Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
Nature ; 613(7943): 324-331, 2023 01.
Article in English | MEDLINE | ID: mdl-36599989

ABSTRACT

Pathogens generate ubiquitous selective pressures and host-pathogen interactions alter social behaviours in many animals1-4. However, very little is known about the neuronal mechanisms underlying pathogen-induced changes in social behaviour. Here we show that in adult Caenorhabditis elegans hermaphrodites, exposure to a bacterial pathogen (Pseudomonas aeruginosa) modulates sensory responses to pheromones by inducing the expression of the chemoreceptor STR-44 to promote mating. Under standard conditions, C. elegans hermaphrodites avoid a mixture of ascaroside pheromones to facilitate dispersal5-13. We find that exposure to the pathogenic Pseudomonas bacteria enables pheromone responses in AWA sensory neurons, which mediate attractive chemotaxis, to suppress the avoidance. Pathogen exposure induces str-44 expression in AWA neurons, a process regulated by a transcription factor zip-5 that also displays a pathogen-induced increase in expression in AWA. STR-44 acts as a pheromone receptor and its function in AWA neurons is required for pathogen-induced AWA pheromone response and suppression of pheromone avoidance. Furthermore, we show that C. elegans hermaphrodites, which reproduce mainly through self-fertilization, increase the rate of mating with males after pathogen exposure and that this increase requires str-44 in AWA neurons. Thus, our results uncover a causal mechanism for pathogen-induced social behaviour plasticity, which can promote genetic diversity and facilitate adaptation of the host animals.


Subject(s)
Caenorhabditis elegans , Pheromones , Pseudomonas aeruginosa , Reproduction , Sexual Behavior, Animal , Animals , Female , Male , Caenorhabditis elegans/metabolism , Caenorhabditis elegans/microbiology , Caenorhabditis elegans/physiology , Caenorhabditis elegans Proteins/metabolism , Glycolipids/metabolism , Hermaphroditic Organisms/physiology , Pheromones/metabolism , Pseudomonas aeruginosa/pathogenicity , Pseudomonas aeruginosa/physiology , Receptors, Pheromone/metabolism , Reproduction/physiology , Sensory Receptor Cells/metabolism
2.
PLoS Genet ; 17(7): e1009678, 2021 07.
Article in English | MEDLINE | ID: mdl-34260587

ABSTRACT

Animals can adapt to dynamic environmental conditions by modulating their developmental programs. Understanding the genetic architecture and molecular mechanisms underlying developmental plasticity in response to changing environments is an important and emerging area of research. Here, we show a novel role of cAMP response element binding protein (CREB)-encoding crh-1 gene in developmental polyphenism of C. elegans. Under conditions that promote normal development in wild-type animals, crh-1 mutants inappropriately form transient pre-dauer (L2d) larvae and express the L2d marker gene. L2d formation in crh-1 mutants is specifically induced by the ascaroside pheromone ascr#5 (asc-ωC3; C3), and crh-1 functions autonomously in the ascr#5-sensing ASI neurons to inhibit L2d formation. Moreover, we find that CRH-1 directly binds upstream of the daf-7 TGF-ß locus and promotes its expression in the ASI neurons. Taken together, these results provide new insight into how animals alter their developmental programs in response to environmental changes.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism , Adaptation, Physiological/genetics , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Cell Cycle , Cell Growth Processes , Cyclic AMP Response Element-Binding Protein/physiology , Gene Expression/genetics , Gene Expression Regulation/genetics , Gene Expression Regulation, Developmental/genetics , Larva/genetics , Larva/growth & development , Pheromones/metabolism , Sensory Receptor Cells/metabolism , Signal Transduction/genetics , Transcription Factors/genetics , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/physiology
3.
J Neurosci ; 42(5): 720-730, 2022 02 02.
Article in English | MEDLINE | ID: mdl-34862187

