Blumenthal, T., Meyer, B. J. & Priess, J. R. (Cold Spring Harbor Lab. Press,
Plainview, NY), pp. 611– 643.
14. Bargmann, C. I. & Mori, I. (1997) in C. elegans II, eds. Riddle, D. L.,
Blumenthal, T., Meyer, B. J. & Priess, J. R. (Cold Spring Harbor Lab. Press,
Plainview, NY), pp. 717–737.
15. Bargmann, C. I. (1998) Science 282, 2028 –2033.
16. Ahren, B., Taborsky, G. J., Jr., & Porte, D., Jr. (1986) Diabetologia 29, 827–836.
17. Miller, R. E. (1981) Endocr. Rev. 2, 471– 494.
18. Hawdon, J. M. & Schad, G. A. (1993) Exp. Parasitol. 77, 489– 491.
19. Sulston, J. & Hodgkin, J. (1988) in The Nematode Caenorhabditis elegans, ed.
Wood, W. B. (Cold Spring Harbor Lab. Press, Plainview, NY), pp. 587– 606.
20. Gottlieb, S. & Ruvkun, G. (1994) Genetics 137, 107–120.
21. Hawdon, J. M. & Schad, G. A. (1991) J. Helm. Soc. Wash. 58, 140 –142.
22. Hawdon, J. M., Jones, B. F., Perregaux, M. A. & Hotez, P. J. (1995) Exp.
Parasitol. 80, 205–211.
23. Hawdon, J. M. & Schad, G. A. (1990) J. Parasitol. 76, 394–398.
24. Aver y, L., Bargmann, C. I. & Horvitz, H. R. (1993) Genetics 134, 455–464.
25. Lef kowitz, R. J., Hoffman, B. B. & Taylor, P. (1996) in The Phar macological
Basis of Therapeutics, eds. Hardman, J. G. & Limbird, L. E. (McGraw–Hill, New
York), pp. 105–139.
26. Brown, J. H. & Taylor, P. (1996) in The Pharmacological Basis of Therapeutics,
eds. Hardman, J. G. & Limbird, L. E. (McGraw–Hill, New York), pp. 141–160.
27. Haim, N., Nahum, S. & Dudai, Y. (1979) J. Neurochem. 32, 543–552.
28. Lee, Y. S., Park, Y. S., Chang, D. J., Hwang, J. M., Min, C. K., Kaang, B. K.
& Cho, N. J. (1999) J. Neurochem. 72, 58– 65.
29. Yamamura, H. I. & Snyder, S. H. (1974) Proc. Natl. Acad. Sci. USA 1725–1729.
30. Burgermeister, W., Klein, W. L., Nirenberg, M. & Witkop, B. (1978) Mol.
Pharmacol. 14, 240 –256.
31. Woods, S. C. & Porte, D., Jr. (1974) Physiol. Rev. 54, 596– 619.
32. Latifpour, J., Gousse, A., Yoshida, M. & Weiss, R. M. (1992) J. Urol. 147,
760–763.
33. Henquin, J.-C. (1994) in Joslin’s Diabetes Mellitus, eds. Kahn, C. R. & Weir,
G. C. (Lea & Febiger, Malvern, PA), pp. 56– 80.
34. Bonner Weir, S. & Smith, F. E. (1994) in Joslin’s Diabetes Mellitus, eds. Kahn,
C. R. & Weir, G. C. (Lea & Febiger, Malvern, PA), pp. 15–28.
35. Roovers, E., Vincent, M. E., van Kesteren, E., Geraets, P. M., Planta, R. J.,
Vreugdenhil, E. & van Heerikhuizen, H. (1995) Gene 162, 181–188.
36. Mori, I. & Ohshima, Y. (1995) Nature (London) 376, 344 –348.
Tissenbaum et al. PNAS
u
January 4, 2000
u
vol. 97
u
no. 1
u
465
NEUROBIOLOGY
... Activation is thought to represent initial steps in the transition to the parasitic stage of the life cycle, and is analogous to feeding associated with recovery of the analogous C. elegans dauer larva from developmental arrest (Hawdon and Schad, 1991a;Hotez et al., 1993;Crook, 2014). The molecular components of the A. caninum activation pathway are highly conserved with those of the C. elegans dauer pathway, and include cGMP and insulin-like signalling (ILS) pathways (Tissenbaum et al., 2000;Hawdon and Datu, 2003;Brand and Hawdon, 2004;Gao et al., 2009;2010;Kiss et al., 2009;Wang et al., 2009). Here a triple resistant isolate of A. caninum that exhibits an alternate activation phenotype compared to our susceptible isolate is described. ...
