Download PDF

Plos Genetics

Publication date: 2021-11-01
Volume: 17
Publisher: Public Library of Science (PLoS)

Author:

Marques, Filipe
Falquet, Laurent ; Vandewyer, Elke ; Beets, Isabel ; Glauser, Dominique A

Keywords:

Science & Technology, Life Sciences & Biomedicine, Genetics & Heredity, PROTEIN-COUPLED RECEPTORS, CAENORHABDITIS-ELEGANS, MOLECULAR-MECHANISMS, PARKINSONS-DISEASE, NERVOUS-SYSTEM, PAIN, HABITUATION, HEAT, SENSITIZATION, CHANNELS, Animals, Caenorhabditis elegans, Caenorhabditis elegans Proteins, Escape Reaction, Genes, Helminth, Hot Temperature, Neurons, Neuropeptides, Nociception, Protein Isoforms, Receptors, G-Protein-Coupled, Signal Transduction, G093419N#54969810, 0604 Genetics, Developmental Biology, 3105 Genetics

Abstract:

In order to thrive in constantly changing environments, animals must adaptively respond to threatening events. Noxious stimuli are not only processed according to their absolute intensity, but also to their context. Adaptation processes can cause animals to habituate at different rates and degrees in response to permanent or repeated stimuli. Here, we used a forward genetic approach in Caenorhabditis elegans to identify a neuropeptidergic pathway, essential to prevent fast habituation and maintain robust withdrawal responses to repeated noxious stimuli. This pathway involves the FRPR-19A and FRPR-19B G-protein coupled receptor isoforms produced from the frpr-19 gene by alternative splicing. Loss or overexpression of each or both isoforms can impair withdrawal responses caused by the optogenetic activation of the polymodal FLP nociceptor neuron. Furthermore, we identified FLP-8 and FLP-14 as FRPR-19 ligands in vitro. flp-14, but not flp-8, was essential to promote withdrawal response and is part of the same genetic pathway as frpr-19 in vivo. Expression and cell-specific rescue analyses suggest that FRPR-19 acts both in the FLP nociceptive neurons and downstream interneurons, whereas FLP-14 acts from interneurons. Importantly, genetic impairment of the FLP-14/FRPR-19 pathway accelerated the habituation to repeated FLP-specific optogenetic activation, as well as to repeated noxious heat and harsh touch stimuli. Collectively, our data suggest that well-adjusted neuromodulation via the FLP-14/FRPR-19 pathway contributes to promote nociceptive signals in C. elegans and counteracts habituation processes that otherwise tend to rapidly reduce aversive responses to repeated noxious stimuli.