ABSTRACT

Chemical communication controls a wide range of behaviors via conserved signaling networks. Axon regeneration in response to injury is determined by the interaction between the extracellular environment and intrinsic growth potential. In this study, we investigated the role of chemical signaling in axon regeneration in Caenorhabditis elegans We find that the enzymes involved in ascaroside pheromone biosynthesis, ACOX-1.1, ACOX-1.2, and DAF-22, participate in axon regeneration by producing a dauer-inducing ascaroside, ascr#5. We demonstrate that the chemoreceptor genes, srg-36 and srg-37, which encode G-protein-coupled receptors for ascr#5, are required for adult-specific axon regeneration. Furthermore, the activating mutation in egl-30 encoding Gqα suppresses axon regeneration defective phenotype in acox-1.1 and srg-36 srg-37 mutants. Therefore, the ascaroside signaling system provides a unique example of a signaling molecule that regulates the regenerative pathway in the nervous system.SIGNIFICANCE STATEMENT In Caenorhabditis elegans, axon regeneration is positively regulated by the EGL-30 Gqα-JNK MAP kinase cascade. However, it remains unclear what signals activate the EGL-30 pathway in axon regeneration. Here, we show that SRG-36 and SRG-37 act as upstream G-protein-coupled receptors (GPCRs) that activate EGL-30. C. elegans secretes a family of small-molecule pheromones called ascarosides, which serve various functions in chemical signaling. SRG-36 and SRG-37 are GPCRs for the dauer-inducing ascaroside ascr#5. Consistent with this, we found that ascr#5 activates the axon regeneration pathway via SRG-36/SRG-37 and EGL-30. Thus, ascaroside signaling promotes axon regeneration by activating the GPCR-Gqα pathway.


Subject(s)
Axons/physiology , Caenorhabditis elegans Proteins/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Nerve Regeneration/physiology , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/physiology , Animals , Animals, Genetically Modified , Caenorhabditis elegans , Caenorhabditis elegans Proteins/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , Receptors, G-Protein-Coupled/genetics
4.
Nature ; 542(7639): 43-48, 2017 02 02.
Article in English | MEDLINE | ID: mdl-28099418

ABSTRACT

Interleukin-17 (IL-17) is a major pro-inflammatory cytokine: it mediates responses to pathogens or tissue damage, and drives autoimmune diseases. Little is known about its role in the nervous system. Here we show that IL-17 has neuromodulator-like properties in Caenorhabditis elegans. IL-17 can act directly on neurons to alter their response properties and contribution to behaviour. Using unbiased genetic screens, we delineate an IL-17 signalling pathway and show that it acts in the RMG hub interneurons. Disrupting IL-17 signalling reduces RMG responsiveness to input from oxygen sensors, and renders sustained escape from 21% oxygen transient and contingent on additional stimuli. Over-activating IL-17 receptors abnormally heightens responses to 21% oxygen in RMG neurons and whole animals. IL-17 deficiency can be bypassed by optogenetic stimulation of RMG. Inducing IL-17 expression in adults can rescue mutant defects within 6 h. These findings reveal a non-immunological role of IL-17 modulating circuit function and behaviour.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/cytology , Caenorhabditis elegans/physiology , Interleukin-17/metabolism , Sensation/physiology , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Caenorhabditis elegans/drug effects , HEK293 Cells , Humans , Interneurons/drug effects , Interneurons/metabolism , Oxygen/metabolism , Oxygen/pharmacology , Receptors, Interleukin-17/metabolism , Sensation/drug effects , Signal Transduction/drug effects
5.
EMBO J ; 37(15)2018 08 01.
Article in English | MEDLINE | ID: mdl-29925517

ABSTRACT

Animals change sensory responses and their eventual behaviors, depending on their internal metabolic status and external food availability. However, the mechanisms underlying feeding state-dependent behavioral changes remain undefined. Previous studies have shown that Caenorhabditis elegans hermaphrodite exhibits avoidance behaviors to acute exposure of a pheromone, ascr#3 (asc-ΔC9, C9). Here, we show that the ascr#3 avoidance behavior is modulated by feeding state via the insulin signaling pathway. Starvation increases ascr#3 avoidance behavior, and loss-of-function mutations in daf-2 insulin-like receptor gene dampen this starvation-induced ascr#3 avoidance behavior. DAF-2 and its downstream signaling molecules, including the DAF-16 FOXO transcription factor, act in the ascr#3-sensing ADL neurons to regulate synaptic transmission to downstream target neurons, including the AVA command interneurons. Moreover, we found that starvation decreases the secretion of INS-18 insulin-like peptides from the intestine, which antagonizes DAF-2 function in the ADL neurons. Altogether, this study provides insights about the molecular communication between intestine and sensory neurons delivering hunger message to sensory neurons, which regulates avoidance behavior from pheromones to facilitate survival chance.


