You are on page 1of 12

Developmental Biology 475 (2021) 265–276

Contents lists available at ScienceDirect

Developmental Biology
journal homepage: www.elsevier.com/locate/developmentalbiology

Developmental plasticity and the response to nutrient stress in


Caenorhabditis elegans
Sabih Rashid 1, Christopher Wong 1, Richard Roy *
Department of Biology, McGill University, 1205 Avenue Docteur Penfield, Montreal, QC, H3A 1B1, Canada

A R T I C L E I N F O A B S T R A C T

Keywords: Developmental plasticity refers the ability of an organism to adapt to various environmental stressors, one of
Caenorhabditis elegans which is nutritional stress. Caenorhabditis elegans require various nutrients to successfully progress through all the
Developmental plasticity larval stages to become a reproductive adult. If nutritional criteria are not satisfied, development can slow or
Starvation
completely arrest. In poor growth conditions, the animal can enter various diapause stages, depending on its
AMPK
Diapause
developmental progress. In C. elegans, there are three well-characterized diapauses: the L1 arrest, the dauer
Dauer larva diapause, and adult reproductive diapause, each associated with drastic changes in metabolism and germline
Germ line development. At the centre of these changes is AMP-activated protein kinase (AMPK). AMPK is a metabolic
Quiescence regulator that maintains energy homeostasis, particularly during times of nutrient stress. Without AMPK, meta-
bolism is disrupted during dauer, leading to the rapid consumption of lipid stores as well as misregulation of
metabolic enzymes, leading to reduced survival. During the L1 arrest and dauer diapause, AMPK is responsible for
ensuring germline quiescence by modifying the germline chromatin landscape to maintain germ cell integrity
until conditions improve. Similar to classic hormonal signalling, small RNAs also play a critical role in regulating
development and behaviour in a cell non-autonomous fashion. Thus, during the challenges associated with
developmental plasticity, AMPK summons an army of signalling pathways to work collectively to preserve
reproductive fitness during these periods of unprecedented uncertainty.

1. Caenorhabditis elegans as a model for developmental plasticity Caenorhabditis elegans is an ideal model to study various aspects of
developmental plasticity. In the wild, they are free-living nematodes that
The development of an organism is heavily influenced by environ- feed primarily on microorganisms present in decaying organic material
mental factors. These include temperature, weather, and even the pres- (Frezal and Felix, 2015). Because of their simple anatomy, quick devel-
ence of other species, all of which can impact development at various opment time, and adaptation to laboratory environments, a vast array of
levels (R
egniere et al., 2012; Prudic et al., 2011; O’Leary et al., 2017). molecular and genetic tools have been developed to interrogate almost
However, while different species display diverse regulation of develop- every aspect of their cellular and organismal function.
ment through environmental factors, growth in all organisms is influ- Development in C. elegans is regulated by the activity and cross talk
enced by nutrient availability. Lack of essential nutrients can slow or between a number of conserved signalling pathways (Greenwald et al.,
even arrest growth outright, and in some cases trigger organisms to un- 1983; Friedman et al., 1988; Kimura et al., 1997; Long et al., 2002; Jia
dergo alternative developmental programs (Lafuente and Beldade, et al., 2004; Lapierre and Hansen, 2012; Blackwell et al., 2019).
2019). This developmental plasticity serves as an adaptive means for Numerous temporally oscillating proteins and small RNAs, which regu-
organisms to survive acute or chronic environmental stress, allowing late the timing of development, have also been identified (Lee et al.,
them to adjust their physiology and growth until environmental condi- 1993; Johnstone and Barry, 1996). The effects of these pathways are
tions improve (Braendle and Felix, 2008; Sommer, 2020). This form of particularly robust, resulting in an essentially invariant pattern of cell
organismal adaptation requires that key cellular sensors must respond to division and development, provided that environmental fluctuation is
such cues and transduce this information globally through a robust, minimal. However, during periods of environmental challenge, C. elegans
well-established network of signalling pathways. can also modulate its developmental rate and even undergo alternative

* Corresponding author.
E-mail address: richard.roy@mcgill.ca (R. Roy).
1
These authors contributed equally to this work.

https://doi.org/10.1016/j.ydbio.2021.01.015
Received 13 October 2020; Received in revised form 24 December 2020; Accepted 29 January 2021
Available online 4 February 2021
0012-1606/© 2021 The Authors. Published by Elsevier Inc. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-
nc-nd/4.0/).
S. Rashid et al. Developmental Biology 475 (2021) 265–276

developmental trajectories to survive bouts of environmental stress to reversed upon refeeding and execution of the larval developmental
ultimately enhance fitness. program (Roux et al., 2016).
Nutrition is one of the major determinants of C. elegans growth and In addition to the L1 arrest, C. elegans can arrest their development in
development. In addition to the absence of food, which can slow or arrest later larval stages, following a ‘nutritional checkpoint’ that exists
development, C. elegans also requires the presence of various micro- following some of the later larval moults (Schindler et al., 2014).
nutrients for proper growth. These include vitamins, amino acids, heme, C. elegans may also enter an adult reproductive diapause (ARD) upon
and cholesterol (Zecic et al., 2019). These and other nutrients are starvation in the L4 stage, which is characterized by a shrinking of the
required throughout development or at specific life stages, often germ line and an increase in longevity (Angelo and Gilst, 2009).
reflecting a distinct developmental requirement for a given metabolite. While temporary developmental arrests may potentially provide a
Heme deficiency, for instance, will only cause larval arrest at the L4 short-term survival advantage, animals can adjust their developmental
stage, prior to which C. elegans can develop without the nutrient and not progression for longer diapauses by entering into an alternate L3 stage
suffer any apparent consequences (Rao et al., 2005). called the dauer diapause. Dauer larvae are characterized by quiescent
Based on the numerous nutritional needs of the organism, a chemi- behaviour, altered metabolism, a hardened protective cuticle, and sig-
cally defined minimal medium has been developed, which provides the nificant changes in gene regulation (Cassada and Russell, 1975; Wang
essential nutrients required to support C. elegans development (Szewczyk and Kim, 2003). In this alternate stage, wild-type animals can survive for
et al., 2003). The medium can serve as a powerful tool to test the effects up to four months, until more favourable environmental conditions arise.
of various metabolite supplements on growth. Given the natural habitat In their natural habitat, wild-type animals display a behaviour called
of C. elegans, animals can subsist on a wide variety of diets, each of which nictation, where the animal stands on its tail and waves its head, pre-
modulates their growth in different ways. In some cases, the specific sumably as an attempt to attach onto a passing host (Felix and Braendle,
dietary metabolites within their food that regulate growth have been 2010). This likely increases the chance of larval dispersal followed by a
identified (MacNeil et al., 2013). Recent efforts have attempted to cate- subsequent change in environment. Because this can lead to dramatic
gorize the various natural diets of C. elegans in order to better understand changes in nutrient status in a very short period of time, dauer larvae are
how the organism’s developmental and metabolic processes have able to quickly readjust their metabolism and resume their growth,
evolved alongside its environment (Schulenburg and Felix, 2017). allowing them to return to a reproductive mode of development.
One notable feature of C. elegans development is their ability to Nevertheless, animals that have transited through the dauer diapause
reversibly arrest their development in response to environmental stress. have an altered life history that is recorded in their transcriptome (Hall
The two most well-studied of these “diapauses” occur following et al., 2010), suggesting that developmental plasticity of such a drastic
embryogenesis at the L1 diapause, or later in larval development during nature can have long-term consequences for the organism.
the dauer diapause (Fig. 1). The L1 diapause occurs during the first (L1)
larval stage and is characterized by a global developmental quiescence 2. Metabolic responses to nutrient stress
that is triggered in emergent larvae that hatch in the absence of nutrients
(Hong et al., 1998; Baugh and Sternberg 2006). While wild type animals 2.1. The dauer detour
can survive for up to ten days without food, their capacity to initiate
post-embryonic development decreases with the duration of the When faced with overcrowding, high temperatures, or a lack of food,
diapause, suggesting that this developmental plasticity comes at a cost late stage L1 larvae can alter their developmental trajectory and delay
(Zhang et al., 2011). Curiously, as animals remain in the diapause, they reproduction by executing a preparatory second larval stage (L2d) fol-
begin to exhibit features typical of aged adult animals including accu- lowed by a quiescent third larval stage (dauer) to enhance survival until
mulation of proteotoxic aggregates, though many of these are completely environmental conditions become more favourable and capable of sup-
porting their progeny (Cassada and Russell, 1975). This dauer diapause is
notable as it results in significant changes in animal morphology and
behaviour, as opposed to simply slowing or arresting growth, and thus
serves as a striking example of developmental plasticity. (Riddle et al.,
1981; Hu, 2007). In addition, this diapause is highly conserved among
numerous nematode species, including various parasites, where it aids in
host infection (Hotez et al., 1993; Hawdon and Schad, 1991). This has led
to some speculation that the evolution of some nematode species from
soil-dwelling roundworms to parasites that live within hosts may have
been aided by the dauer adaptation (Crook, 2014), which facilitated the
infection of other organisms by allowing parasites to adapt to the often
harsh internal environments typical of their hosts (Harvey et al., 2000).
Dauer formation is mediated by a dauer pheromone which is secreted
continuously by the animals (Golden and Riddle, 1982; 1984; Fielenbach
and Antebi, 2008). When levels of the pheromone in a population surpass
a threshold, animals will activate the dauer program. The ensuing
response is relayed to amphid neurons in the head region of the larva
(Bargmann, 1991), and the subsequent morphological and metabolic
adaptations are regulated by a number of signalling pathways, including
the insulin/IGF-1, TGFβ, and cyclic GMP signalling pathways (Kimura
et al., 1997; Ren et al., 1996; Riddle and Albert, 1997; Birnby et al.,
2000). This complex regulation is expected, given the substantial
changes in gene expression that distinguish the dauer larva from L3
Fig. 1. C. elegans can undergo developmental arrest at various stages in larvae which undergo continuous reproductive development (Jeong
response to starvation. Upon encountering energetic or environmental et al., 2009).
stresses, the animal can enter a diapause stage depending on its current devel- Animals that have passed through dauer exhibit significantly altered
opmental stage. In all of the described diapause stages, the germ cells undergo gene expression profiles compared to animals that develop in replete
G2 stage cell cycle arrest. conditions (Hall et al., 2010). This transcriptional memory may be