... Due to its similarity to recovery from the dauer stage in C. elegans, hookworm larval activation is believed to represent an early step in the transition to parasitism that occurs during infection of a permissive host (Hawdon and Schad, 1991a;Hawdon and Hotez, 1996). The activation pathway in hookworms is highly conserved with recovery from the dauer stage in C. elegans, and is mediated by both cGMP and ILS pathways (Tissenbaum et al., 2000;Hawdon and Datu, 2003;Brand and Hawdon, 2004;Gao et al., 2009;Kiss et al., 2009;Gelmedin et al., 2011) (Fig. 5A). When BCR iL3 were tested in our activation assay, their maximum feeding level was significantly lower (approximately 40-50%) than the feeding level of the wild-type WMD and the double-resistant KGR isolates (Fig. 5B). ...
... Dauer recovery occurs in response to food and lower nematode density and is regulated by the mGC DAF-11 expressed in amphidial neurons ASI, ASJ and ASK (Hanna-Rose and Han, 2000). DAF-11 binds a bacterial food signal, initiating cGMP synthesis and subsequent signalling through a conserved ILS pathway to cause recovery (Tissenbaum et al., 2000;Murakami et al., 2001;Chung et al., 2013). The dauer pathway is well-studied genetically and biochemically, and the molecular components of the pathway have been identified (Riddle and Albert, 1997;Hu, 2007). ...
Article
Full-text available
Parasitic gastrointestinal nematodes pose significant health risks to humans, livestock, and companion animals, and their control relies heavily on the use of anthelmintic drugs. Overuse of these drugs has led to the emergence of resistant nematode populations. Herein, a naturally occurring isolate (referred to as BCR) of the dog hookworm, Ancylostoma caninum, that is resistant to 3 major classes of anthelmintics is characterized. Various drug assays were used to determine the resistance of BCR to thiabendazole, ivermectin, moxidectin and pyrantel pamoate. When compared to a drug-susceptible isolate of A. caninum, BCR was shown to be significantly resistant to all 4 of the drugs tested. Multiple single nucleotide polymorphisms have been shown to impart benzimidazole resistance, including the F167Y mutation in the β-tubulin isotype 1 gene, which was confirmed to be present in BCR through molecular analysis. The frequency of the resistant allele in BCR was 76.3% following its first passage in the lab, which represented an increase from approximately 50% in the founding hookworm population. A second, recently described mutation in codon 134 (Q134H) was also detected at lower frequency in the BCR population. Additionally, BCR exhibits an altered larval activation phenotype compared to the susceptible isolate, suggesting differences in the signalling pathways involved in the activation process which may be associated with resistance. Further characterization of this isolate will provide insights into the mechanisms of resistance to macrocyclic lactones and tetrahydropyrimidine anthelmintics.
... Dauer induction by food scarcity is mediated by the insulin pathway, since the perception of food changes the subcellular localization of DAF-16, requiring the insulinlike peptide DAF-28 [64]. Moreover, at this point, temperature cues feed into the insulin/IGF-1 pathway via ttx-3 [65][66][67]. Hence, not all environmental cues feed into the pathway at the level of the GPCRs. ...
... Moreover, chemical inhibition of Hc-DAF-9 prevents recovery from developmental arrest [111], suggesting that zooparasites synthesize an endogenous ligand for DAF-12. Indeed, earlier studies showed that host sterol molecules feed into the dauer pathway upstream of the insulin pathway, rather than into the DA biosynthesis pathway [65,97,112]. ...