Subject(s)
Avoidance Learning/physiology , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Insulin/metabolism , Receptor, Insulin/metabolism , Starvation/metabolism , Synaptic Transmission/physiology , Animals , Caenorhabditis elegans Proteins/genetics , Forkhead Transcription Factors/genetics , Neurons/metabolism , Peptide Hormones/metabolism , Pheromones/metabolism , Receptor, Insulin/genetics , Signal Transduction , Synaptic Transmission/genetics
6.
Nature ; 539(7628): 254-258, 2016 11 10.
Article in English | MEDLINE | ID: mdl-27799655

ABSTRACT

The optimal foraging strategy in a given environment depends on the number of competing individuals and their behavioural strategies. Little is known about the genes and neural circuits that integrate social information into foraging decisions. Here we show that ascaroside pheromones, small glycolipids that signal population density, suppress exploratory foraging in Caenorhabditis elegans, and that heritable variation in this behaviour generates alternative foraging strategies. We find that natural C. elegans isolates differ in their sensitivity to the potent ascaroside icas#9 (IC-asc-C5). A quantitative trait locus (QTL) regulating icas#9 sensitivity includes srx-43, a G-protein-coupled icas#9 receptor that acts in the ASI class of sensory neurons to suppress exploration. Two ancient haplotypes associated with this QTL confer competitive growth advantages that depend on ascaroside secretion, its detection by srx-43 and the distribution of food. These results suggest that balancing selection at the srx-43 locus generates alternative density-dependent behaviours, fulfilling a prediction of foraging game theory.


Subject(s)
Caenorhabditis elegans/genetics , Caenorhabditis elegans/physiology , Feeding Behavior , Selection, Genetic , Animals , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/isolation & purification , Caenorhabditis elegans Proteins/metabolism , Feeding Behavior/drug effects , Food , Game Theory , Haplotypes , Hexoses/metabolism , Hexoses/pharmacology , Indoles/pharmacology , Male , Pheromones/metabolism , Pheromones/pharmacology , Population Density , Quantitative Trait Loci , Receptors, G-Protein-Coupled/metabolism , Sensory Receptor Cells/metabolism , Social Behavior
7.
J Am Chem Soc ; 142(32): 13645-13650, 2020 08 12.
Article in English | MEDLINE | ID: mdl-32702987

ABSTRACT

The nematode Caenorhabditis elegans produces a broad family of pheromones, known as the ascarosides, that are modified with a variety of groups derived from primary metabolism. These modifications are essential for the diverse activities of the ascarosides in development and various behaviors, including attraction, aggregation, avoidance, and foraging. The mechanism by which these different groups are added to the ascarosides is poorly understood. Here, we identify a family of over 30 enzymes, which are homologous to mammalian carboxylesterase (CES) enzymes, and show that a number of these enzymes are responsible for the selective addition of specific modifications to the ascarosides. Through stable isotope feeding experiments, we demonstrate the in vivo activity of the CES-like enzymes and provide direct evidence that the acyl-CoA synthetase ACS-7, which was previously implicated in the attachment of certain modifications to the ascarosides in C. elegans, instead activates the side chains of certain ascarosides for shortening through ß-oxidation. Our data provide a key to the combinatorial logic that gives rise to different modified ascarosides, which should greatly facilitate the exploration of the specific biological functions of these pheromones in the worm.


Subject(s)
Caenorhabditis elegans/enzymology , Carboxylesterase/metabolism , Coenzyme A Ligases/metabolism , Animals , Glycolipids/biosynthesis , Glycolipids/chemistry , Molecular Structure
8.
Nat Chem Biol ; 13(6): 577-586, 2017 05 17.
Article in English | MEDLINE | ID: mdl-28514418

ABSTRACT

The existence of small-molecule signals that influence development in Caenorhabditis elegans has been known for several decades, but only in recent years have the chemical structures of several of these signals been established. The identification of these signals has enabled connections to be made between these small molecules and fundamental signaling pathways in C. elegans that influence not only development but also metabolism, fertility, and lifespan. Spurred by these important discoveries and aided by recent advances in comparative metabolomics and NMR spectroscopy, the field of nematode chemistry has the potential to expand dramatically in the coming years. This Perspective will focus on small-molecule pheromones and hormones that influence developmental events in the nematode life cycle (ascarosides, dafachronic acids, and nemamides), will cover more recent work regarding the biosynthesis of these signals, and will explore how the discovery of these signals is transforming our understanding of nematode development and physiology.