266
S. Rashid et al. Developmental Biology 475 (2021) 265–276

preserved epigenetically through changes in histone modifications, as promoting exit and resumption of reproductive development.
post-dauer animals generally exhibit decreased levels of H3K4me3 and In addition to the effects of NADPH in regulating dauer formation,
H4panAc. Post-dauer animals also exhibit a detectable extension in previous work suggests that a redox catalyzing enzyme called thio-
lifespan and larger brood sizes, potentially increasing reproductive redoxin trx-1 also regulates dauer formation through interactions with
fitness. It is so far unclear whether there are negative consequences the cGMP pathway (Sanzo-Machuca et al., 2019). This study reveals a
associated with the altered post-dauer chromatin signature. Hall et al. redox-dependent role of trx-1 in maintaining the longevity of long-lived
speculate that this phenotypic diversity that arises due to this alternate mutants, including the daf-2 (insulin-like receptor) mutant. Interestingly,
life history may provide an evolutionary advantage, in addition to the the loss of the catalytic or regulatory subunits of AMPK also severely
survival advantage conferred by the stress-resistant dauer stage. suppressed the lifespan extension conferred by daf-2, while also
dramatically decreasing survival of the dauer larva (Narbonne and Roy,
2006). AMPK plays a role in maintaining redox balance during periods of
2.2. Redox state and the regulation of dauer formation
metabolic stress, such as glucose starvation (Ren and Shen, 2019). AMPK
is also a target of Trx1 in mice (Shao et al., 2014), suggesting that the
Dauer formation is triggered by increased levels of dauer pheromone
dauer survival defect seen in AMPK mutants could be compounded by a
in a growing population of animals. This affects a number of downstream
misregulation of redox balance, although it is unclear whether the defect
steps that converge on DAF-12, a nuclear hormone receptor-like tran-
is a result of damage due to elevated ROS levels, or reductive stress
scription factor that can bind several steroidal ligands known as
resulting from the progressive accumulation of toxic redox/metabolic
dafachronic acids (DAs) (Motola et al., 2006) (Fig. 2). Reproductive
by-products (Tilton et al., 1991).
growth is maintained when DAF-12 is bound by DAs, but when DAF-12 is
in its unliganded state it binds to DIN-1S, forming a transcriptional
complex that blocks gene expression required for reproductive develop- 2.3. Metabolic shifts in the dauer stage
ment, while activating dauer-specific genes (Antebi et al., 2000; Ludewig
et al., 2004). Given that dauer larvae often must survive for extended durations
DA formation is itself regulated by levels of the electron donor until their surrounding environmental conditions improve, their global
NADPH. Penkov et al., discovered that a tug of war between DA synthesis metabolism must adjust to accommodate a potentially lengthy quies-
and the biosynthesis of the saccharide trehalose helps determine dauer cence. Oxidative metabolism is greatly reduced in dauer larvae,
formation. Synthesis of trehalose from glucose-6-phosphate (G6P) pre- compared to animals that bypassed this diapause (Wang and Kim, 2003).
vents G6P entry into the pentose phosphate pathway (Wamelink et al., Instead, dauer larvae shift towards glyoxylate and gluconeogenic path-
2008), thus reducing levels of cellular NADPH which is required for DA ways to generate energy. The shift towards gluconeogenesis, in partic-
synthesis by DAF-9 (Motola et al., 2006). Without DAs to bind it, DAF-12 ular, results in the accumulation of glycogen and trehalose. SAGE and
is activated and dauer formation is promoted. Notably, trehalose levels microarray analysis of dauer gene expression also indicates increases in
are increased during starvation, promoting stress resistance and genes involved in β-oxidation of fatty acids (Jones et al., 2001).
longevity (Hibshman et al., 2017). Dauer formation thus invokes com- Many of these changes in metabolic gene expression represent a shift
mon stress response pathways, highlighting the coordination of devel- towards a regulated consumption of existing energy stores, as dauer
opmental progression and metabolic adaptation. Other methods of larvae do not feed, and thus must subsist entirely on stored nutrients. The
reducing cellular NADPH, such as disrupting the activity of isocitrate shift towards gluconeogenesis is regulated primarily by AAK-2, one of the
dehydrogenase, also induces dauer formation, suggesting internal two catalytic components of AMPK, while the switch to the glyoxylate
metabolite concentrations, as well as redox state, are significant con- cycle was mediated by AAK-2 and DAF-12 (Penkov et al., 2020). This
tributors to dauer formation. DAF-12 itself appears to induce the shift occurs as a result of stoichiometric ratios of enzymes within dauer
biosynthesis of trehalose, reducing levels of NADPH in response to dauer larvae. Specifically, ICL-1, a glyoxylate cycle enzyme, is upregulated in
pheromone, thus creating a feedback loop to promote dauer formation. dauer larvae, whereas this ratio is reduced in aak-2 mutants. In the
NADþ, meanwhile, is sufficient to trigger dauer exit in C. elegans absence of an appropriate metabolic adjustment, the survival of the dauer
(Mylenko et al., 2016). NADþ, and potentially other bacterial compo- larva may be compromised. Loss of the insulin signalling effector daf-16,
nents, can thus serve as signals that indicate the presence of food, for example, leads to a misregulation of catabolism, causing many sugars,

Fig. 2. Dauer formation in C. elegans is tightly regulated and invokes numerous metabolic changes. Environmental stressors such as lack of food or high
population density can trigger entry into the dauer stage. In this stage, a metabolic shift occurs that promotes the catabolism of existing energy stores through
anaerobic reactions in order to sustain development. Animals may exit dauer when environmental conditions, such as food availability, improve.

267
S. Rashid et al. Developmental Biology 475 (2021) 265–276

lipids, and amino acids to be rapidly degraded in daf-16 mutants, ulti- developmental arrest in C. elegans. This arrest may allow animals to
mately resulting in premature expiration compared to wild-type dauer temporarily slow or halt developmental processes until foraging allows
larvae (Penkov et al., 2020). A similar loss of energy stores is observed in them to identify food sources that satisfy their needs, which is likely not
dauer animals lacking AMPK, further suggesting daf-16 and AMPK may difficult given the diversity of their natural environment (Schulenberg,
work in concert to regulate long-term survival in dauer through meta- 2017).
bolic regulation (Narbonne and Roy, 2009). Recently, it has been shown
that the addition of ethanol can increase the duration of dauer survival by 2.5. Amino acids
allowing the animals to metabolize it as an energy source (Kaptan et al.,
2020). This supplementation induces a metabolic shift to improve sur- C. elegans require several amino acids in their diet, including the same
vival, and specifically prevents the deterioration of mitochondria nine amino acids essential in humans (histidine, lysine, tryptophan,
commonly seen in dauer animals. The mitochondria may thus serve as phenylalanine, methionine, threonine, leucine, isoleucine and valine), as
important metabolic regulators of survival during the dauer stage. well as arginine (Braeckman et al., 2009). The remaining amino acids can
Analysis of gene expression in dauer larvae indicate that various be synthesized de novo. Amino acids serve as the basic building blocks for
fermentation pathways are upregulated, suggesting that dauer larvae are proteins, but certain amino acids can be used for the synthesis of me-
able to utilize carbon sources for energy in the absence of oxygen, which tabolites which serve as energy sources in low-nutrient conditions (Kühnl
may be important for animals that do not feed and thus have reduced et al., 2005). Supplementation with amino acids can also extend the
internal oxygen (Wang and Kim, 2003; Holt and Riddle, 2003). Some lifespan of animals in a daf-16-dependent manner (Edwards et al., 2015).
nematode species are able to undergo anaerobic mitochondrial fermen- Conversely, lack of essential amino acids can have severe conse-
tation, converting phosphoenolpyruvate (PEP), a glycolytic intermediate, quences on C. elegans development. The branched amino acids, leucine,
into oxaloacetate (Komuniecki and Harris, 1995). Malate de- isoleucine, and valine, may be required for progression past the L2 larval
hydrogenases, or malic enzymes, can reversibly convert oxaloacetate to stage (Jia et al., 2016). Mutations in the branched-chain α-ketoacid de-
malate, which were upregulated in dauer larvae (McElwee et al., 2006). hydrogenase, part of the branched chain amino acid (BCAA) catabolic
Malate dismutation in anaerobic mitochondria may be one possible pathway, can cause developmental arrest as well as embryonic lethality
method of producing energy and maintaining redox balance in dauer (Jia et al., 2016). In addition to serving as protein building blocks, these
larvae (McElwee et al., 2006). Indeed, there is evidence that C. elegans amino acids are also required for the synthesis of certain lipids, and have
may utilize alternate, anaerobic metabolic pathways such as fumarate been implicated in metabolic disease in humans, suggesting that they
respiration, as rhodoquinone, a mitochondrial component required for have physiological roles beyond protein synthesis (Newgard, 2012).
anaerobic respiration is found in all stages of the C. elegans life cycle In order for animals to regulate developmental plasticity in response
(Takamiya et al., 1999). to nutrient signals, they must integrate nutrient sensing with downstream
regulation of metabolism and growth. In C. elegans, the amphid neurons
2.4. Essential nutrients play a role in amino acid sensing. Specifically, MGL-1 and MGL-2, both of
which are metabotrophic glutamate receptors that are expressed in the
C. elegans have a specific set of essential nutrients required amphids, are involved in food sensing, as loss of mgl-1 results in animals
throughout their lifespan, without which they will stop developing that do not delay reproduction even in the absence of food (Jeong and
entirely, or face numerous deleterious effects (Braeckman et al., 2008; Paik, 2017). Additionally, leucine-mediated rescue of starvation pheno-
Zecic et al., 2019) (Fig. 3). In their natural environment their diet consists types, such as stress resistance and increased longevity, is abolished
primarily of different bacterial species, although fungal, plant, and ani- when mgl-1 and mgl-2 are disrupted, suggesting that they play a role
mal matter has also been identified in their intestine (Schulenberg, specifically in amino acid sensing (Kang and Avery, 2009). Signals from
2017). This finding may indicate that a diet solely of bacteria is not these neural receptors likely get transduced to downstream signalling
sufficient for the animal’s dietary essentials. Indeed, media used in lab- pathways, which further regulate development in response to nutrient
oratories must also be supplemented with cholesterol in order to facili- signals. The TOR pathway is a primary candidate for this regulation, as it
tate proper growth, indicating that a diet that consists solely of E. coli is has a known role in mediating amino acid signals in other organisms
insufficient. The regulation of developmental rate by various metabolites (Jewell et al., 2013). In particular, Rag GTPases are likely involved, as
is likely mediated through insulin/IGF-1 and TOR signalling, as both of they activate the TOR pathway in the presence of amino acids (Sancak
these signalling pathways are responsible for sensing and responding to et al., 2008). Interestingly, a cocktail of amino acids can trigger devel-
nutrients, and have defined roles in developmental regulation (Iser and opment in quiescent animals undergoing L1 arrest, and this also occurs
Wolkow, 2007; Jia et al., 2004). However, there are instances where following hypodermal activation of Rag-TORC1, suggesting amino acid
metabolites can mediate C. elegans development independent of major signalling activates TOR to promote development (Fukuyama 2015).
signalling pathways (MacNeil et al., 2013). Nucleotides, for example, are Thus, developmental plasticity requires the integration of external
metabolites that are required for proper development, and germ cell environmental cues with an exquisitely controlled network of metabolic
proliferation is inhibited when nucleotide uptake is low by compro- modulators of developmental progression.
mising biosynthetic or scavenging pathways (Chi et al., 2016; Jia et al.,
2020). 2.6. Vitamin deficiencies
The minimal medium used for axenic growth of C. elegans contains
precise amounts of every metabolite that animals require for develop- In C. elegans, as in most organisms, vitamins act as cofactors and
ment (Szewczyk et al., 2003). However, animals generally grow slowly in donor molecules to catalyze a wide spectrum of metabolic reactions (Bito
this medium compared to traditional bacterial diets. The ‘typical’ et al., 2013). Many vitamins are essential for C. elegans development,
C. elegans life cycle has been defined primarily by its growth on labora- including riboflavin, thiamin, and vitamin B12 (Dougherty et al., 1959).
tory strains of E. coli, such as OP50 or HT115. In the natural world, Like amino acids, a deficiency of these essential vitamins can slow or
C. elegans development is highly plastic, with growth rates varying arrest development. However, the mechanisms by which development is
dramatically based on its diets (Samuel et al., 2016). This highlights the regulated by the presence or absence of vitamins remain largely un-
possibility that C. elegans can respond to natural metabolites in remark- known. Deficiencies of vitamin B2 result in a decrease of TOR activity in
ably different ways. Vitamin B12 found in diets of Comamonas aquaticas, animals, as well as a reduction of protease secretion in the intestine, a
for example, can alter gene expression and cause accelerated develop- process by which animals sense and avoid low quality food (Qi et al.,
mental rates and altered timing of egg-laying (Watson et al., 2014). 2017). Vitamin B12, as well as vitamin B9, or folate, act in the methio-
Conversely, bacteria lacking certain essential nutrients may induce nine/SAM cycle (Yilmaz and Walhout, 2014) and the reduction of folate