Article
Full-text available
Nematodes are presumably the most abundant Metazoa on Earth, and can even be found in some of the most hostile environments of our planet. Various types of hypobiosis evolved to adapt their life cycles to such harsh environmental conditions. The five most distal major clades of the phylum Nematoda (Clades 8–12), formerly referred to as the Secernentea, contain many economically relevant parasitic nematodes. In this group, a special type of hypobiosis, dauer, has evolved. The dauer signalling pathway, which culminates in the biosynthesis of dafachronic acid (DA), is intensively studied in the free-living nematode Caenorhabditis elegans , and it has been hypothesized that the dauer stage may have been a prerequisite for the evolution of a wide range of parasitic lifestyles among other nematode species. Biosynthesis of DA is not specific for hypobiosis, but if it results in exit of the hypobiotic state, it is one of the main criteria to define certain behaviour as dauer. Within Clades 9 and 10, the involvement of DA has been validated experimentally, and dauer is therefore generally accepted to occur in those clades. However, for other clades, such as Clade 12, this has hardly been explored. In this review, we provide clarity on the nomenclature associated with hypobiosis and dauer across different nematological subfields. We discuss evidence for dauer-like stages in Clades 8 to 12 and support this with a meta-analysis of available genomic data. Furthermore, we discuss indications for a simplified dauer signalling pathway in parasitic nematodes. Finally, we zoom in on the host cues that induce exit from the hypobiotic stage and introduce two hypotheses on how these signals might feed into the dauer signalling pathway for plant-parasitic nematodes. With this work, we contribute to the deeper understanding of the molecular mechanisms underlying hypobiosis in parasitic nematodes. Based on this, novel strategies for the control of parasitic nematodes can be developed.
... Specifically, FOXO3 expression has been associated with long-lived populations of humans in several independent cohorts (Broer et al., 2015;Morris et al., 2015;Soerensen et al., 2015) suggesting that activation of this pathway is one of the most promising routes to benefiting The healthspan and lifespan extensions induced by atracurium were dependent on the AChR subunit unc-38, demonstrating that atracurium works to promote longevity through its canonical mechanism. Though the specific role of AChR antagonists was previously unstudied, agonists of the AChR were shown to promote recovery from the developmentally arrested dauer phase (Tissenbaum et al., 2000). This effect is dependent on insulin-like neuroendocrine signaling (Tissenbaum et al., 2000), a major regulator of DAF-16 (Gottlieb & Ruvkun, 1994). ...
... Though the specific role of AChR antagonists was previously unstudied, agonists of the AChR were shown to promote recovery from the developmentally arrested dauer phase (Tissenbaum et al., 2000). This effect is dependent on insulin-like neuroendocrine signaling (Tissenbaum et al., 2000), a major regulator of DAF-16 (Gottlieb & Ruvkun, 1994). In that study, the authors suggest a mechanism by which ACh activation of the AChR activates DAF-2, which would then inhibit activation of DAF-16. ...
Article
Full-text available
Transcriptome-based drug screening is emerging as a powerful tool to identify geroprotective compounds to intervene in age-related disease. We hypothesized that, by mimicking the transcriptional signature of the highly conserved longevity intervention of FOXO3 (daf-16 in worms) overexpression, we could identify and repurpose compounds with similar downstream effects to increase longevity. Our in silico screen, utilizing the LINCS transcriptome database of genetic and compound interventions, identified several FDA-approved compounds that activate FOXO downstream targets in mammalian cells. These included the neuromuscular blocker atracurium, which also robustly extends both lifespan and healthspan in Caenorhabditis elegans. This longevity is dependent on both daf-16 signaling and inhibition of the neuromuscular acetylcholine receptor subunit unc-38. We found unc-38 RNAi to improve healthspan, lifespan, and stimulate DAF-16 nuclear localization, similar to atracurium treatment. Finally, using RNA-seq transcriptomics, we identify atracurium activation of DAF-16 downstream effectors. Together, these data demonstrate the capacity to mimic genetic lifespan interventions with drugs, and in doing so, reveal that the neuromuscular acetylcholine receptor regulates the highly conserved FOXO/DAF-16 longevity pathway.
... TGF-β & IIS) (Bargmann 1998;Fielenbach and Antebi 2008;Hilliard et al. 2002). Also, the binding to amphid chemical receptors may trigger subordinate signaling pathways as for example muscarinic signaling (Tissenbaum et al. 2000). Aumann and Ehlers (2001) observed that n-hexane and chloroform eluates of the bacterial supernatant were biologically inactive, suggesting the presence of a molecule acting antagonistically to the recovery-inducing signal. ...