Subject(s)
Hormones/metabolism , Nematoda/growth & development , Pheromones/metabolism , Animals , Caenorhabditis elegans/growth & development , Hormones/chemistry , Life Cycle Stages/physiology , Molecular Structure , Pheromones/chemistry , Reproduction
9.
J Exp Biol ; 222(Pt 18)2019 09 25.
Article in English | MEDLINE | ID: mdl-31511342

ABSTRACT

In this study, we assessed the effect of symbiotic (cognate and non-cognate) and non-symbiotic bacteria on ascaroside production of first-generation adults in two Steinernema spp.: S. carpocapsae All strain and S. feltiae SN strain. Each nematode species was reared under three bacterial scenarios: (1) cognate symbiotic, (2) non-cognate symbiotic strain and (3) non-cognate symbiotic species. Our results showed S. carpocapsae produced four quantifiable ascaroside molecules: asc-C5, asc-C6, asc-C7 and asc-C11, whereas in S. feltiae only three molecules were detected: asc-C5, asc-C7 and asc-C11. Bacterial conditions did not significantly affect the quantity of the secreted ascarosides in first-generation adults of S. carpocapsae However, in S. feltiae, Xenorhabdus nematophila All strain influenced the production of two ascaroside molecules: asc-C5 and asc-C11.


Subject(s)
Pheromones/metabolism , Rhabditida/metabolism , Rhabditida/microbiology , Xenorhabdus , Animals , Bacteria , Bacterial Physiological Phenomena , Glycolipids/metabolism , Symbiosis
10.
Proc Natl Acad Sci U S A ; 113(36): 10055-60, 2016 09 06.
Article in English | MEDLINE | ID: mdl-27551084

ABSTRACT

Caenorhabditis elegans secretes ascarosides as pheromones to communicate with other worms and to coordinate the development and behavior of the population. Peroxisomal ß-oxidation cycles shorten the side chains of ascaroside precursors to produce the short-chain ascaroside pheromones. Acyl-CoA oxidases, which catalyze the first step in these ß-oxidation cycles, have different side chain-length specificities and enable C. elegans to regulate the production of specific ascaroside pheromones. Here, we determine the crystal structure of the acyl-CoA oxidase 1 (ACOX-1) homodimer and the ACOX-2 homodimer bound to its substrate. Our results provide a molecular basis for the substrate specificities of the acyl-CoA oxidases and reveal why some of these enzymes have a very broad substrate range, whereas others are quite specific. Our results also enable predictions to be made for the roles of uncharacterized acyl-CoA oxidases in C. elegans and in other nematode species. Remarkably, we show that most of the C. elegans acyl-CoA oxidases that participate in ascaroside biosynthesis contain a conserved ATP-binding pocket that lies at the dimer interface, and we identify key residues in this binding pocket. ATP binding induces a structural change that is associated with tighter binding of the FAD cofactor. Mutations that disrupt ATP binding reduce FAD binding and reduce enzyme activity. Thus, ATP may serve as a regulator of acyl-CoA oxidase activity, thereby directly linking ascaroside biosynthesis to ATP concentration and metabolic state.


Subject(s)
Acyl-CoA Oxidase/chemistry , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans/chemistry , Pheromones/chemistry , Acyl-CoA Oxidase/genetics , Acyl-CoA Oxidase/metabolism , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Animals , Binding Sites , Caenorhabditis elegans/enzymology , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Crystallography, X-Ray , Flavin-Adenine Dinucleotide/chemistry , Flavin-Adenine Dinucleotide/metabolism , Gene Expression , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Kinetics , Models, Molecular , Mutation , Oxidation-Reduction , Pheromones/biosynthesis , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Multimerization , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Substrate Specificity
11.
PLoS Genet ; 12(7): e1006219, 2016 07.
Article in English | MEDLINE | ID: mdl-27467070