268
S. Rashid et al. Developmental Biology 475 (2021) 265–276

intake in C. elegans results in reproductive defects, as well as a shorter in terms of its major regulators. Both daf-16 and AMPK appear to play
lifespan (Austin et al., 2010). vital roles in both contexts, as loss of these factors lead to an inability to
Conversely, supplementation with vitamins can have significant ef- arrest, thus leading to decreased survival in starved L1 animals (Mu~ noz
fects on the physiology of C. elegans. Addition of low doses of folic acid and Riddle, 2003; Baugh and Sternberg, 2006; Narbonne and Roy, 2009;
can increase lifespan and delay aging through inhibition of both TOR and Baugh, 2013). L1 diapause is also regulated by a number of insulin-like
insulin signalling, suggesting that it may affect C. elegans developmental peptides, which are ligands that can serve as both agonists and antago-
rate as well (Rathor et al., 2015). Vitamin D can also affect longevity by nists for the insulin receptor DAF-2 (Pierce et al., 2001). Specifically,
activating stress response genes including skn-1/Nrf, which regulates ins-6, ins-4, and daf-28 appear to regulate L1 arrest in concert, as over-
oxidative stress responses, and ire-1/ERN1, which is required for the expression of these ILPs leads to reduced survival during L1 starvation, as
unfolded protein response (Mark et al., 2016). Dietary variation in animals continue to grow despite the lack of nutrients (Chen and Baugh,
Comamonas aquaticas lead to an increased developmental rate through 2014). Conversely, the loss of these ILPs leads to increased survival in the
the regulation of gene expression that is independent of TOR and insulin L1 diapause. Interestingly, these ILPs appear to play a role in regulating
signalling (MacNeil et al., 2013). Specifically, this effect is induced by development by facilitating communication between neurons and the
Vitamin B12 via its role as a cofactor in the methionine/SAM cycle intestine (Hung et al., 2014), suggesting that intestinal cells are crucial
(Watson et al., 2014). This suggests that the supply of certain vitamins for relaying information in order to appropriately tune developmental
may limit the growth rate of C. elegans, and faster rates of development progression to nutrient status.
may be more prevalent than that of the standard wild-type Bristol strain Notably, while the L1 diapause promotes survival in the short term
fed a laboratory-derived E. coli diet. It is, however, unclear whether a when faced with starvation, it also comes with several drawbacks,
faster developmental rate would confer a fitness advantage to C. elegans including damage to the gonad, as well as a delay in development
in a natural context. Given the number of development adjustments following recovery from the arrest (Lee et al., 2012). Indeed, while ani-
C. elegans likely undergoes in nature, most of which primarily involve the mals can survive for up to 21 days in this arrest, they display numerous
slowing or ceasing of development in order to improve survival (Frezal characteristics typical of aging, including mitochondrial fragmentation
and Felix, 2015), it is certainly advantageous to tune developmental rate and granulated tissues (Roux and Kenyon, 2016). However, many of
to the quality of food that is consumed, as a hard-wired rapid rate of these phenotypes are reversed upon feeding of L1 worms, though protein
development would oblige animals to increase nutrient intake to a level aggregation does persist even into adulthood.
that cannot be systematically accommodated by their natural Neuronal signals also impinge on survival during the L1 diapause, as
environments. survival was seen to increase in mutants lacking unc-31 function, which
plays a role in neuroendocrine signalling through ligands such as ILPs
2.7. L1 arrest (Lee and Ashrafi, 2008). Indeed, the mere sensing of food, without
ingestion, was sufficient to trigger gene expression changes in
The L1 arrest shares similarities with the dauer diapause, particularly L1-arrested larvae, including upregulation of the IIS pathway (Kaplan

Fig. 3. C. elegans have diverse nutritional needs. Various nutrients, such as vitamins, heme, and amino acids, are required for optimal growth of C. elegans.
Conversely, deficiencies in these nutrients can induce a variety of defects, from shortened lifespans, reduced reproductive capabilities, to developmental arrest.
Beneficial effects from nutrients are depicted in green, deleterious effects from a lack of those nutrients are in red.

269
S. Rashid et al. Developmental Biology 475 (2021) 265–276

et al., 2018). In particular, expression of the ILP daf-28, a DAF-2 agonist, stem cell population expands throughout and beyond the late L3 larval
was increased in the amphid neurons. This suggests external nutritional stage (Michaelson et al., 2010; Schindler et al., 2014), after which the
cues are transduced by neurons, leading to downstream effects that proximal-most germ cells become displaced outside the signalling range
prepare larvae for development in response to food availability. of the DTCs, resulting in a switch from mitotic divisions to the onset of
Coupling nutrient sensing with the maintenance of quiescence is meiosis (Austin and Kimble, 1987; Henderson et al., 1994; Nadarajan
likely crucial for prolonged survival of L1 arrested larvae, although the et al., 2009).
precise molecular mechanisms behind this have not been elucidated. In both the L1 diapause and the dauer stage, the germline stem cells
Regulation by small RNAs, such as microRNAs, may also contribute to undergo a G2/M phase cell cycle arrest (Narbonne and Roy, 2006;
survival of the starved L1 larvae. AIN-1 and AIN-2, components of the Ouellet et al., 2008; Seidel and Kimble, 2015) (Fig. 1) Interestingly, no
miRNA-induced silencing complexes (miRISC), are required for L1 sur- germ cell proliferation occurs throughout the dauer diapause despite
vival during the arrest (Zhang and Han, 2011). Indeed, miRNAs such as active Notch signalling. The C. elegans ortholog of the PTEN tumour
mir-71 promote survival during the L1 arrest survival through the inhi- suppressor, DAF-18 is required, not only for the formation of the dauer
bition of insulin receptor/PI3K pathway genes (Zhang et al., 2011), while larva, but also the extended survival conferred by this stage (Solari et al.,
the miRNA mir-235, the ortholog of mammalian mir-92, (Kasuga et al., 2004). In addition, the C. elegans orthologues of various components of
2013), is required for the quiescence of blast cells. Specifically, mir-235 the LKB1/AMPK signalling pathway were found to be critical to establish
targets nhr-91, a homolog of mammalian germ cell nuclear factor and/or maintain cell cycle quiescence in the dauer stage (Narbonne &
(GCNF), to suppress its expression during the L1 arrest and thus inhibit Roy, 2006, 2009). The loss of AMPK leads to supernumerary germ cells in
blast cell progression. Notably, mir-235 is negatively regulated by the IIS the dauer germ line, although the defect occurs during the execution of
pathway, suggesting its activity is also nutritionally controlled (Kasuga dauer, when animals are still feeding and nutrients are present, and not
et al., 2013). during the diapause per se.
The integrity of the germ cells is compromised in AMPK mutant dauer
2.8. AMPK, a major regulator of the starvation response larvae, and they therefore do not produce functional sperm or oocytes,
rendering the post-dauer mutants sterile (Kadekar and Roy, 2019). While
AMPK is a highly conserved enzyme that has numerous roles in wild type animals that transit through dauer display characteristic
regulating metabolism. AMPK’s function during periods of energy stress chromatin modifications and corresponding gene expression, animals
has been well characterized, particularly in C. elegans. Loss of AMPK that lack AMPK signalling misregulate both endogenous small RNA
further reduces the viability of animals that remain in the L1 diapause for pathways and their chromatin landscape in the germ line. These alter-
extended periods, suggesting that the kinase has an important role in ations impact gene expression in the dauer and post-dauer animals, and
managing the metabolic stress caused by acute starvation throughout the are likely the basis of the observed post-dauer sterility (Kadekar and Roy,
L1 diapause (Lee et al., 2012; Demoinet et al., 2017). 2019). For example, the expression levels of certain transcripts (ppk-2,
Conversely, animals lacking AMPK display numerous defects during pmk-1, spe-26, pro-2, mek-2) are altered in wild-type animals that transit
and after the dauer diapause (Narbonne and Roy, 2009; Kadekar and through the dauer stage compared to those animals that did not (Ow
Roy, 2019). AMPK mutant animals have greatly reduced survival in the et al., 2018). In animals that lack both catalytic subunits and hence all
dauer stage (Narbonne and Roy, 2009). Given the role of AMPK in AMPK signalling (aak(0) mutants), the expression of these five genes was
regulating enzyme levels (Penkov et al., 2020), the reduced dauer sur- dramatically misregulated compared to wild-type animals (Kadekar and
vival may be a result of metabolic misregulation that causes animals to be Roy, 2019). Since these incorrectly deposited chromatin marks persist
unable to respond to the nutrient stresses of dauer. Loss of AMPK also into the post-dauer animals, the abnormal expression of these genes also
interferes with the ability of dauer animals to effectively utilize energy persists, likely disrupting a multitude of reproductive processes. Thus,
stores, a process that is crucial for extended dauer survival. Specifically, the changes that must take place in the germ line to adapt gene expres-
triglyceride stores are rapidly metabolized in AMPK dauer larvae, sug- sion to the energetic stress of the dauer stage are under the control of
gesting these animals have a much smaller reservoir of energy on which AMPK. Through its activation during the dauer diapause, it remodels the
to subsist (Narbonne and Roy, 2009). Those AMPK mutant larvae that do chromatin landscape to regulate germline quiescence and preserve germ
survive through dauer display numerous somatic defects, such as pro- cell integrity for the duration of the arrest, whether it will be 24 h or 24
truding or burst vulva, as well as dauer germline hyperplasia and days.
post-dauer sterility (Kadekar and Roy, 2019).
3.2. Germline quiescence during the L1 arrest
3. Protecting the germ line during acute nutrient stress
During the L1 arrest, larvae adjust their germline development,
3.1. Germline quiescence during the dauer stage instructing the PGCs to undergo a reversible arrest until a nutrient con-
tingency is met. A fusion protein GFP:histone-H2A, which marks all
The capacity of C. elegans to exercise its inherent developmental chromosomes in the early embryonic cells, revealed that the embryonic
plasticity often manifests in various contexts where animals are obliged germline precursor cells Z2 and Z3 undergo G2 cell cycle arrest
to adapt to starvation, such as by altering their cell cycle and/or delaying (Fukuyama et al., 2006) (Fig. 1). These precursor cells condense their
aspects of tissue development or differentiation (Budirahardja and chromosomes before embryogenesis and remain condensed for the
G€onczy, 2009). A crucial process in this adaptation is the preservation of remainder of the L1 arrest. Conversely, somatic cells only condense their
totipotent germ cells during extended periods of nutrient deprivation in chromosomes briefly during mitosis (Fukuyama et al., 2006). Moreover,
order to ensure that animals are able to reproduce once they resume immunofluorescence analysis of γ-tubulin revealed that the Z2 and Z3
reproductive development (Fukuyama, 2018). precursor cells contain two centrosomes in the starved L1 larvae rather
When the L1 larva hatches from the egg, it contains four primordial than a single centrosome in the continuously fed counterparts consistent
gonadal cells, two of which make up the somatic primordium, while the with a G2 and not a G1 arrest. When starved animals were recovered with
other two correspond to the primordial germ cells (PGCs) that will pro- a DNA synthesis inhibitor hydroxyurea, 98% of larvae contained more
liferate to generate the entire germ line (Hirsh et al., 1976). During than three PGCs, indicating that these cells were able to divide without
C. elegans development, germ cell proliferation is dependent on activa- any requirement for S phase (Fukuyama et al., 2006).
tion of the Notch receptor GLP-1, which is present on the germ cells, by The germ cell arrest that occurs during the L1 arrest stage is depen-
its Delta-like ligand LAG-2 and APX-1, which are expressed on the distal dent on the activity of daf-18/PTEN, a negative regulator of the insulin-
tip cells (Austin and Kimble, 1987; Nadarajan et al., 2009). This germline like signalling pathway (Fukuyama et al., 2006; Ogg and Ruvkun, 1998).