Article
Full-text available
The entomopathogenic nematode (EPN) Heterorhabditis bacteriophora is an effective biological-control agent of insect pests. The dauer juveniles (DJs) seek for, infect insects, and release cells of the carried symbiotic bacterium of the genus Photorhabdus. Inside the host, the DJs perceive signals from the insect’s haemolymph that trigger the exit from the arrested stage and the further development to mature adults. This developmental step is called DJ recovery. In commercial production, a high and synchronous DJ recovery determines the success of liquid-culture mass production. To enhance the understanding about genetic components regulating DJ recovery, more than 160 mutant- and 25 wild type inbred lines (WT ILs) were characterized for DJ recovery induced by cell-free bacterial supernatant. The mutant lines exhibited a broader DJ recovery range than WT ILs (4.6–67.2% vs 1.6–35.7%). A subset of mutant lines presented high variability of virulence against mealworm (Tenebrio molitor) (from 22 to 78% mortality) and mean time survival under oxidative stress (70 mM H2O2; from 10 to 151 h). Genotyping by sequencing of 96 mutant lines resulted in more than 150 single nucleotide polymorphisms (SNPs), of which four results are strongly associated with the DJ recovery trait. The present results are the basis for future approaches in improving DJ recovery by breeding under in vitro liquid-culture mass production in H. bacteriophora. This generated platform of EMS-mutants is as well a versatile tool for the investigation of many further traits of interest in EPNs. Keypoints • Exposure to bacterial supernatants of Photorhabdus laumondii induces the recovery of Heterorhabditis bacteriophora dauer juveniles (DJs). Both, the bacteria and the nematode partner, influence this response. However, the complete identity of its regulators is not known. • We dissected the genetic component of DJ recovery regulation in H. bacteriophora nematodes by generating a large array of EMS mutant lines and characterizing their recovery pheno- and genotypes. • We determined sets of mutants with contrasting DJ recovery and genotyped a subset of the EMS-mutant lines via genotyping by sequencing (GBS) and identified SNPs with significant correlation to the recovery trait.
... Some cell death was noticeable for all treatments, except acetylcholine, at high concentrations (>1 mM) at 48 h (Fig. 1F). While the OXO-mediated effects suggest that perturbation of Bma-GAR-3 may elicit phenotypes of interest, the slower neuromodulatory action of this general receptor class may require assays sensitized to other subtle but important phenotypes in the host context (72,73). More insight into the tissue-specific expression patterns of this highlyexpressed receptor would allow better prediction of its physiological roles. ...
Article
Full-text available
The diversification of anthelmintic targets and mechanisms of action will help ensure the sustainable control of nematode infections in response to the growing threat of drug resistance. G protein-coupled receptors (GPCRs) are established drug targets in human medicine but remain unexploited as anthelmintic substrates despite their important roles in nematode neuromuscular and physiological processes. Bottlenecks in exploring the druggability of parasitic nematode GPCRs include a limited helminth genetic toolkit and difficulties establishing functional heterologous expression. In an effort to address some of these challenges, we profile the function and pharmacology of muscarinic acetylcholine receptors in the human parasite Brugia malayi, an etiological agent of human lymphatic filariasis. While acetylcholine-gated ion channels are intensely studied as targets of existing anthelmintics, comparatively little is known about metabotropic receptor contributions to parasite cholinergic signaling. Using multivariate phenotypic assays in microfilariae and adults, we show that nicotinic and muscarinic compounds disparately affect parasite fitness traits. We identify a putative G protein-linked acetylcholine receptor of B. malayi (Bma-GAR-3) that is highly expressed across intramammalian life stages and adapt spatial RNA in situ hybridization to map receptor transcripts to critical parasite tissues. Tissue-specific expression of Bma-gar-3 in Caenorhabditis elegans (body wall muscle, sensory neurons, and pharynx) enabled receptor deorphanization and pharmacological profiling in a nematode physiological context. Finally, we developed an image-based feeding assay as a reporter of pharyngeal activity to facilitate GPCR screening in parasitized strains. We expect that these receptor characterization approaches and improved knowledge of GARs as putative drug targets will further advance the study of GPCR biology across medically important nematodes.
... However some larvae will bypass the dauer stage and continuously develop (Narasimhan et al., 2011;Ogg et., 1997). Arrested animals may spontaneously recover from dauer while still at 24°C (Tissenbaum et al., 2000;Nika et al., 2016). ...
Article
Caenorhabditis elegans can enter a diapause stage called “dauer” when it senses that the environment is not suitable for development. This implies a detour from the typical developmental trajectory and requires a tight control of the developmental clock and a massive tissue remodeling. In the last decades, core components of the signaling pathways that govern the dauer development decision have been identified, but the tissues where they function for the acquisition of dauer-specific traits are still under intense study. Growing evidence demonstrates that these pathways engage in complex cross-talk and feedback loops. In this review, we summarize the current knowledge regarding the transcriptional regulation of the dauer program and the relevant tissues for its achievement. A better understanding of this process will provide insight on how developmental plasticity is achieved and how development decisions are under a robust regulation to ensure an all-or-nothing response. Furthermore, this developmental decision can also serve as a simplified model for relevant developmental disorders. Abbreviations: AID Auxin Induced Degron DA dafachronic acid Daf-c dauer formation constitutive Daf-d dauer formation defective DTC Distal Tip Cells ECM modified extracellular matrix GPCRs G protein-coupled receptors IIS insulin/IGF-1 signaling ILPs insulin-like peptides LBD Ligand Binding Domain PDL4 Post Dauer L4 TGF-β transforming growth factor beta WT wild-type
Preprint
Full-text available
The diversification of anthelmintic targets and mechanisms of action will help ensure the sustainable control of nematode infections in response to the growing threat of drug resistance. G protein-coupled receptors (GPCRs) are established drug targets in human medicine but remain unexploited as anthelmintic substrates despite their important roles in nematode neuromuscular and physiological processes. Bottlenecks in exploring the druggability of parasitic nematode GPCRs include a limited helminth genetic toolkit and difficulties establishing functional heterologous expression. In an effort to address some of these challenges, we profile the function and pharmacology of muscarinic acetylcholine receptors in the human parasite Brugia malayi, an etiological agent of human lymphatic filariasis. While acetylcholine-gated ion channels are intensely studied as targets of existing anthelmintics, comparatively little is known about metabotropic receptor contributions to parasite cholinergic signaling. Using multivariate phenotypic assays in microfilariae and adults, we show that nicotinic and muscarinic compounds disparately affect parasite fitness traits. We identify a putative G protein-linked acetylcholine receptor (Bma-GAR-3) that is highly expressed across intra-mammalian life stages and adapt spatial RNA in situ hybridization to map receptor transcripts to critical parasite tissues. Tissue-specific expression of Bma-gar-3 in Caenorhabditis elegans (body wall muscle, sensory neurons, and pharynx) enabled receptor deorphanization and pharmacological profiling in a nematode physiological context. Lastly, we developed an image-based feeding assay as a reporter of pharyngeal activity to facilitate GPCR screening in parasitized strains. We expect that these receptor characterization approaches and improved knowledge of GARs as putative drug targets will further advance the study of GPCR biology across medically important nematodes.
Conference Paper
Effects of food deprivation and octopamine on the sensitivity of behavior of nematode C. elegans to agonist of nAChRs levamisole and partial inhibition of ACh-esterase by aldicarb were investigated. Either food deprivation or octopamine caused sensitization of swimming as induced by mechanical stimulus to aldicarb and levamisole action, which was revealed in disturbances of coordination of muscles necessary for swimming. These results show that both food deprivation and exogenous octopamine caused activation of C. elegans cholinergic system. The joint action of food deprivation and octopamine revealed their additive effects. Therefore, food deprivation caused adaptive activation of cholinergic system via nonidentified mechanism independently from octopamine secretion. Activation of cholinergic system by food deprivation may be an adaptive reaction of C. elegans nervous system necessary for movement speed elevation to avoid local unfavorable environment.
Article
Mechanisms governing morphogenesis and development of infectious third-stage larvae (L3i) of parasitic nematodes have been likened to those regulating dauer development in Caenorhabditis elegans. Dauer regulatory signal transduction comprises initial G protein-coupled receptor (GPCR) signaling in chemosensory neurons of the amphidial complex that regulates parallel insulin- and TGFβ-like signaling in the tissues. Insulin- and TGFβ-like signals converge to co-regulate steroid signaling through the nuclear receptor (NR) DAF-12. Discovery of the steroid ligands of DAF-12 opened a new avenue of small molecule physiology in C. elegans. These signaling pathways are conserved in parasitic nematodes and an increasing body of evidence supports their function in formation and developmental regulation of L3i during the infectious process in soil transmitted species. This review presents these lines of evidence for G protein-coupled receptor (GPCR), insulin- and TGFβ-like signaling in brief and focuses primarily on signaling through parasite orthologs of DAF-12. We discuss in some depth the deployment of sensitive analytical techniques to identify Δ7-dafachronic acid as the natural ligand of DAF-12 homologs in Strongyloides stercoralis and Haemonchus contortus and of targeted mutagenesis by CRISPR/Cas9 to assign dauer-like regulatory function to the NR Ss-DAF-12, its coactivator Ss-DIP-1 and the key ligand biosynthetic enzyme Ss-CYP-22a9. Finally, we present published evidence of the potential of Ss-DAF-12 signaling as a chemotherapeutic target in human strongyloidiasis.
Article
Full-text available
In mammals, insulin signalling regulates glucose transport together with the expression and activity of various metabolic enzymes. In the nematode Caenorhabditis elegans, a related pathway regulates metabolism, development and longevity. Wild-type animals enter the developmentally arrested dauer stage in response to high levels of a secreted pheromone, accumulating large amounts of fat in their intestines and hypodermis. Mutants in DAF-2 (a homologue of the mammalian insulin receptor) and AGE-1 (a homologue of the catalytic subunit of mammalian phosphatidylinositol 3-OH kinase) arrest development at the dauer stage. Moreover, animals bearing weak or temperature-sensitive mutations in daf-2 and age-1 can develop reproductively, but nevertheless show increased energy storage and longevity. Here we show that null mutations in daf-16 suppress the effects of mutations in daf-2 or age-1; lack of daf-16 bypasses the need for this insulin receptor-like signalling pathway. The principal role of DAF-2/AGE-1 signalling is thus to antagonize DAF-16. daf-16 is widely expressed and encodes three members of the Fork head family of transcription factors. The DAF-2 pathway acts synergistically with the pathway activated by a nematode TGF-β-type signal, DAF-7, suggesting that DAF-16 cooperates with nematode SMAD proteins in regulating the transcription of key metabolic and developmental control genes. The probable human ortholognes of DAF-16, FKHR and AFX, may also act downstream of insulin signalling and cooperate with TGF-β effectors in mediating metabolic regulation. These genes may be dysregulated in diabetes.
Article
Full-text available
We have identified three new families of insulin homologs in Caenorhabditis elegans. In two of these families, concerted mutations suggest that an additional disulfide bond links B and A domains, and that the A-domain internal disulfide bond is substituted by a hydrophobic interaction. Homology modeling remarkably confirms these predictions and shows that despite this atypical disulfide bond pattern and the absence of C-like peptide, all these proteins may adopt the same fold as the insulin. Interestingly, whereas we identified 10 insulin-like peptides, only one insulin-like-receptor (daf-2) has been found. We propose that these insulin-related peptides may correspond to different activators or inhibitors of the daf-2 insulin-regulating pathway.
Article
In Caenorhabditis elegans, formation of the developmentally arrested dauer larva is induced by high levels of a constitutively secreted pheromone. Synergy between two groups of incompletely penetrant dauer-constitutive (Daf-c) mutations has recently led to a proposal that these two groups of genes are partially redundant and function in two parallel pathways that regulate dauer formation. A possible weakness in this reasoning is that the mutations used to identify the synergy were specifically obtained as incompletely penetrant mutations. Here we use screens to identify new Daf-c alleles without any requirement for partial penetrance. Nevertheless, 22 of the 25 new mutations are incompletely penetrant mutations in 6 previously identified genes. Among these are mutations in daf-8 and daf-19, genes for which only one mutation had been previously identified. Also included in this group are three daf-1 alleles that do not exhibit the maternal rescue characteristic of other daf-1 alleles. Two of the 25 new mutations are fully penetrant and are alleles of daf-2, the one gene in which a fully penetrant mutation had been found earlier. Finally, one of the 25 new mutations is semidominant, temperature-sensitive, and identifies a new gene, daf-28. The results demonstrate that an incompletely penetrant Daf-c phenotype is characteristic of mutations in most Daf-c genes other than daf-2. This finding strengthens the hypothesis that a branched genetic pathway controls dauer formation.
Article
With the assistance of two new associate editors, Alfred G. Gilman and George B. Koelle, the fifth edition retains the excellence set by previous editions of this classic text. To maintain the same overall length, several outmoded areas have been abbreviated or eliminated. There are discussions of many new drugs, as well as promising drugs whose availability in the United States is probable in the near future.The section on chemotherapy of neoplastic diseases has undergone extensive revision. The introductory section contains a very readable and informative discussion of pharmacokinetics, pharmacodynamics, and an interesting account of the process of development and approval of new drugs. The section on Parkinson disease has been extensively revised, with major emphasis on levodopa. The chapter on neurohumoral transmission and the autonomic nervous system remains the best source of information in this complex and rapidly changing area. The treatment of diuretics is completely up to
Article
Abstract—The powerful muscarinic antagonist [3H]quinuclidinyl benzilate (QNB) specifically binds to homogenates of Drosophila melanogaster head at a level of 65 ± 6 fmol/mg protein, with an apparent dissociation constant of 0.15–0.7 nM. The half-life of the ligand-receptor complex at 25°C is 30–40 min. Binding is inhibited by low concentrations of muscarinic ligands but not by low concentrations of nicotinic ligands, anticholinesterases or non-cholinergic drugs. Binding-sites are membrane bound and are inactivated by trypsin and by Triton X-100. Part of the activity (<20%) is released into a high speed supernatant by 2 M-NaCI. The gene coding for the putative muscarinic receptor in Drosophila is apparently not located adjacent to the gene for acetylcholinesterase
Article
In the pond snail Lymnaea stagnalis (Ls), growth and associated processes are likely to be controlled by a family of molluscan insulin-related peptides (MIP). Here we report on the cloning of a cDNA encoding a putative receptor for these MIP. This cDNA was isolated from Ls via PCR with degenerate oligodeoxynucleotides corresponding to conserved parts of the tyrosine kinase domain of the human insulin receptor and its Drosophila homologue. Many of the typical insulin-receptor features, including a cysteine-rich domain, a single transmembrane domain and a tyrosine-kinase domain are conserved in the predicted, 1607-amino acid (aa) protein. Comparison of the aa sequence of the molluscan receptor to other insulin-receptor sequences revealed strong variations in the percentage of sequence identity for the different domains, ranging from 70% sequence identity in the tyrosine-kinase domain to virtually no sequence identity in the C-terminal sequence. Striking differences are the absence of a clear tetrabasic cleavage site, and the extremely long C-terminus of 308 aa that contains seven Tyr residues. Southern blot analyses at varying stringencies, extensive screening of cDNA- and genomic libraries, and PCR experiments indicate the presence of a single putative MIP receptor. This suggests that the four different MIP may exert their functional role in Ls by binding to the same receptor.
Article
Article
The infective dauer juvenile (DJ2) of Anguina agrostis, a stage capable of surviving desiccation, is up to sixfold more resistant to the detergent sodium dodecyl sulfate than are freshly hatched juveniles or adult males, and twofold more resistant to the anesthetic phenoxypropanol. Thus, the DJ2, like dauer stages of other species, may also be more resistant to various types of environmental stress in its natural habitat. In A. agrostis, however, resistance appears to be acquired gradually during development of the second juvenile stage, rather than during a molt.
Article
Previous studies from our laboratory demonstrated that there is an up-regulation of muscarinic receptor density in the bladder dome of the 8-wks diabetic rat compared to control. To determine whether the changes observed in receptor density can be corrected by insulin or dietary myoinositol, five groups of rats were maintained for eight weeks: control (C), diabetic (D), diabetic insulin-treated (DI), diabetic myoinositol-treated (DMI), and control myoinositol-treated (CMI). Diabetes was induced by i.v. injection of 65 mg./kg. of streptozotocin. D and DMI animals were smaller, had higher serum glucose and lower serum insulin levels, higher water intakes and urine outputs, and larger bladder domes than the other groups. The density of the muscarinic receptors measured by radioligand receptor binding assays using [3H]quinuclidinyl benzilate were (in fmol./mg. protein): C, 88 +/- 13; D, 176 +/- 19; DI, 94 +/- 5; DMI, 158 +/- 8; CMI, 112 +/- 10. These data indicate that insulin, but not myoinositol treatment normalized diabetes induced abnormalities observed in the general features of streptozotocin-injected rats and prevented the diabetic-induced upregulation of bladder dome muscarinic receptors.