ABSTRACT

Evolutionary life history theory seeks to explain how reproductive and survival traits are shaped by selection through allocations of an individual's resources to competing life functions. Although life-history traits evolve rapidly, little is known about the genetic and cellular mechanisms that control and couple these tradeoffs. Here, we find that two laboratory-adapted strains of C. elegans descended from a single common ancestor that lived in the 1950s have differences in a number of life-history traits, including reproductive timing, lifespan, dauer formation, growth rate, and offspring number. We identified a quantitative trait locus (QTL) of large effect that controls 24%-75% of the total trait variance in reproductive timing at various timepoints. Using CRISPR/Cas9-induced genome editing, we show this QTL is due in part to a 60 bp deletion in the 3' end of the nurf-1 gene, which is orthologous to the human gene encoding the BPTF component of the NURF chromatin remodeling complex. Besides reproduction, nurf-1 also regulates growth rate, lifespan, and dauer formation. The fitness consequences of this deletion are environment specific-it increases fitness in the growth conditions where it was fixed but decreases fitness in alternative laboratory growth conditions. We propose that chromatin remodeling, acting through nurf-1, is a pleiotropic regulator of life history trade-offs underlying the evolution of multiple traits across different species.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/genetics , Chromosomal Proteins, Non-Histone/genetics , Evolution, Molecular , Selection, Genetic/genetics , Animals , CRISPR-Cas Systems , Chromatin/genetics , Chromatin Assembly and Disassembly/genetics , Humans , Phenotype , Quantitative Trait Loci/genetics , Reproduction/genetics
12.
Nat Chem Biol ; 12(10): 770-2, 2016 10.
Article in English | MEDLINE | ID: mdl-27501395

ABSTRACT

Polyketides and nonribosomal peptides are two important types of natural products that are produced by many species of bacteria and fungi but are exceedingly rare in metazoans. Here, we elucidate the structure of a hybrid polyketide-nonribosomal peptide from Caenorhabditis elegans that is produced in the canal-associated neurons (CANs) and promotes survival during starvation-induced larval arrest. Our results uncover a novel mechanism by which animals respond to nutrient fluctuations to extend survival.


Subject(s)
Caenorhabditis elegans/growth & development , Caenorhabditis elegans/metabolism , Larva/growth & development , Peptides/metabolism , Polyketides/metabolism , Animals , Biological Products/chemistry , Biological Products/metabolism , Biological Products/pharmacology , Caenorhabditis elegans/cytology , Caenorhabditis elegans/drug effects , Larva/drug effects , Neurons/metabolism , Peptides/chemistry , Peptides/pharmacology , Polyketides/chemistry , Polyketides/pharmacology
13.
Proc Natl Acad Sci U S A ; 112(13): 3955-60, 2015 Mar 31.
Article in English | MEDLINE | ID: mdl-25775534

ABSTRACT

Caenorhabditis elegans uses ascaroside pheromones to induce development of the stress-resistant dauer larval stage and to coordinate various behaviors. Peroxisomal ß-oxidation cycles are required for the biosynthesis of the fatty acid-derived side chains of the ascarosides. Here we show that three acyl-CoA oxidases, which catalyze the first step in these ß-oxidation cycles, form different protein homo- and heterodimers with distinct substrate preferences. Mutations in the acyl-CoA oxidase genes acox-1, -2, and -3 led to specific defects in ascaroside production. When the acyl-CoA oxidases were expressed alone or in pairs and purified, the resulting acyl-CoA oxidase homo- and heterodimers displayed different side-chain length preferences in an in vitro activity assay. Specifically, an ACOX-1 homodimer controls the production of ascarosides with side chains with nine or fewer carbons, an ACOX-1/ACOX-3 heterodimer controls the production of those with side chains with seven or fewer carbons, and an ACOX-2 homodimer controls the production of those with ω-side chains with less than five carbons. Our results support a biosynthetic model in which ß-oxidation enzymes act directly on the CoA-thioesters of ascaroside biosynthetic precursors. Furthermore, we identify environmental conditions, including high temperature and low food availability, that induce the expression of acox-2 and/or acox-3 and lead to corresponding changes in ascaroside production. Thus, our work uncovers an important mechanism by which C. elegans increases the production of the most potent dauer pheromones, those with the shortest side chains, under specific environmental conditions.