270
S. Rashid et al. Developmental Biology 475 (2021) 265–276

Genetic analysis reveals that DAF-18 functions downstream of age-1/- animals (Demoinet et al., 2017). Therefore, in response to acute starva-
PI3K and therefore partially bypasses the requirement for insulin-like tion, L1 diapause larvae tune their metabolic state through
signalling in order to respond to nutritional changes. In blast cells, AMPK-mediated adjustment, while the immortal germ line lineage is
cki-1, a cyclin-dependent kinase inhibitor is required for quiescence protected by at least three AMPK-dependent pathways: modulation of
during the L1 arrest (Fukuyama et al., 2003; Hong et al., 1998; Kostic TOR activity to block premature PGC divisions; regulation of the chro-
et al., 2003). In contrast to somatic cells, cki-1 is not required for germ matin modifying COMPASS-like complex; and regulation of the 14-3-3
cell arrest. Instead, germ cell arrest is dependent on the mitotic spindle protein PAR-5, to block the deposition of inappropriate epigenetic
checkpoint protein mdf-1/Mad1p, a downstream effector of DAF-18 and instructions.
AKT-1/Akt/PKB signalling (Watanabe et al., 2008).
The role of AMPK in blocking somatic cell divisions during the L1 3.3. Adult reproductive diapause
diapause was recently expanded to include the post-embryonic Q neu-
roblasts. These cells divide and also migrate in aak(0) mutants (Zheng While the dauer and L1 diapauses have been well characterized, the
et al., 2018). Therefore, the loss of AMPK allows a subset of somatic cells existence of ARD, a hypometabolic stage, has been debated since it was
to execute aspects of their post-embryonic developmental program first introduced approximately 10 years ago (Angelo and Van Gilst, 2009;
without satisfying a food signal contingency that normally ensures global Carranza-García & Navarro, 2020). Early and late L4 animals that
developmental quiescence. encounter starvation can enter the ARD by altering their somatic and
AMPK is required for germ cell arrest during the L1 diapause germline tissue. When L4 larval stage animals commit towards the
(Demoinet et al., 2017). It is also critical to block the aberrant chromatin reproductive adult stage and encounter starvation, they can execute the
modifications that occur in response to starvation. Animals that lack all ARD. Animals can spend up to 30 days in this state without compro-
AMPK signalling (aak(0)) die prematurely in the L1 diapause, but if they mising their adult lifespan (Angelo and Van Gilst, 2009). During this
are recovered before they expire, most of these animals will become period, the gonad undergoes shrinkage. While the majority of somatic
sterile adults (~70% of the population), while those individuals cells continue to undergo cell division and endoreduplication without
(remaining 30%) that are fertile produce few progeny (Demoinet et al., disruption, vulval development is arrested (Angelo and Van Gilst, 2009;
2017). In L1 arrested aak(0) mutants, the activating chromatin mark Seidel and Kimble, 2011). Therefore, because of this defect, entry into the
H3K4me3 accumulates abnormally in the PGCs and persists. In fact, the ARD stage is optimal at the early L4 larval stage, before any embryos
marks continue to remain elevated in subsequent generations that were begin to develop. Should the animals enter ARD at the late L4 larval
never starved. Instead they accumulate and presumably cause atypical stage, animals will produce embryos that are retained within the uterus,
germline gene expression, consequently disrupting germ cell integrity. resulting in internal hatching and death of the mother (Seidel and Kim-
The loss of AMPK in this context results in reproductive defects, not only ble, 2011).
in starved animals, but also their non-starved progeny, revealing a The germ line undergoes drastic changes during ARD. After three
transgenerational effect likely conferred through the accumulation of the days of starvation in wild-type animals, the number of the germ cells
ectopic histone modifications. This transgenerational epigenetic conse- decreases from 150 to 30 germ cells per arm, causing the gonad to shrink
quence of developmental plasticity that is revealed by the loss of AMPK (Angelo and Van Gilst, 2009; Seidel and Kimble, 2011). In wild-type
may represent a more common evolutionary phenomenon whereby or- animals, starvation affects the number of oocytes produced. In replete
ganisms prime not only themselves, but also future generations, for conditions, each gonad arm can possess six to ten oocytes with ovulation
potentially limiting nutrient conditions (Heijmans et al., 2008). occurring once every 23 min. In ARD animals, each gonad arm only holds
The compromise of components of the COMPASS-like complex during one oocyte and the rate of ovulation decreases to once every 8 h (Seidel
the L1 diapause is sufficient to partially suppress both the sterility and the and Kimble, 2011). By the tenth day, embryo production ceases.
reduced brood size in aak(0) mutants (Demoinet et al., 2017). In Starvation also alters the cell cycle of the germ cells. In the absence of
C. elegans, the COMPASS-like complex catalyzes the trimethylation of food, the mitotic germ cell cycle becomes quiescent (Angelo and Van
H3K4. RBR-2 is a histone demethylase that catalyzes the demethylation Gilst, 2009). The cell cycle slows and eventually most germ cells arrest at
of H3K4 (Mariani et al., 2016). Based on similarities in somatic defects the G2 phase. In young adults, the germ cells arrest their cell cycle after
and reduced brood sizes between aak(0) and rbr-2 mutants, the mis- 30 min of starvation, while L4 larval stage animals do not halt their cell
regulation of the levels of H3K4me3 could be a cause of sterility in aak(0) cycle immediately. Instead, they will moult into adulthood then slow
mutants (Demoinet et al., 2017). The compromise of any one of the down and arrest their mitotic germ cell cycle (Seidel and Kimble, 2015).
COMPASS-like complex components during the L1 diapause was suffi- Not surprisingly, starvation also affects the rate of meiosis in the germ
cient to restore the fertility and correct the reduced brood size. Inter- cells. The expression of GLD-1, a meiotic marker, revealed that the rate of
estingly, most of the somatic defects of starved aak(0) mutants were not meiosis in the gonad of the starved animals is lower than continuously
suppressed when treated with set-2 and set-16 RNAi, suggesting that these fed animals, suggesting that nutrient availability affects the development
defects could be due to an H3K4me3-independent pathway. Bio- of the germ cells (Seidel and Kimble, 2015). However, most of these
informatic analysis also indicates that all members of the COMPASS-like phenotypes are reversible upon the resumption of feeding.
complex components with the exception of DPY-30, a histone methyl- After spending five days in ARD, the mitotic index of germ cells,
transferase complex regulatory subunit, possess consensus AMPK phos- defined as the ratio of cells undergoing mitosis to the total number of
phorylation motifs. In the PGCs of starved L1 diapause mutants, the levels proliferative cells, is very low (Roy et al., 2016). Yet after only 1.5 h
of SET-2 were higher compared to starved wild-type PGCs, indicating following the resumption of feeding, the germ cells enter M phase and the
that AMPK could be involved in regulating the levels of SET-2 and mitotic index recovers substantially. Consequently, the DNA content
COMPASS-like complex elements in the PGC nuclei. The absence of matches the changes in the mitotic index. The DNA content is high in the
AMPK activity therefore results in an increase in H3K4me3 abundance first 24 h post-ARD, but by 48 h, there is a significant reduction in DNA
and inappropriate gene expression in the starved aak(0) mutant germ content that persists for the following four days, which correlates with
cells. the decline in sperm number. Refed or recovered animals return to their
H3K4me3 is most often associated with transcriptional start sites and original body size following ARD and regenerate a normal-sized gonad,
enhanced elongation (Lin et al., 2011). Interestingly, the aberrant tran- although the recovered animals do not have as many germ cells and
scriptional elongation that occurs as a result of the aberrant chromatin therefore produce approximately 70% of the brood size of their contin-
H3K4me3 levels may also contribute to the sterility in aak(0) animals, uously fed counterparts (Angelo and Van Gilst, 2009; Carranza-García
since treatment with a well-characterized transcription elongation in- and Navarro, 2019). Although germ cell proliferation and gonad size
hibitor partially restored fertility in the post-L1 diapause aak(0) mutant largely recover after ARD, this period of nutrient deprivation in ARD

271
S. Rashid et al. Developmental Biology 475 (2021) 265–276

dramatically affects overall germ cell numbers. a daf-12 mutant. Liganded DAF-12 promotes the transcription of the
There is debate regarding the molecular mechanisms that underlie let-7-related miRNA family, which in turn downregulates the expression
successful entry and exit from ARD. One group reported that the tran- of hbl-1/hunchback during the L2/L3 transition, allowing the animal to
scription factor nhr-49/HNF4 is responsible for the gonad size reduction execute the L3 larval stage gene expression program. Interestingly, these
during starvation and fertility recovery upon re-feeding (Angelo and Van let-7-related miRNAs negatively feed back on DAF-12. This relationship
Gilst, 2009; Carranza-García and Navarro, 2019). ced-3 mutants which illustrates how nuclear receptors and small RNAs couple physiological
are deficient in apoptosis could not undergo gonad shrinkage and did not and environmental events.
recover from ARD, suggesting that germline apoptosis was critical for Similar to traditional peptide hormones and their signalling path-
surviving this brief period of starvation (Angelo and Van Gilst, 2009). ways, small RNAs are emerging as beacons of developmental and phys-
Later, another group reported that apoptosis is not required for recovery iological adaptation, tuning gene expression to match environmental
from ARD, suggesting that the gonad shrinkage is not dependent on change. Perhaps the most notable among these is the pioneer microRNA
germline apoptosis (Carranza-García and Navarro, 2019). Studies also lin-4, which is required for post-embryonic developmental timing (Lee
show that LIN-35/Rb, the C. elegans homolog of the retinoblastoma et al., 1993). When lin-4 is expressed in the late L1 larval stage, it binds to
protein which regulates starvation-induced germ cell apoptosis, is not the 30 UTR of lin-14 and lin-28 mRNA to suppress their expression. In lin-4
required for gonad shrinking or recovery from the ARD stage mutants, the L1 program is reiterated, resulting in seam cell defects,
(Lascarez-Lagunas et al., 2014). While ARD does not use any of the ca- absence of egg-laying structures, long body shape, and uncoordinated
nonical longevity pathways, autophagy, and fat metabolism for other movements in the adult animals. Thus, lin-4 is a small RNA that is crucial
forms of diapause, ARD requires the HLH-30/TFEB transcription factor for timely progression through the early larval stages. Although this
that also plays an important role in the L1 arrest through its ability to example demonstrates the importance of small RNAs in coordinating
regulate efficient entry and exit of this quiescent stage (Gerisch et al., cellular/developmental decisions that ultimately determine the entire
2020). developmental trajectory of the organism, a hormone can be generally
Proper RNA metabolism is required for ARD recovery. While germ defined as a molecule that is produced in one tissue and can exert its
cells undergo G2 cell cycle arrest, somatic cell divisions or endor- effects in either the same (autocrine) or another tissue(s) (paracrine),
eduplication are not postponed during prolonged starvation (Burnaev- often invoking some degree of cell non-autonomy.
skiy et al., 2018) (Fig. 1). cye-1 mutants, which are cyclin E-deficient, are As in the case with many peptide hormones, neurons have recently
able to pass through ARD normally, suggesting that cell cycle reactivation been placed at the centre of small RNA-mediated effects in C. elegans. It
in somatic tissue was not required for recovery. Three-week starved ARD has been demonstrated that double-stranded RNA (dsRNA) expressed in
animals that were treated with a thymidylate synthase inhibitor 5-fluo- the neurons can travel to other somatic tissues as well as the germ line to
ro-20 -deoxyuridine (FUDR) were unable to restore their body and gonad silence its target(s) (Devanapally et al., 2015). Fluorescently labelled
size, normal pumping rate, or thrashing after ARD, suggesting that dsRNA that is injected into the body cavity accumulates within the oo-
nucleic acid metabolism is critical for ARD recovery. To examine if cytes likely through the non-specific import of material from the body
mitochondrial DNA synthesis was the underlying cause for the defects cavity. These cell non-autonomous acting dsRNAs are known as mobile
seen in FUDR-treated recovering ARD animals, polg-1 mutants, which are RNAs, and the silencing that they trigger in the germ line relies on sec-
deficient for mitochondrial DNA polymerase, were subjected to ARD and ondary RNAs produced by RDE-1 processing, the secondary nuclear
allowed to recover. These mutants recovered normally and had an un- Argonuate HRDE-1, and the RNase D homolog MUT-7. Thus, extracel-
altered mitochondrial DNA content, suggesting that the recovery from lular dsRNA produced in the neurons could be taken up and processed
ARD does not require mitochondrial DNA synthesis in the soma. To assess within the parental germ line where they can elicit robust gene silencing
if RNA metabolism is a requirement for ARD recovery, animals were and transgenerational inheritance.
treated with FUDR and specific nucleotides were supplemented into the Endogenous siRNAs of neuronal origin may also produce a heritable
growth media. Only supplementation of uracil, uridine, and deoxyur- response to regulate behaviour in subsequent generations through the
idine were able to reverse the defects associated with FUDR treatment. modification of gene expression in the germ line in a manner that is
The type of RNA molecules that are important for morphological and dependent on the activity of the nuclear Argonaute proteins HRDE-1 and
functional restoration during the recovery from ARD still remain CSR-1 (Posner et al., 2019). The transmission of these endogenous siR-
uncharacterized. NAs is dependent on RDE-4, a neuron-enriched small RNA binding pro-
tein and was previously shown to play a role in neuronal migration,
4. Coordinating developmental and physiological adaptation learning, and memory (Juang et al., 2013; Sims et al., 2016; Tonkin and
during situations of developmental plasticity Bass, 2003). Although it was not specifically shown, findings from this
group imply that neuronal-derived small RNAs travel to the germ line.
Hormones enable communication between tissues to regulate physi- Small RNAs produced in animals expressing RDE-4 exclusively in the
ology and behaviour in many organisms. They play a key role in regu- neurons can affect gene expression cell non-autonomously in affected
lating development in response to nutritional stress to ensure the survival progeny for up to three generations, underscoring an adaptive means of
of the organism. As previously mentioned, dauer formation is triggered communicating information that is sensed by the neurons and trans-
by reduced levels of a bile acid-like hormone Dafachronic acid, resulting mitted, quite far from the source of their biosynthesis, to the germ line.
in a number of somatic and germline changes that include global cell However, despite the excitement generated by this possible link between
cycle and developmental arrest that are mediated by the nuclear receptor the neurons and the germ line, it is important to note that the authors did
called DAF-12 (Antebi et al., 2000). not show that RDE-4-dependent neuronal small RNAs have homology to
In addition to regulating dauer entry and co-regulating the dauer the germline genes that were differentially expressed in the presence of
metabolic transition, DAF-12 also functions as a ligand-regulated miRNA neuronal RDE-4.
switch in response to nutrient availability (Shen et al., 2012). Some Perhaps even more intriguing, small RNAs have recently been shown
daf-12 mutants display a heterochronic delay of the third larval stage to reinforce learning avoidance behaviours in a transgenerational
program and reiterate the L2 seam cell division programs (Antebi et al., manner. C. elegans consume a variety of bacterial food sources in the
2000; Hammell et al., 2009). In the daf-12 mutant, the expression levels environment. C. elegans that have consumed Pseudomonas aeruginosa, a
of the let-7-related miRNAs mir-48, mir-84, and mir-241 are reduced, pathogenic strain known to cause disease in plants and animals, develop
suggesting that active DAF-12 functions as a positive regulator of these an avoidance behaviour within 4 h of consuming the bacteria (Moore
miRNAs. Consistent with these findings, mutants with a triple knockout et al., 2019). This learned behaviour is inherited up to four generations
of these let-7-related miRNAs repeat the L2 seam cell division, resembling through the germ line, despite the previous generations never

272
S. Rashid et al. Developmental Biology 475 (2021) 265–276

5. Conclusion

The natural environment of any organism can pose numerous, often


unpredictable challenges to health and survival. Developmental plas-
ticity is thus crucial for organisms to be able to adapt and maximize their
fitness. This is particularly true for C. elegans, and we have highlighted
the potential of this animal to adapt to nutrient stress at various points of
their life cycle. This adaptation may be as simple as slowing growth when
faced with a dearth of essential nutrients, as seen in animals grown on
axenic ‘minimal media’ (Szewczyk et al., 2003), or even accelerating
development upon encountering excess quantities of specific metabo-
lites, as is the case with animals fed Vitamin B12 (MacNeil et al., 2013).
Alternatively, developmental plasticity can involve more drastic
shifts, such as a developmental arrest in larvae that hatch in the absence
of food, or the thoroughly studied dauer diapause, where animals couple
nutrient signals with dramatic changes in gene expression, ultimately
resulting in altered metabolism, morphology, and even behaviour. The
dauer stage in particular highlights the extent to which developmental
plasticity can benefit survival. By entering the dauer diapause, animals
can survive environmental stresses for extended periods, then resume
reproductive development only when conditions improve. This adapta-
tion not only benefits survival of individuals, but may also have led to
evolutionary advantages, such as allowing nematodes to evolve from soil-
dwelling bacterivores to infectious parasites (Crook, 2014).
We also attempted to highlight the extensive metabolic challenges
Fig. 4. Neuron-derived small RNAs play various roles in C. elegans. Small
faced by even simple organisms such as C. elegans when faced with acute
RNAs synthesized in the neurons can influence behaviour and somatic devel-
starvation. Lack of adequate food resources will trigger arrest at several
opment. Small RNAs can be inherited transgenerationally to influence
P. aeruginosa avoidance, chemotaxis behaviour, and proper L2/L3 larval stage
points during C. elegans development, highlighted by phenomena such as
transition. Small RNAs could potentially play a role in preserving germline the L1 diapause or the ARD. Even after development has ceased, drastic
quiescence and germ cell integrity during the dauer stage in AMPK animals. shifts in metabolic pathways must occur in order for animals to survive
the period of nutrient stress seen prominently in dauer larvae, which
prioritize fatty acid oxidation, gluconeogenesis, and fermentation path-
ways to provide them with the energy they require for survival. Aside
encountering P. aeruginosa. The progeny of the exposed mother express from severe caloric deprivation, lack of several key metabolites, such as
daf-7/TGFβ at significantly high levels in the ASI neurons, while daf-7 amino acids or vitamins, can limit the development of C. elegans through
mutants do not inherit this avoidance behaviour. Mutations that disrupt distinct mechanisms, highlighting the diverse and even “process-specific”
the nuclear RNAi pathway also perturb the normal aversion to nutritional needs that organisms require.
P. aeruginosa, suggesting that small RNA-mediated epigenetic adaptation Developmental plasticity in response to nutrient stress often includes
might be involved in learning and transmitting this avoidance behaviour. cell cycle quiescence in animals, including the germ cells. Maintaining
RNA-sequencing revealed that microRNAs and piRNAs are significantly the integrity of the germ line is crucial, as the totipotent nature of the
altered upon exposure to P. aeruginosa. Mutants that lack prg-1, the germ cells must be preserved so that following the arrest, fertilization
C. elegans ortholog of the PIWI Argonaute, display a naïve choice pref- will trigger appropriate embryonic and post-embryonic developmental
erence and normal pathogenic avoidance after exposure, however, this progression. AMPK is a notable mediator of quiescence in the germ line,
avoidance behaviour is not inherited. Moreover, exposure to the purified and is required for the survival and post-arrest fertility in both the L1 and
small RNAs isolated from P. aeruginosa is sufficient to induce an avoid- dauer diapauses through interactions with a number of different signal-
ance behaviour (Kaletskey et al., 2020). Further analysis shows that a ling pathways and mechanisms, highlighting the complexity of gene
single non-coding RNA, P11, that targets the C. elegans maco-1 is regulation during these developmental arrests (Narbonne and Roy, 2006,
required. In addition to the requirement of intact RNAi and piRNA 2009; Demoinet et al., 2017). Of particular interest is the idea that AMPK
pathways, the ASI neuron and the germ line are required for transmitting signalling may be mediated partly through small RNAs (Kadekar and
this transgenerational inheritance behaviour. These data are consistent Roy, 2019). Indeed, emerging data indicate that small RNAs play a
with a regulatory role of neuronally-derived small RNAs that are trans- crucial role as hormone-like signals throughout development. Small
mitted to future generations through the germ line to affect learned RNAs, particularly those produced in the neurons, may also act trans-
pathogenic avoidance behaviour in order to identify nutritious food generationally to regulate behaviour, and likely many other aspects of
sources, although how these small RNAs are packaged and expedited to physiology. How these small RNAs toggle between mounting an imme-
their final destinations remains undetermined. diate versus a long-term transgenerational adaptive response should help
Like hormones, small RNAs coordinate development among different us to understand how developmental plasticity may at times confer an
tissues according to both developmental and environmental cues (Fig. 4). advantage to the individual, while at other times confer a benefit to the
Although many have unique functions in development, like many hor- population as a whole.
mones, their effects depend greatly on their downstream effectors and/or
the response of their target tissues. However, although hormones elicit Acknowledgements
changes within the organism to adjust to immediate environmental sig-
nals, the effects of small RNAs can be both immediate or they can be long- We thank Sider Penkov for valuable insights and discussion con-
term, even transgenerational, adjusting gene expression to enhance the cerning many of these ideas. RR is supported by a project grant from the
survival of progeny over multiple generations. Canadian Institutes for Health Research (CIHR).

273
S. Rashid et al. Developmental Biology 475 (2021) 265–276

References Developmental Strategies. Diversity and Commonality in Animals. Springer, Tokyo.