Subject(s)
Acyl-CoA Oxidase/metabolism , Caenorhabditis elegans/enzymology , Pheromones/metabolism , Animals , Catalysis , Chromatography, Liquid , Mutation , Oxygen/metabolism , Polymerase Chain Reaction , Protein Multimerization , Substrate Specificity , Tandem Mass Spectrometry , Temperature
14.
Nat Prod Rep ; 34(5): 472-477, 2017 May 10.
Article in English | MEDLINE | ID: mdl-28386618

ABSTRACT

The nematode Caenorhabditis elegans produces tens, if not hundreds, of different ascarosides as pheromones to communicate with other members of its species. Overlapping mixtures of these pheromones affect the development of the worm and a variety of different behaviors. The ascarosides represent a unique tool for dissecting the neural circuitry that controls behavior and that connects to important signaling pathways, such as the insulin and TGFß pathways, that lie at the nexus of development, metabolism, and lifespan in C. elegans. However, the exact physiological roles of many of the ascarosides are unclear, especially since many of these pheromones likely have multiple functions depending on their concentrations, the presence of other pheromones, and a variety of other factors. Determining these physiological roles will be facilitated by top-down approaches to characterize the pheromone receptors and their function, as well as bottom-up approaches to characterize the pheromone biosynthetic enzymes and their regulation.


Subject(s)
Caenorhabditis elegans/chemistry , Pheromones/physiology , Animals , Molecular Structure , Pheromones/chemistry
15.
Nature ; 477(7364): 321-5, 2011 Aug 17.
Article in English | MEDLINE | ID: mdl-21849976

ABSTRACT

Evolution can follow predictable genetic trajectories, indicating that discrete environmental shifts can select for reproducible genetic changes. Conspecific individuals are an important feature of an animal's environment, and a potential source of selective pressures. Here we show that adaptation of two Caenorhabditis species to growth at high density, a feature common to domestic environments, occurs by reproducible genetic changes to pheromone receptor genes. Chemical communication through pheromones that accumulate during high-density growth causes young nematode larvae to enter the long-lived but non-reproductive dauer stage. Two strains of Caenorhabditis elegans grown at high density have independently acquired multigenic resistance to pheromone-induced dauer formation. In each strain, resistance to the pheromone ascaroside C3 results from a deletion that disrupts the adjacent chemoreceptor genes serpentine receptor class g (srg)-36 and -37. Through misexpression experiments, we show that these genes encode redundant G-protein-coupled receptors for ascaroside C3. Multigenic resistance to dauer formation has also arisen in high-density cultures of a different nematode species, Caenorhabditis briggsae, resulting in part from deletion of an srg gene paralogous to srg-36 and srg-37. These results demonstrate rapid remodelling of the chemoreceptor repertoire as an adaptation to specific environments, and indicate that parallel changes to a common genetic substrate can affect life-history traits across species.


Subject(s)
Biological Evolution , Caenorhabditis elegans/genetics , Caenorhabditis elegans/physiology , Receptors, Pheromone/genetics , Adaptation, Physiological/genetics , Adaptation, Physiological/physiology , Animals , Caenorhabditis elegans/classification , Caenorhabditis elegans/drug effects , Environment , Evolution, Molecular , Glycolipids/metabolism , Glycolipids/pharmacology , Hibernation/genetics , Hibernation/physiology , Larva/growth & development , Pheromones/metabolism , Pheromones/pharmacology , Population Density , Quantitative Trait Loci/genetics , Receptors, Pheromone/metabolism
16.
Biochem J ; 473(11): 1507-21, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27009306

ABSTRACT

L-Rhamnose is a common component of cell-wall polysaccharides, glycoproteins and some natural products in bacteria and plants, but is rare in fungi and animals. In the present study, we identify and characterize a biosynthetic pathway for dTDP-rhamnose in Caenorhabditis elegans that is highly conserved across nematode species. We show that RML-1 activates glucose 1-phosphate (Glc-1-P) in the presence of either dTTP or UTP to yield dTDP-glucose or UDP-glucose, respectively. RML-2 is a dTDP-glucose 4,6-dehydratase, converting dTDP-glucose into dTDP-4-keto-6-deoxyglucose. Using mass spectrometry and NMR spectroscopy, we demonstrate that coincubation of dTDP-4-keto-6-deoxyglucose with RML-3 (3,5-epimerase) and RML-4 (4-keto-reductase) produces dTDP-rhamnose. RML-4 could only be expressed and purified in an active form through co-expression with a co-regulated protein, RML-5, which forms a complex with RML-4. Analysis of the sugar nucleotide pool in C. elegans established the presence of dTDP-rhamnose in vivo Targeting the expression of the rhamnose biosynthetic genes by RNAi resulted in significant reductions in dTDP-rhamnose, but had no effect on the biosynthesis of a closely related sugar, ascarylose, found in the ascaroside pheromones. Therefore, the rhamnose and ascarylose biosynthetic pathways are distinct. We also show that transcriptional reporters for the rhamnose biosynthetic genes are expressed highly in the embryo, in the hypodermis during molting cycles and in the hypodermal seam cells specifically before the molt to the stress-resistant dauer larval stage. These expression patterns suggest that rhamnose biosynthesis may play an important role in hypodermal development or the production of the cuticle or surface coat during molting.


Subject(s)
Caenorhabditis elegans/metabolism , Molting/physiology , Rhamnose/metabolism , Thymine Nucleotides/metabolism , Animals , Biosynthetic Pathways/physiology , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Chromatography, High Pressure Liquid , Deoxyglucose/analogs & derivatives , Deoxyglucose/metabolism , Glucose/analogs & derivatives , Glucose/metabolism , Glucose-1-Phosphate Adenylyltransferase/genetics , Glucose-1-Phosphate Adenylyltransferase/metabolism , Kinetics , Magnetic Resonance Spectroscopy , Mass Spectrometry , Molting/genetics , Phylogeny , RNA Interference , Uridine Diphosphate Glucose/metabolism
17.
Nature ; 458(7242): 1171-5, 2009 Apr 30.
Article in English | MEDLINE | ID: mdl-19349961

ABSTRACT

Innate social behaviours emerge from neuronal circuits that interpret sensory information on the basis of an individual's own genotype, sex and experience. The regulated aggregation behaviour of the nematode Caenorhabditis elegans, a simple animal with only 302 neurons, is an attractive system to analyse these circuits. Wild social strains of C. elegans aggregate in the presence of specific sensory cues, but solitary strains do not. Here we identify the RMG inter/motor neuron as the hub of a regulated circuit that controls aggregation and related behaviours. RMG is the central site of action of the neuropeptide receptor gene npr-1, which distinguishes solitary strains (high npr-1 activity) from wild social strains (low npr-1 activity); high RMG activity is essential for all aspects of social behaviour. Anatomical gap junctions connect RMG to several classes of sensory neurons known to promote aggregation, and to ASK sensory neurons, which are implicated in male attraction to hermaphrodite pheromones. We find that ASK neurons respond directly to pheromones, and that high RMG activity enhances ASK responses in social strains, causing hermaphrodite attraction to pheromones at concentrations that repel solitary hermaphrodites. The coordination of social behaviours by RMG suggests an anatomical hub-and-spoke model for sensory integration in aggregation, and points to functions for related circuit motifs in the C. elegans wiring diagram.


Subject(s)
Caenorhabditis elegans/physiology , Neural Pathways/physiology , Pheromones/physiology , Social Behavior , Animals , Caenorhabditis elegans/cytology , Caenorhabditis elegans/drug effects , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Disorders of Sex Development , Feeding Behavior/drug effects , Feeding Behavior/physiology , Male , Models, Neurological , Mutation , Neural Pathways/drug effects , Neurons/drug effects , Neurons/physiology , Pheromones/pharmacology , Receptors, Neuropeptide Y/genetics , Receptors, Neuropeptide Y/metabolism
18.
Front Mol Biosci ; 11: 1396587, 2024.
Article in English | MEDLINE | ID: mdl-39055986