https://doi.org/10.1007/978-4-431-56609-0_5.
Fukuyama, M., Gendreau, S.B., Derry, W.B., Rothman, J.H., 2003. Essential embryonic
Angelo, G., Van Gilst, M.R., 2009. Starvation protects germline stem cells and extends
roles of the CKI-1 cyclin-dependent kinase inhibitor in cell-cycle exit and
reproductive longevity in C. elegans. Science 326 (5955), 954–958. https://doi.org/
morphogenesis in C. elegans. Dev. Biol. 260 (1), 273–286. https://doi.org/10.1016/
10.1126/science.1178343.
s0012-1606(03)00239-2.
Antebi, A., Yeh, W.H., Tait, D., Hedgecock, E.M., Riddle, D.L., 2000. daf-12 encodes a
Fukuyama, M., Rougvie, A.E., Rothman, J.H., 2006. C. elegans DAF-18/PTEN mediates
nuclear receptor that regulates the dauer diapause and developmental age in
nutrient-dependent arrest of cell cycle and growth in the germline. Curr. Biol. 16 (8),
C. elegans. Genes Dev. 14 (12), 1512–1527.
773–779. https://doi.org/10.1016/j.cub.2006.02.073.
Austin, J., Kimble, J., 1987. glp-1 is required in the germ line for regulation of the
Fukuyama, M., Kontani, K., Katada, T., Rougvie, A.E., 2015. The C. elegans hypodermis
decision between mitosis and meiosis in C. elegans. Cell 51 (4), 589–599. https://
couples progenitor cell quiescence to the dietary state. Curr. Biol. 25 (9), 1241–1248.
doi.org/10.1016/0092-8674(87)90128-0.
https://doi.org/10.1016/j.cub.2015.03.016.
Austin, M.U., Liau, W.-S., Balamurugan, K., Ashokkumar, B., Said, H.M., LaMunyon, C.W.,
Gerisch, B., Tharyan, R.G., Mak, J., Denzel, S.I., Popkes-van Oepen, T., Henn, N.,
2010. Knockout of the folate transporter folt-1 causes germline and somatic defects in
Antebi, A., 2020. HLH-30/TFEB is a master regulator of reproductive quiescence.
C. elegans. BMC Dev. Biol. 10 (1), 46. https://doi.org/10.1186/1471-213x-10-46.
Dev. Cell 53 (3), 316–329. https://doi.org/10.1016/j.devcel.2020.03.014 e5.
Baugh, L.R., 2013. To grow or not to grow: nutritional control of development during
Golden, J.W., Riddle, D.L., 1982. A pheromone influences larval development in the
Caenorhabditis elegans L1 arrest. Genetics 194 (3), 539–555. https://doi.org/
nematode Caenorhabditis elegans. Science 218 (4572), 578–580.
10.1534/genetics.113.150847.
Golden, J.W., Riddle, D.L., 1984. A pheromone-induced developmental switch in
Baugh, L.R., Sternberg, P.W., 2006. DAF-16/FOXO regulates transcription of cki-1/Cip/
Caenorhabditis elegans: temperature-sensitive mutants reveal a wild-type temperature-
Kip and repression of lin-4 during C. elegans L1 arrest. Curr. Biol. 16 (8), 780–785.
dependent process. Proc. Natl. Acad. Sci. U.S.A. 81 (3), 819–823.
https://doi.org/10.1016/j.cub.2006.03.021.
Greenwald, I.S., Sternberg, P.W., Horvitz, H.R., 1983. The lin-12 locus specifies cell fates
Birnby, D.A., Link, E.M., Vowels, J.J., Tian, H., Colacurcio, P.L., Thomas, J.H., 2000.
in Caenorhabditis elegans. Cell 34 (2), 435–444. https://doi.org/10.1016/0092-
A transmembrane guanylyl cyclase (DAF-11) and Hsp90 (DAF-21) regulate a
8674(83)90377-x.
common set of chemosensory behaviors in Caenorhabditis elegans. Genetics 155 (1),
Hall, S.E., Beverly, M., Russ, C., Nusbaum, C., Sengupta, P., 2010. A cellular memory of
85–104.
developmental history generates phenotypic diversity in C. elegans. Curr. Biol. : CB 20
Bito, T., Matsunaga, Y., Yabuta, Y., Kawano, T., Watanabe, F., 2013. Vitamin B12
(2), 149–155. https://doi.org/10.1016/j.cub.2009.11.035.
deficiency in Caenorhabditis elegans results in loss of fertility, extended life cycle, and
Hammell, C.M., Karp, X., Ambros, V., 2009. A feedback circuit involving let-7-family
reduced lifespan. FEBS open bio 3, 112–117. https://doi.org/10.1016/
miRNAs and DAF-12 integrates environmental signals and developmental timing in
j.fob.2013.01.008.
Caenorhabditis elegans. Proc. Natl. Acad. Sci. U. S. A. 106 (44), 18668–18673. https://
Blackwell, T.K., Sewell, A.K., Wu, Z., Han, M., 2019. TOR signalling in Caenorhabditis
doi.org/10.1073/pnas.0908131106.
elegans. development, metabolism, and aging. Genetics 213 (2), 329–360. https://
Harvey, S.C., Gemmill, A.W., Read, A.F., Viney, M.E., 2000. The control of morph
doi.org/10.1534/genetics.119.302504.
development in the parasitic nematode Strongyloides ratti. Proceedings: Biol. Sci. 267
Braeckman, B.P., 2009. Intermediary metabolism. In: WormBook, pp. 1–24. https://
(1457), 2057–2063. https://doi.org/10.1098/rspb.2000.1249.
doi.org/10.1895/wormbook.1.146.1.
Hawdon, J.M., Schad, G.A., 1991. Albumin and a dialyzable serum factor stimulate
Braendle, C., Felix, M.A., 2008. Plasticity and errors of a robust developmental system in
feeding in vitro by third-stage larvae of the canine hookworm Ancylostoma caninum.
different environments. Dev. Cell 15 (5), 714–724. https://doi.org/10.1016/
J. Parasitol. 77 (4), 587–591.
j.devcel.2008.09.011.
Heijmans, B.T., Tobi, E.W., Stein, A.D., Putter, H., Blauw, G.J., Susser, E.S.,
Budirahardja, Y., G€ onczy, P., 2009. Coupling the cell cycle to development.
Slagboom, P.E., Lumey, L.H., 2008. Persistent epigenetic differences associated with
J. Development 136 (17), 2861–2872. https://doi.org/10.1242/dev.021931.
prenatal exposure to famine in humans. Proc. Natl. Acad. Sci. U.S.A. 105 (44),
Burnaevskiy, N., Chen, S., Mailig, M., Reynolds, A., Karanth, S., Mendenhall, A.,
17046–17049. https://doi.org/10.1073/pnas.0806560105.
Kaeberlein, M., 2018. Reactivation of RNA metabolism underlies somatic restoration
Henderson, S.T., Gao, D., Lambie, E.J., Kimble, J., 1994. lag-2 may encode a signaling
after adult reproductive diapause in C. elegans. Elife 7, e36194. https://doi.org/
ligand for the GLP-1 and LIN-12 receptors of C. elegans. Development 120 (10),
10.7554/eLife.36194.
2913–2924.
Carranza-García, E., Navarro, R.E., 2019. Apoptosis contributes to protect germ cells from
Hibshman, J.D., Doan, A.E., Moore, B.T., Kaplan, R.E., Hung, A., Webster, A.K.,
the oogenic germline starvation response but is not essential for the gonad shrinking
Bhatt, D.P., Chitrakar, R., Hirschey, M.D., Baugh, L.R., 2017. daf-16/FoxO promotes
or recovery observed during adult reproductive diapause in C. elegans. PloS One 14
gluconeogenesis and trehalose synthesis during starvation to support survival. eLife
(6), e0218265. https://doi.org/10.1371/journal.pone.0218265.
6, e30057. https://doi.org/10.7554/eLife.30057.
Carranza-García, E., Navarro, R.E., 2020. Insights into the hypometabolic stage caused by
Hirsh, D., Oppenheim, D., Klass, M., 1976. Development of the reproductive system of
prolonged starvation in L4-adult Caenorhabditis elegans hermaphrodites. Front Cell
Caenorhabditis elegans. Dev. Biol. 49 (1), 200–219.
Dev. Biol. 8, 124. https://doi.org/10.3389/fcell.2020.00124.
Holt, S.J., Riddle, D.L., 2003. SAGE surveys C. elegans carbohydrate metabolism: evidence
Cassada, R.C., Russell, R.L., 1975. The dauerlarva, a post-embryonic developmental
for an anaerobic shift in the long-lived dauer larva. Mech. Ageing Develop. 124 (7),
variant of the nematode Caenorhabditis elegans. Dev. Biol. 46 (2), 326–342. https://
779–800. https://doi.org/10.1016/s0047-6374(03)00132-5.
doi.org/10.1016/0012-1606(75)90109-8.
Hong, Y., Roy, R., Ambros, V., 1998. Developmental regulation of a cyclin-dependent
Chen, Y., Baugh, L.R., 2014. ins-4 and daf-28 function redundantly to regulate C. elegans
kinase inhibitor controls postembryonic cell cycle progression in Caenorhabditis
L1 arrest. Dev. Biol. 394 (2), 314–326. https://doi.org/10.1016/
elegans. Development 125 (18), 3585–3597.
j.ydbio.2014.08.002.
Hotez, P., Hawdon, J., Schad, G.A., 1993. Hookworm larval infectivity, arrest and
Chi, C., Ronai, D., Than, M.T., Walker, C.J., Sewell, A.K., Han, M., 2016. Nucleotide levels
amphiparatenesis: the Caenorhabditis elegans Daf-c paradigm. Parasitol. today 9 (1),
regulate germline proliferation through modulating GLP-1/Notch signaling in
23–26. https://doi.org/10.1016/0169-4758(93)90159-d.
C. elegans. Genes Dev. 30 (3), 307–320. https://doi.org/10.1101/gad.275107.115.
Hu, Patrick J., 2007. Dauer. In: WormBook, pp. 1–19. https://doi.org/10.1895/
Crook, M., 2014. The dauer hypothesis and the evolution of parasitism: 20 years on and
wormbook.1.144.1.
still going strong. Int. J. Parasitol. 44 (1), 1–8. https://doi.org/10.1016/
Hung, W.L., Wang, Y., Chitturi, J., Zhen, M., 2014. A Caenorhabditis elegans
j.ijpara.2013.08.004.
developmental decision requires insulin signaling-mediated neuron-intestine
Demoinet, E., Li, S., Roy, R., 2017. AMPK blocks starvation-inducible transgenerational
communication. Development 141 (8), 1767–1779. https://doi.org/10.1242/
defects in Caenorhabditis elegans. Proc. Natl. Acad. Sci. U.S.A. 114 (13),
dev.103846.
E2689–E2698. https://doi.org/10.1073/pnas.1616171114.
Iser, W.B., Wolkow, C.A., 2007. DAF-2/Insulin-Like signalling in C. elegans modifies
Devanapally, S., Ravikumar, S., Jose, A.M., 2015. Double-stranded RNA made in
effects of dietary restriction and nutrient stress on aging, stress and growth. PloS One
C. elegans neurons can enter the germline and cause transgenerational gene silencing.
2 (11), e1240. https://doi.org/10.1371/journal.pone.0001240.
Proc. Natl/ Acad. Sci. U. S. A. 112 (7), 2133–2138. https://doi.org/10.1073/
Jeong, H., Paik, Y.-K., 2017. MGL-1 on AIY neurons translates starvation to reproductive
pnas.1423333112.
plasticity via neuropeptide signalling in Caenorhabditis elegans. Dev. Biol. 430 (1),
Dougherty, E.C., 1959. Introduction to axenic culture of invertebrate metazoan: a goal.
80–89.
Ann. N. Y. Acad. Sci. 77, 27–54. https://doi.org/10.1111/j.1749-
Jeong, P.Y., Kwon, M.S., Joo, H.J., Paik, Y.K., 2009. Molecular time-course and the
6632.1959.tb36891.x.
metabolic basis of entry into dauer in Caenorhabditis elegans. PloS One 4 (1), e4162.
Edwards, C., Canfield, J., Copes, N., Brito, A., Rehan, M., Lipps, D., Brunquell, J.,
https://doi.org/10.1371/journal.pone.0004162.
Westerheide, S.D., Bradshaw, P.C., 2015. Mechanisms of amino acid-mediated
Jewell, J.L., Russell, R.C., Guan, K.L., 2013. Amino acid signalling upstream of mTOR.
lifespan extension in Caenorhabditis elegans. BMC Genet. 16 (1), 8. https://doi.org/
Nat. Rev. Mol. Cell Biol. 14 (3), 133–139. https://doi.org/10.1038/nrm3522.
10.1186/s12863-015-0167-2.
Jia, K., Chen, D., Riddle, D.L., 2004. The TOR pathway interacts with the insulin
F
elix, M.-A., Braendle, C., 2010. The natural history of Caenorhabditis elegans. Curr. Biol.
signalling pathway to regulate C. elegans larval development, metabolism and life
20 (2), R965–R969. https://doi.org/10.1016/j.cub.2010.09.050.
span. Development (Cambridge, England) 131 (16), 3897–3906. https://doi.org/
Fielenbach, N., Antebi, A., 2008. C. elegans dauer formation and the molecular basis of
10.1242/dev.01255.
plasticity. Genes Dev. 22 (16), 2149–2165. https://doi.org/10.1101/gad.1701508.
Jia, F., Cui, M., Than, M.T., Han, M., 2016. Developmental defects of Caenorhabditis
Frezal, L., F
elix, M.A., 2015. C. elegans outside the Petri dish. eLife 4, e05849. https://
elegans lacking branched-chain α-ketoacid dehydrogenase are mainly caused by
doi.org/10.7554/eLife.05849.
monomethyl branched-chain fatty acid deficiency. J. Biol. Chem. 291 (6),
Friedman, D.B., Johnson, T.E., 1988. A mutation in the age-1 gene in Caenorhabditis
2967–2973. https://doi.org/10.1074/jbc.M115.676650.
elegans lengthens life and reduces hermaphrodite fertility. Genetics 118 (1), 75–86.
Jia, F., Chi, C., Han, M., 2020. Regulation of nucleotide metabolism and germline
Fukuyama, M., 2018. Nutritional control of the germline development in Caenorhabditis
proliferation in response to nucleotide imbalance and genotoxic stresses by EndoU
elegans. In: Kobayashi, K., Kitano, T., Iwao, Y., Kondo, M. (Eds.), Reproductive and

274
S. Rashid et al. Developmental Biology 475 (2021) 265–276

nuclease. Cell Rep. 30 (6), 1848–1861. https://doi.org/10.1016/ Michaelson, D., Korta, D.Z., Capua, Y., Hubbard, E.J., 2010. Insulin signalling promotes
j.celrep.2020.01.050 e5. germline proliferation in C. elegans. Development 137 (4), 671–680. https://doi.org/
Johnstone, I.L., Barry, J.D., 1996. Temporal reiteration of a precise gene expression 10.1242/dev.042523.
pattern during nematode development. EMBO J. 15 (14), 3633–3639. Moore, R.S., Kaletsky, R., Murphy, C.T., 2019. Piwi/PRG-1 Argonaute TGF-β mediate
Jones, S.J., Riddle, D.L., Pouzyrev, A.T., Velculescu, V.E., Hillier, L., Eddy, S.R., transgenerational learned pathogenic avoidance. Cell 177 (7), 1827–1841. https://
Stricklin, S.L., Baillie, D.L., Waterston, R., Marra, M.A., 2001. Changes in gene doi.org/10.1016/j.cell.2019.05.024 e1812.
expression associated with developmental arrest and longevity in Caenorhabditis Motola, D.L., Cummins, C.L., Rottiers, V., Sharma, K.K., Li, T., Li, Y., Suino-Powell, K.,
elegans. Genome Res. 11 (8), 1346–1352. Xu, H.E., Auchus, R.J., Antebi, A., Mangelsdorf, D.J., 2006. Identification of ligands
Juang, B.T., Gu, C., Starnes, L., Palladino, F., Goga, A., Kennedy, S., L’Etoile, N.D., 2013. for DAF-12 that govern dauer formation and reproduction in C. elegans. Cell 124 (6),
Endogenous nuclear RNAi mediates behavioral adaptation to odor. Cell 154 (5), 1209–1223. https://doi.org/10.1016/j.cell.2006.01.037.
1010–1022. https://doi.org/10.1016/j.cell.2013.08.006. Mu~ noz, M.J., Riddle, D.L., 2003. Positive selection of Caenorhabditis elegans mutants with
Kadekar, P., Roy, R., 2019. AMPK regulates germline stem cell quiescence and integrity increased stress resistance and longevity. Genetics 163 (1), 171–180.
through an endogenous small RNA pathway. PLoS Biol. 17 (6), e3000309 https:// Mylenko, M., Boland, S., Penkov, S., Sampaio, J.L., Lombardot, B., Vorkel, D.,
doi.org/10.1371/journal.pbio.3000309. Verbavatz, J.M., Kurzchalia, T.V., 2016. NADþ is a food component that promotes
Kaletsky, R., Moore, R.S., Vrla, G.D., Parsons, L.R., Gitai, Z., Murphy, C.T., 2020. exit from dauer diapause in Caenorhabditis elegans. PloS One 11 (12), e0167208.
C. elegans interprets bacterial non-coding RNAs to learn pathogenic avoidance. https://doi.org/10.1371/journal.pone.0167208.
Nature 586 (7829), 445–451. https://doi.org/10.1038/s41586-020-2699-5. Nadarajan, S., Govindan, J.A., McGovern, M., Hubbard, E.J., Greenstein, D., 2009. MSP
Kang, C., Avery, L., 2009. Systemic regulation of starvation response in Caenorhabditis and GLP-1/Notch signaling coordinately regulate actomyosin-dependent cytoplasmic
elegans. Genes Dev. 23 (1), 12–17. https://doi.org/10.1101/gad.1723409. streaming and oocyte growth in C. elegans. Development 136 (13), 2223–2234.
Kaplan, R., Webster, A.K., Chitrakar, R., Dent, J.A., Baugh, L.R., 2018. Food perception https://doi.org/10.1242/dev.034603.
without ingestion leads to metabolic changes and irreversible developmental arrest in Narbonne, P., Roy, R., 2006. Inhibition of germline proliferation during C. elegans dauer
C. elegans. BMC Biol. 16 (1), 112. https://doi.org/10.1186/s12915-018-0579-3. development requires PTEN, LKB1 and AMPK signalling. Development 133 (4),
Kaptan, D., et al., 2020. Exogenous ethanol induces a metabolic switch that prolongs the 611–619. https://doi.org/10.1242/dev.02232%JDevelopment.
survival of Caenorhabditis elegans dauer larva and enhances its resistance to Narbonne, P., Roy, R., 2009. Caenorhabditis elegans dauers need LKB1/AMPK to ration
desiccation. Aging cell 19 (10), e13214. https://doi.org/10.1111/acel.13214. lipid reserves and ensure long-term survival. Nature 457 (7226), 210–214. https://
Kasuga, H., Fukuyama, M., Kitazawa, A., Kontani, K., Katada, T., 2013. The microRNA doi.org/10.1038/nature07536.
miR-235 couples blast-cell quiescence to the nutritional state. Nature 497 (7450), Newgard, C.B., 2012. Interplay between lipids and branched-chain amino acids in
503–506. https://doi.org/10.1038/nature12117. development of insulin resistance. Cell Metabol. 15 (5), 606–614. https://doi.org/
Kimura, K.D., Tissenbaum, H.A., Liu, Y., Ruvkun, G., 1997. daf-2, an insulin receptor-like 10.1016/j.cmet.2012.01.024.
gene that regulates longevity and diapause in Caenorhabditis elegans. Science 277 O’Leary, J.K., Barry, J.P., Gabrielson, P.W., Rogers-Bennett, L., Potts, D.C., Palumbi, S.R.,
(5328), 942–946. Micheli, F., 2017. Calcifying algae maintain settlement cues to larval abalone
Komuniecki, R., Harris, B.G., 1995. Carbohydrate and energy metabolism in helminths. following algal exposure to extreme ocean acidification. Sci. Rep. 7 (1), 5774.
In: Marr, J.J., Muller, M. (Eds.), Biochemistry and Molecular Biology of Parasites. https://doi.org/10.1038/s41598-017-05502-x.
Academic Press, London, pp. 49–66. Ogg, S., Ruvkun, G., 1998. The C. elegansPTEN homolog, DAF-18, acts in the insulin
Kostic, I., Li, S., Roy, R., 2003. cki-1 links cell division and cell fate acquisition in the receptor-like metabolic signalling pathway. Mol. Cell. 2 (6), 887–893. https://
C. eleganssomatic gonad. Dev. Biol. 263 (2), 242–252. https://doi.org/10.1016/ doi.org/10.1016/s1097-2765(00)80303-2.
j.ydbio.2003.07.001. Ouellet, J., Li, S., Roy, R., 2008. Notch signalling is required for both dauer maintenance
Kühnl, J., Bobik, T., Procter, J.B., Burmeister, C., H€ oppner, J., Wilde, I., Lüersen, K., and recovery in C. elegans. Development 135 (15), 2583–2592. https://doi.org/
Torda, A.E., Walter, R.D., Liebau, E., 2005. Functional analysis of the methylmalonyl- 10.1242/dev.012435%JDevelopment.
CoA epimerase from Caenorhabditis elegans. FEBS J. 272 (6), 1465–1477. https:// Ow, M.C., Borziak, K., Nichitean, A.M., Dorus, S., Hall, S.E., 2018. Early experiences
doi.org/10.1111/j.1742-4658.2005.04579.x. mediate distinct adult gene expression and reproductive programs in Caenorhabditis
Lafuente, E., Beldade, P., 2019. Genomics of developmental plasticity in animals. Front. elegans. PLoS Genet. 14 (2), e1007219 https://doi.org/10.1371/
Genet. 10, 720. journal.pgen.1007219.
Lapierre, L.R., Hansen, M., 2012. Lessons from C. elegans: signalling pathways for Penkov, S., Raghuraman, B.K., Erkut, C., Oertel, J., Galli, R., Ackerman, E., Vorkel, D.,
longevity. Trends Endocrinol. Metabol.: TEM (Trends Endocrinol. Metab.) 23 (12), Verbavatz, J.M., Koch, E., Fahmy, K., Shevchenko, A., Kurzchalia, T.V., 2020.
637–644. https://doi.org/10.1016/j.tem.2012.07.007. A metabolic switch regulates the transition between growth and diapause in
L
ascarez-Lagunas, L.I., Silva-García, C.G., Dinkova, T.D., Navarro, R.E., 2014. LIN-35/Rb C. elegans. BMC Biol. 18 (1), 31. https://doi.org/10.1186/s12915-020-0760-3.
causes starvation-induced germ cell apoptosis via CED-9/Bcl2 downregulation in Pierce, S.B., Costa, M., Wisotzkey, R., Devadhar, S., Homburger, S.A., Buchman, A.R.,
Caenorhabditis elegans. Mol. Cell Biol. 34 (13), 2499–2516. https://doi.org/10.1128/ Ferguson, K.C., Heller, J., Platt, D.M., Pasquinelli, A.A., Liu, L.X., Doberstein, S.K.,
mcb.01532-13. Ruvkun, G., 2001. Regulation of DAF-2 receptor signalling by human insulin and ins-
Lee, B.H., Ashrafi, K., 2008. A TRPV channel modulates C. elegans neurosecretion, larval 1, a member of the unusually large and diverse C. elegans insulin gene family. Genes
starvation survival, and adult lifespan. PLoS Genet. 4 (10), e1000213 https:// Dev. 15 (6), 672–686. https://doi.org/10.1101/gad.867301.
doi.org/10.1371/journal.pgen.1000213. Posner, R., Toker, I.A., Antonova, O., Star, E., Anava, S., Azmon, E., Rechavi, O., 2019.
Lee, R.C., Feinbaum, R.L., Ambros, V., 1993. The C. elegans heterochronic gene lin-4 Neuronal small RNAs control behaviour transgenerationally. Cell 177 (7),
encodes small RNAs with antisense complementarity to lin-14. Cell 75 (5), 843–854. 1814–1826. https://doi.org/10.1016/j.cell.2019.04.029 e1815.
https://doi.org/10.1016/0092-8674(93)90529-y. Prudic, K.L., Jeon, C., Cao, H., Monteiro, A., 2011. Developmental plasticity in sexual
Lee, I., Hendrix, A., Kim, J., Yoshimoto, J., You, Y.J., 2012. Metabolic rate regulates L1 roles of butterfly species drives mutual sexual ornamentation. Science 331 (6013),
longevity in C. elegans. PloS One 7 (9), e44720. https://doi.org/10.1371/ 73–75. https://doi.org/10.1126/science.1197114.
journal.pone.0044720. Qi, B., Kniazeva, M., Han, M., 2017. A vitamin-B2-sensing mechanism that regulates gut
Lin, C., Garrett, A.S., De Kumar, B., Smith, E.R., Gogol, M., Seidel, C., Krumlauf, R., protease activity to impact animal’s food behaviour and growth. eLife 6, e26243.
Shilatifard, A., 2011. Dynamic transcriptional events in embryonic stem cells https://doi.org/10.7554/elife.26243.
mediated by the super elongation complex (SEC). Genes Dev. 25 (14), 1486–1498. Rao, A.U., Carta, L.K., Lesuisse, E., Hamza, I., 2005. Lack of heme synthesis in a free-living
https://doi.org/10.1101/gad.2059211. eukaryote. Proc. Natl. Acad. Sci. U.S.A. 102 (12), 4270–4275. https://doi.org/
Long, X., Spycher, C., Han, Z.S., Rose, A.M., Müller, F., Avruch, J., 2002. TOR deficiency 10.1073/pnas.0500877102.
in C. elegans causes developmental arrest and intestinal atrophy by inhibition of Rathor, L., Akhoon, B.A., Pandey, S., Srivastava, S., Pandey, R., 2015. Folic acid
mRNA translation. Curr. Biol. 12 (17), 1448–1461. https://doi.org/10.1016/s0960- supplementation at lower doses increases oxidative stress resistance and longevity in
9822(02)01091-6. Caenorhabditis elegans. Age 37 (6), 113. https://doi.org/10.1007/s11357-015-9850-
Ludewig, A.H., Kober-Eisermann, C., Weitzel, C., Bethke, A., Neubert, K., Gerisch, B., 5.
Hutter, H., Antebi, A., 2004. A novel nuclear receptor/coregulator complex controls Regniere, J., Powell, J., Bentz, B., Nealis, V., 2012. Effects of temperature on
C. elegans lipid metabolism, larval development, and aging. Genes Dev. 18 (17), development, survival and reproduction of insects: experimental design, data analysis
2120–2133. https://doi.org/10.1101/gad.312604. and modeling. J. Insect Physiol. 58 (5), 634–647. https://doi.org/10.1016/
MacNeil, L.T., Watson, E., Arda, H.E., Zhu, L.J., Walhout, A.J., 2013. Diet-induced j.jinsphys.2012.01.010.
developmental acceleration independent of TOR and insulin in C. elegans. Cell 153 Ren, Y., Shen, H.M., 2019. Critical role of AMPK in redox regulation under glucose
(1), 240–252. https://doi.org/10.1016/j.cell.2013.02.049. starvation. Redox Biol. 25, 101154. https://doi.org/10.1016/j.redox.2019.101154.
Mariani, L., Lussi, Y.C., Vandamme, J., Riveiro, A., Salcini, A.E., 2016. The H3K4me3/2 Ren, P., Lim, C.S., Johnsen, R., Albert, P.S., Pilgrim, D., Riddle, D.L., 1996. Control of
histone demethylase RBR-2 controls axon guidance by repressing the actin- C. elegans larval development by neuronal expression of a TGF-beta homolog. Science
remodeling gene wsp-1. Development 143 (5), 851–863. https://doi.org/10.1242/ 274 (5291), 1389–1391.
dev.132985%JDevelopment. Riddle, D.L., Albert, P.S., 1997. Genetic and environmental regulation of dauer larva
Mark, K.A., Dumas, K.J., Bhaumik, D., Schilling, B., Davis, S., Oron, T.R., Sorensen, D.J., development. In: Riddle, D.L., Blumenthal, T., Meyer, B.J., et al. (Eds.), C. elegans II,
Lucanic, M., Brem, R.B., Melov, S., Ramanathan, A., Gibson, B.W., Lithgow, G.J., second ed. Cold Spring Harbor Laboratory Press, Cold Spring Harbor (NY) (Chapter
2016. Vitamin D promotes protein homeostasis and longevity via the stress response 26).
pathway genes skn-1, ire-1, and xbp-1. Cell Rep. 17 (5), 1227–1237. Riddle, D.L., Swanson, M.M., Albert, P.S., 1981. Interacting genes in nematode dauer
McElwee, J.J., Schuster, E., Blanc, E., Thornton, J., Gems, D., 2006. Diapause-associated larva formation. Nature 290 (5808), 668–671. https://doi.org/10.1038/290668a0.
metabolic traits reiterated in long-lived daf-2 mutants in the nematode Caenorhabditis Roux, A.E., Langhans, K., Huynh, W., Kenyon, C., 2016. Reversible age-related
elegans. Mech. Ageing Develop. 127 (5), 458–472. https://doi.org/10.1016/ phenotypes induced during larval quiescence in C. elegans. Cell Metabol. 23 (6),
j.mad.2006.01.006. 1113–1126. https://doi.org/10.1016/j.cmet.2016.05.024.

275
S. Rashid et al. Developmental Biology 475 (2021) 265–276

Roy, D., Michaelson, D., Hochman, T., Santella, A., Bao, Z., Goldberg, J.D., Sommer, R.J., 2020. Phenotypic plasticity: from theory and genetics to current and future
Hubbard, E.J.A., 2016. Cell cycle features of C. elegans germline stem/progenitor cells challenges. Genetics 215 (1), 1–13. https://doi.org/10.1534/genetics.120.303163.
vary temporally and spatially. Dev. Biol. 409 (1), 261–271. https://doi.org/10.1016/ Szewczyk, N.J., Kozak, E., Conley, C.A., 2003. Chemically defined medium and
j.ydbio.2015.10.031. Caenorhabditis elegans. BMC Biotechnol. 3, 19. https://doi.org/10.1186/1472-6750-
Samuel, B.S., Rowedder, H., Braendle, C., Felix, M.-A., Ruvkun, G., 2016. Caenorhabditis 3-19.
elegans responses to bacteria from its natural habitats. Proc. Natl. Acad. Sci. U.S.A. Takamiya, S., Matsui, T., Taka, H., Murayama, K., Matsuda, M., Aoki, T., 1999. Free-living
113 (27), E3941–E3949. nematodes Caenorhabditis elegans possess in their mitochondria an additional
Sancak, Y., Peterson, T.R., Shaul, Y.D., Lindquist, R.A., Thoreen, C.C., Bar-Peled, L., rhodoquinone, an essential component of the eukaryotic fumarate reductase system.
Sabatini, D.M., 2008. The Rag GTPases bind raptor and mediate amino acid signalling Arch. Biochem. Biophys. 371 (2), 284–289. https://doi.org/10.1006/
to mTORC1. Science 320 (5882), 1496–1501. https://doi.org/10.1126/ abbi.1999.1465.
science.1157535. Tilton, W.M., Seaman, C., Carriero, D., Piomelli, S., 1991. Regulation of glycolysis in the
Sanzo-Machuca, A., Monje Moreno, J.M., Casado-Navarro, R., Karakuzu, O., Guerrero- erythrocyte: role of the lactate/pyruvate and NAD/NADH ratios. J. Lab. Clin. Med.
Gomez, D., Fierro-Gonz alez, J.C., Swoboda, P., Mu~ noz, M.J., Garsin, D.A., 118 (2), 146–152.
Pedrajas, J.R., Barrios, A., Miranda-Vizuete, A., 2019. Redox-dependent and redox- Tonkin, L.A., Bass, B.L., 2003. Mutations in RNAi rescue aberrant chemotaxis of ADAR
independent functions of Caenorhabditis elegans thioredoxin 1. Redox Biol. 24, mutants. Science 302 (5651), 1725. https://doi.org/10.1126/science.1091340.
101178. https://doi.org/10.1016/j.redox.2019.101178. Wamelink, M.M.C., Struys, E.A., Jakobs, C., 2008. The biochemistry, metabolism and
Schindler, A.J., Baugh, L.R., Sherwood, D.R., 2014. Identification of late larval stage inherited defects of the pentose phosphate pathway: a review. J. Inherit. Metab. Dis.
developmental checkpoints in Caenorhabditis elegans regulated by insulin/IGF and 31 (6), 703–717.
steroid hormone signalling pathways. PLoS Genet. 10 (6), e1004426 https://doi.org/ Wang, J., Kim, S.K., 2003. Global analysis of dauer gene expression in Caenorhabditis
10.1371/journal.pgen.1004426. elegans. Development 130 (8), 1621–1634.
Schulenburg, H., Felix, M.A., 2017. The natural biotic environment of Caenorhabditis Watanabe, S., Yamamoto, T.G., Kitagawa, R., 2008. Spindle assembly checkpoint gene
elegans. Genetics 206 (1), 55–86. https://doi.org/10.1534/genetics.116.195511. mdf-1 regulates germ cell proliferation in response to nutrition signals in C. elegans.
Seidel, H.S., Kimble, J., 2011. The oogenic germline starvation response in C. elegans. PloS EMBO J. 27 (7), 1085–1096. https://doi.org/10.1038/emboj.2008.32.
One 6 (12), e28074. https://doi.org/10.1371/journal.pone.0028074. Watson, E., MacNeil, L.T., Ritter, A.D., Yilmaz, L.S., Rosebrock, A.P., Caudy, A.A.,
Seidel, H.S., Kimble, J., 2015. Cell-cycle quiescence maintains Caenorhabditis elegans Walhout, A.J., 2014. Interspecies systems biology uncovers metabolites affecting
germline stem cells independent of GLP-1/Notch. eLife 4, e10832. https://doi.org/ C. elegans gene expression and life history traits. Cell 156 (4), 759–770. https://
10.7554/eLife.10832. doi.org/10.1016/j.cell.2014.01.047.
Shao, D., Oka, S., Liu, T., Zhai, P., Ago, T., Sciarretta, S., Li, H., Sadoshima, J., 2014. Yilmaz, L.S., Walhout, A.J., 2014. Worms, bacteria, and micronutrients: an elegant model
A redox-dependent mechanism for regulation of AMPK activation by Thioredoxin1 of our diet. Trends Genet. 30 (11), 496–503. https://doi.org/10.1016/
during energy starvation. Cell Metabol. 19 (2), 232–245. https://doi.org/10.1016/ j.tig.2014.07.010.
j.cmet.2013.12.013. Zecic, A., Dhondt, I., Braeckman, B.P., 2019. The nutritional requirements of
Shen, Y., Wollam, J., Magner, D., Karalay, O., Antebi, A., 2012. A steroid receptor- Caenorhabditis elegans. Genes & nutrition 14, 15. https://doi.org/10.1186/s12263-
microRNA switch regulates life span in response to signals from the gonad. Science 019-0637-7.
338 (6113), 1472–1476. https://doi.org/10.1126/science.1228967. Zhang, X., Zabinsky, R., Teng, Y., Cui, M., Han, M., 2011. microRNAs play critical roles in
Sims, J.R., Ow, M.C., Nishiguchi, M.A., Kim, K., Sengupta, P., Hall, S.E., 2016. the survival and recovery of Caenorhabditis elegans from starvation-induced L1
Developmental programming modulates olfactory behaviour in C. elegans via diapause. Proc. Natl. Acad. Sci. U.S.A. 108 (44), 17997–18002.
endogenous RNAi pathways. eLife 5, ee1642. https://doi.org/10.7554/eLife.11642. Zheng, S., Qu, Z., Zanetti, M., Lam, B., Chin-Sang, I., 2018. C. elegans PTEN and AMPK
Solari, F., Bourbon-Piffaut, A., Masse, I., Payrastre, B., Chan, A.M.L., Billaud, M., 2004. block neuroblast divisions by inhibiting a BMP-insulin-PP2A-MAPK pathway.
The human tumour suppressor PTEN regulates longevity and dauer formation in Development 145 (23), dev166876. https://doi.org/10.1242/dev.166876.
Caenorhabditis elegans. Oncogene 24 (1), 20–27. https://doi.org/10.1038/
sj.onc.1207978.

276

You might also like