ABSTRACT

Environmental conditions experienced early in the life of an animal can result in gene expression changes later in its life history. We have previously shown that C. elegans animals that experienced the developmentally arrested and stress resistant dauer stage (postdauers) retain a cellular memory of early-life stress that manifests during adulthood as genome-wide changes in gene expression, chromatin states, and altered life history traits. One consequence of developmental reprogramming in C. elegans postdauer adults is the downregulation of osm-9 TRPV channel gene expression in the ADL chemosensory neurons resulting in reduced avoidance to a pheromone component, ascr#3. This altered response to ascr#3 requires the principal effector of the somatic nuclear RNAi pathway, the Argonaute (AGO) NRDE-3. To investigate the role of the somatic nuclear RNAi pathway in regulating the developmental reprogramming of ADL due to early-life stress, we profiled the mRNA transcriptome of control and postdauer ADL in wild-type and nrde-3 mutant adults. We found 711 differentially expressed (DE) genes between control and postdauer ADL neurons, 90% of which are dependent upon NRDE-3. Additionally, we identified a conserved sequence that is enriched in the upstream regulatory sequences of the NRDE-3-dependent differentially expressed genes. Surprisingly, 214 of the ADL DE genes are considered "germline-expressed", including 21 genes encoding the Major Sperm Proteins and two genes encoding the sperm-specific PP1 phosphatases, GSP-3 and GSP-4. Loss of function mutations in gsp-3 resulted in both aberrant avoidance and attraction behaviors. We also show that an AGO pseudogene, Y49F6A.1 (wago-11), is expressed in ADL and is required for ascr#3 avoidance. Overall, our results suggest that small RNAs and reproductive genes program the ADL mRNA transcriptome during their developmental history and highlight a nexus between neuronal and reproductive networks in calibrating animal neuroplasticity.

19.
Cell Chem Biol ; 31(5): 1011-1022.e6, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38183989

ABSTRACT

Methyl ketone (MK)-ascarosides represent essential components of several pheromones in Caenorhabditis elegans, including the dauer pheromone, which triggers the stress-resistant dauer larval stage, and the male-attracting sex pheromone. Here, we identify an acyl-CoA thioesterase, ACOT-15, that is required for the biosynthesis of MK-ascarosides. We propose a model in which ACOT-15 hydrolyzes the ß-keto acyl-CoA side chain of an ascaroside intermediate during ß-oxidation, leading to decarboxylation and formation of the MK. Using comparative metabolomics, we identify additional ACOT-15-dependent metabolites, including an unusual piperidyl-modified ascaroside, reminiscent of the alkaloid pelletierine. The ß-keto acid generated by ACOT-15 likely couples to 1-piperideine to produce the piperidyl ascaroside, which is much less dauer-inducing than the dauer pheromone, asc-C6-MK (ascr#2, 1). The bacterial food provided influences production of the piperidyl ascaroside by the worm. Our work shows how the biosynthesis of MK- and piperidyl ascarosides intersect and how bacterial food may impact chemical signaling in the worm.


Subject(s)
Caenorhabditis elegans , Pheromones , Animals , Caenorhabditis elegans/metabolism , Caenorhabditis elegans/enzymology , Pheromones/metabolism , Pheromones/biosynthesis , Pheromones/chemistry , Caenorhabditis elegans Proteins/metabolism , Thiolester Hydrolases/metabolism
20.
Bioorg Med Chem ; 21(18): 5754-69, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23920482

ABSTRACT

The nematode Caenorhabditis elegans secretes ascarosides, structurally diverse derivatives of the 3,6-dideoxysugar ascarylose, and uses them in chemical communication. At high population densities, specific ascarosides, which are together known as the dauer pheromone, trigger entry into the stress-resistant dauer larval stage. In order to study the structure-activity relationships for the ascarosides, we synthesized a panel of ascarosides and tested them for dauer-inducing activity. This panel includes a number of natural ascarosides that were detected in crude pheromone extract, but as yet have no assigned function, as well as many unnatural ascaroside derivatives. Most of these ascarosides, some of which have significant structural similarity to the natural dauer pheromone components, have very little dauer-inducing activity. Our results provide a primer to ascaroside structure-activity relationships and suggest that slight modifications to ascaroside structure dramatically influence binding to the relevant G protein-coupled receptors that control dauer formation.


Subject(s)
Caenorhabditis elegans/metabolism , Glycolipids/chemistry , Pheromones/chemistry , Animals , Caenorhabditis elegans/growth & development , Glycolipids/chemical synthesis , Glycolipids/pharmacology , Larva/drug effects , Larva/physiology , Pheromones/chemical synthesis , Pheromones/pharmacology , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL