Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
PLoS Genet ; 17(11): e1009880, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34748554

RESUMEN

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.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/fisiología , Neuropéptidos/metabolismo , Nocicepción , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Animales , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Reacción de Fuga , Genes de Helminto , Calor , Neuronas/metabolismo , Neuropéptidos/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
2.
PLoS Genet ; 16(10): e1009102, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33104696

RESUMEN

Ryanodine receptors (RyR) are essential regulators of cellular calcium homeostasis and signaling. Vertebrate genomes contain multiple RyR gene isoforms, expressed in different tissues and executing different functions. In contrast, invertebrate genomes contain a single RyR-encoding gene and it has long been proposed that different transcripts generated by alternative splicing may diversify their functions. Here, we analyze the expression and function of alternative exons in the C. elegans RyR gene unc-68. We show that specific isoform subsets are created via alternative promoters and via alternative splicing in unc-68 Divergent Region 2 (DR2), which actually corresponds to a region of high sequence variability across vertebrate isoforms. The expression of specific unc-68 alternative exons is enriched in different tissues, such as in body wall muscle, neurons and pharyngeal muscle. In order to infer the function of specific alternative promoters and alternative exons of unc-68, we selectively deleted them by CRISPR/Cas9 genome editing. We evaluated pharyngeal function, as well as locomotor function in swimming and crawling with high-content computer-assisted postural and behavioral analysis. Our data provide a comprehensive map of the pleiotropic impact of isoform-specific mutations and highlight that tissue-specific unc-68 isoforms fulfill distinct functions. As a whole, our work clarifies how the C. elegans single RyR gene unc-68 can fulfill multiple tasks through tissue-specific isoforms, and provide a solid foundation to further develop C. elegans as a model to study RyR channel functions and malfunctions.


Asunto(s)
Empalme Alternativo/genética , Proteínas de Caenorhabditis elegans/genética , Caenorhabditis elegans/genética , Contracción Muscular/genética , Canal Liberador de Calcio Receptor de Rianodina/genética , Animales , Animales Modificados Genéticamente/crecimiento & desarrollo , Caenorhabditis elegans/crecimiento & desarrollo , Señalización del Calcio/genética , Modelos Animales de Enfermedad , Exones , Humanos , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Mutación/genética , Especificidad de Órganos/genética , Isoformas de Proteínas/genética , Rianodina/metabolismo
3.
PLoS Genet ; 15(12): e1008509, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31891575

RESUMEN

Understanding how the nervous system bridges sensation and behavior requires the elucidation of complex neural and molecular networks. Forward genetic approaches, such as screens conducted in C. elegans, have successfully identified genes required to process natural sensory stimuli. However, functional redundancy within the underlying neural circuits, which are often organized with multiple parallel neural pathways, limits our ability to identify 'neural pathway-specific genes', i.e. genes that are essential for the function of some, but not all of these redundant neural pathways. To overcome this limitation, we developed a 'forward optogenetics' screening strategy in which natural stimuli are initially replaced by the selective optogenetic activation of a specific neural pathway. We used this strategy to address the function of the polymodal FLP nociceptors mediating avoidance of noxious thermal and mechanical stimuli. According to our expectations, we identified both mutations in 'general' avoidance genes that broadly impact avoidance responses to a variety of natural noxious stimuli (unc-4, unc-83, and eat-4) and mutations that produce a narrower impact, more restricted to the FLP pathway (syd-2, unc-14 and unc-68). Through a detailed follow-up analysis, we further showed that the Ryanodine receptor UNC-68 acts cell-autonomously in FLP to adjust heat-evoked calcium signals and aversive behaviors. As a whole, our work (i) reveals the importance of properly regulated ER calcium release for FLP function, (ii) provides new entry points for new nociception research and (iii) demonstrates the utility of our forward optogenetic strategy, which can easily be transposed to analyze other neural pathways.


Asunto(s)
Reacción de Prevención , Proteínas de Caenorhabditis elegans/genética , Caenorhabditis elegans/fisiología , Optogenética/métodos , Animales , Caenorhabditis elegans/genética , Señalización del Calcio , Proteínas del Citoesqueleto/genética , Regulación de la Expresión Génica , Proteínas de Homeodominio/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Proteínas de la Membrana/genética , Proteínas Nucleares/genética , Receptores de Glutamato/genética , Canal Liberador de Calcio Receptor de Rianodina/genética , Proteínas de Transporte Vesicular de Glutamato
4.
J Neurosci ; 38(27): 6114-6129, 2018 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-29875264

RESUMEN

The ability to adapt behavior to environmental fluctuations is critical for survival of organisms ranging from invertebrates to mammals. Caenorhabditis elegans can learn to avoid sodium chloride when it is paired with starvation. This behavior may help animals avoid areas without food. Although some genes have been implicated in this salt-aversive learning behavior, critical genetic components, and the neural circuit in which they act, remain elusive. Here, we show that the sole worm ortholog of mammalian CaMKI/IV, CMK-1, is essential for salt-aversive learning behavior in C. elegans hermaphrodites. We find that CMK-1 acts in the primary salt-sensing ASE neurons to regulate this behavior. By characterizing the intracellular calcium dynamics in ASE neurons using microfluidics, we find that loss of cmk-1 has subtle effects on sensory-evoked calcium responses in ASE axons and their modulation by salt conditioning. Our study implicates the expression of the conserved CaMKI/CMK-1 in chemosensory neurons as a regulator of behavioral plasticity to environmental salt in C. elegansSIGNIFICANCE STATEMENT Like other animals, the nematode Caenorhabditis elegans depends on salt for survival and navigates toward high concentrations of this essential mineral. In addition to its role as an essential nutrient, salt also causes osmotic stress at high concentrations. A growing body of evidence indicates that C. elegans balances the requirement for salt with the danger it presents through a process called salt-aversive learning. We show that this behavior depends on expression of a calcium/calmodulin-dependent kinase, CMK-1, in the ASE salt-sensing neurons. Our study identifies CMK-1 and salt-sensitive chemosensory neurons as key factors in this form of behavioral plasticity.


Asunto(s)
Conducta Animal/fisiología , Proteínas de Caenorhabditis elegans/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Células Quimiorreceptoras/metabolismo , Quimiotaxis/fisiología , Aprendizaje/fisiología , Animales , Animales Modificados Genéticamente , Caenorhabditis elegans
5.
PLoS Genet ; 10(10): e1004718, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25340742

RESUMEN

Parkinson's disease (PD) is the most common neurodegenerative movement disorder characterized by the progressive loss of dopaminergic (DA) neurons. Both environmental and genetic factors are thought to contribute to the pathogenesis of PD. Although several genes linked to rare familial PD have been identified, endogenous risk factors for sporadic PD, which account for the majority of PD cases, remain largely unknown. Genome-wide association studies have identified many single nucleotide polymorphisms associated with sporadic PD in neurodevelopmental genes including the transcription factor p48/ptf1a. Here we investigate whether p48 plays a role in the survival of DA neurons in Drosophila melanogaster and Caenorhabditis elegans. We show that a Drosophila p48 homolog, 48-related-2 (Fer2), is expressed in and required for the development and survival of DA neurons in the protocerebral anterior medial (PAM) cluster. Loss of Fer2 expression in adulthood causes progressive PAM neuron degeneration in aging flies along with mitochondrial dysfunction and elevated reactive oxygen species (ROS) production, leading to the progressive locomotor deficits. The oxidative stress challenge upregulates Fer2 expression and exacerbates the PAM neuron degeneration in Fer2 loss-of-function mutants. hlh-13, the worm homolog of p48, is also expressed in DA neurons. Unlike the fly counterpart, hlh-13 loss-of-function does not impair development or survival of DA neurons under normal growth conditions. Yet, similar to Fer2, hlh-13 expression is upregulated upon an acute oxidative challenge and is required for the survival of DA neurons under oxidative stress in adult worms. Taken together, our results indicate that p48 homologs share a role in protecting DA neurons from oxidative stress and degeneration, and suggest that loss-of-function of p48 homologs in flies and worms provides novel tools to study gene-environmental interactions affecting DA neuron survival.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/biosíntesis , Proteínas de Caenorhabditis elegans/biosíntesis , Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón/genética , Estrés Oxidativo/genética , Enfermedad de Parkinson/genética , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/genética , Neuronas Dopaminérgicas/metabolismo , Drosophila melanogaster , Regulación de la Expresión Génica , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología
6.
PLoS Pathog ; 9(8): e1003524, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23950710

RESUMEN

Monepantel is a member of the recently identified class of anthelmintics known as the amino-acetonitrile derivatives (AADs). Monepantel controls all major gastro-intestinal nematodes in sheep including those that are resistant to the classical anthelmintics. Previous studies have shown that the Caenorhabditis elegans acr-23 and the Haemonchus contortus Hco-mptl-1 genes may be prominent targets of monepantel. With this discovery it became possible to investigate the mode of action of monepantel in nematodes at the molecular level. In the present study, we show that a C. elegans mutant acr-23 strain is fully rescued by expressing the wild-type acr-23 gene. Moreover, we present a new mutant allele, and characterize acr-23 alleles genetically. We also show that acr-23 is expressed in body wall muscle cells, and provide therefore a possible explanation for the paralysis caused by monepantel. Furthermore, genetic evidence suggests that the chaperone RIC-3 is required for expression of full monepantel resistance. Finally, we present reconstitution of the C. elegans ACR-23 receptor in Xenopus laevis oocytes and provide direct evidence of its modulation by monepantel. Conversely, co-injection of the chaperone RIC-3 had no impact for channel reconstitution in X. laevis oocytes. These results reinforce the involvement of the ACR-23 family in the mode of action of monepantel and advance our understanding of this new class of anthelmintics.


Asunto(s)
Aminoacetonitrilo/análogos & derivados , Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Resistencia a Medicamentos/fisiología , Canales Iónicos/metabolismo , Aminoacetonitrilo/farmacología , Animales , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Resistencia a Medicamentos/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Canales Iónicos/genética , Mutación , Especificidad de Órganos/efectos de los fármacos , Especificidad de Órganos/genética , Xenopus laevis
7.
BMC Genomics ; 15: 364, 2014 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-24884695

RESUMEN

BACKGROUND: Alternative splicing diversifies the pool of messenger RNA molecules encoded by individual genes. This diversity is particularly high when multiple splicing decisions cause a combinatorial arrangement of several alternate exons. We know very little on how the multiple decisions occurring during the maturation of single transcripts are coordinated and whether specific sequence elements might be involved. RESULTS: Here, the Caenorhabditis elegans genome was surveyed in order to identify sequence elements that might play a specific role in the regulation of multiple splicing decisions. The introns flanking alternate exons in transcripts whose maturation involves multiple alternative splicing decisions were compared to those whose maturation involves a single decision. Fifty-eight penta-, hexa-, and hepta-meric elements, clustered in 17 groups, were significantly over-represented in genes subject to multiple alternative splicing decisions. Most of these motifs relate to known splicing regulatory elements and appear to be well conserved in the related species Caenorhabditis briggsae. The usage of specific motifs is not linked to the gene product function, but rather depends on the gene structure, since it is influenced by the distance separating the multiple splicing decision sites. Two of these motifs are part of the CeRep25B minisatellite, which is also over-represented at the vicinity of alternative splicing regions. Most of the remaining motifs are not part of repeated sequence elements, but tend to occur in specific heterologous pairs in genes subject to multiple alternative splicing decisions. CONCLUSIONS: The existence of specific intronic sequence elements linked to multiple alternative splicing decisions is intriguing and suggests that these elements might have some specialized regulatory role during splicing.


Asunto(s)
Empalme Alternativo/genética , Caenorhabditis elegans/genética , Animales , Secuencia de Bases , Caenorhabditis/genética , Proteínas de Caenorhabditis elegans/química , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Análisis por Conglomerados , Secuencia Conservada , Exones , Genoma , Intrones , Repeticiones de Minisatélite , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo
8.
Proc Natl Acad Sci U S A ; 108(51): 20790-5, 2011 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-22084100

RESUMEN

Alternative splicing is critical for diversifying eukaryotic proteomes, but the rules governing and coordinating splicing events among multiple alternate splice sites within individual genes are not well understood. We developed a quantitative PCR-based strategy to quantify the expression of the 12 transcripts encoded by the Caenorhabditis elegans slo-1 gene, containing three alternate splice sites. Using conditional probability-based models, we show that splicing events are coordinated across these sites. Further, we identify a point mutation in an intron adjacent to one alternate splice site that disrupts alternative splicing at all three sites. This mutation leads to aberrant synaptic transmission at the neuromuscular junction. In a genomic survey, we found that a UAAAUC element disrupted by this mutation is enriched in introns flanking alternate exons in genes with multiple alternate splice sites. These results establish that proper coordination of intragenic alternative splicing is essential for normal physiology of slo-1 in vivo and identify putative specialized cis-regulatory elements that regulate the coordination of intragenic alternative splicing.


Asunto(s)
Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/fisiología , Regulación de la Expresión Génica , Canales de Potasio de Gran Conductancia Activados por el Calcio/genética , Canales de Potasio de Gran Conductancia Activados por el Calcio/fisiología , Alelos , Empalme Alternativo , Animales , Conducta Animal , Caenorhabditis elegans , Exones , Genómica , Intrones , Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Modelos Genéticos , Mutación , Reacción en Cadena de la Polimerasa , Sitios de Empalme de ARN
9.
Proc Natl Acad Sci U S A ; 108(51): 20784-9, 2011 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-22049343

RESUMEN

Most human genes contain multiple alternative splice sites believed to extend the complexity and diversity of the proteome. However, little is known about how interactions among alternative exons regulate protein function. We used the Caenorhabditis elegans slo-1 large-conductance calcium and voltage-activated potassium (BK) channel gene, which contains three alternative splice sites (A, B, and C) and encodes at least 12 splice variants, to investigate the functional consequences of alternative splicing. These splice sites enable the insertion of exons encoding part of the regulator of K(+) conductance (RCK)1 Ca(2+) coordination domain (exons A1 and A2) and portions of the RCK1-RCK2 linker (exons B0, B1, B2, C0, and C1). Exons A1 and A2 are used in a mutually exclusive manner and are 67% identical. The other exons can extend the RCK1-RCK2 linker by up to 41 residues. Electrophysiological recordings of all isoforms show that the A1 and A2 exons regulate activation kinetics and Ca(2+) sensitivity, but only if alternate exons are inserted at site B or C. Thus, RCK1 interacts with the RCK1-RCK2 linker, and the effect of exon variation on gating depends on the combination of alternate exons present in each isoform.


Asunto(s)
Canales de Potasio de Gran Conductancia Activados por el Calcio/química , Empalme Alternativo , Animales , Biofisica/métodos , Caenorhabditis elegans , Electrofisiología/métodos , Exones , Variación Genética , Cinética , Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Modelos Genéticos , Oocitos/metabolismo , Estructura Terciaria de Proteína , Xenopus/metabolismo
10.
MicroPubl Biol ; 20242024.
Artículo en Inglés | MEDLINE | ID: mdl-38660564

RESUMEN

The quest for extending lifespan and promoting a healthy aging has been a longstanding pursuit in the field of aging research. The control of aging and age-related diseases by nitric oxide (NO) and cGMP signaling is a broadly conserved process from worms to human. Here we show that TOP-N53, a dual-acting NO donor and PDE5 inhibitor, can increase both lifespan and health span in C. elegans .

11.
Nature ; 448(7149): 92-6, 2007 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-17611542

RESUMEN

Oxygen-evolving photosynthetic organisms regulate carbon metabolism through a light-dependent redox signalling pathway. Electrons are shuttled from photosystem I by means of ferredoxin (Fdx) to ferredoxin-thioredoxin reductase (FTR), which catalyses the two-electron-reduction of chloroplast thioredoxins (Trxs). These modify target enzyme activities by reduction, regulating carbon flow. FTR is unique in its use of a [4Fe-4S] cluster and a proximal disulphide bridge in the conversion of a light signal into a thiol signal. We determined the structures of FTR in both its one- and its two-electron-reduced intermediate states and of four complexes in the pathway, including the ternary Fdx-FTR-Trx complex. Here we show that, in the first complex (Fdx-FTR) of the pathway, the Fdx [2Fe-2S] cluster is positioned suitably for electron transfer to the FTR [4Fe-4S] centre. After the transfer of one electron, an intermediate is formed in which one sulphur atom of the FTR active site is free to attack a disulphide bridge in Trx and the other sulphur atom forms a fifth ligand for an iron atom in the FTR [4Fe-4S] centre--a unique structure in biology. Fdx then delivers a second electron that cleaves the FTR-Trx heterodisulphide bond, which occurs in the Fdx-FTR-Trx complex. In this structure, the redox centres of the three proteins are aligned to maximize the efficiency of electron transfer from the Fdx [2Fe-2S] cluster to the active-site disulphide of Trxs. These results provide a structural framework for understanding the mechanism of disulphide reduction by an iron-sulphur enzyme and describe previously unknown interaction networks for both Fdx and Trx (refs 4-6).


Asunto(s)
Ferredoxinas/química , Proteínas Hierro-Azufre/química , Oxidorreductasas/química , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Dominio Catalítico , Cristalografía por Rayos X , Transporte de Electrón , Ferredoxinas/metabolismo , Hierro/química , Proteínas Hierro-Azufre/metabolismo , Modelos Moleculares , Oxidación-Reducción , Oxidorreductasas/metabolismo , Proteínas de Plantas/química , Proteínas de Plantas/metabolismo , Conformación Proteica , Proteínas Recombinantes , Spinacia oleracea , Relación Estructura-Actividad , Synechocystis , Tiorredoxinas/química , Tiorredoxinas/metabolismo
12.
Elife ; 122023 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-37166173

RESUMEN

Nociceptive habituation is a conserved process through which pain sensitivity threshold is adjusted based on past sensory experience and which may be dysregulated in human chronic pain conditions. Noxious heat habituation in Caenorhabditis elegans involves the nuclear translocation of CaM kinase-1 (CMK-1) in the FLP thermo-nociceptors neurons, causing reduced animal heat sensitivity and avoidance responses. The phosphorylation of CMK-1 on T179 by CaM kinase kinase-1 (CKK-1) is required for nuclear entry. Recently, we identified a specific nuclear export sequence (NES) required to maintain CMK-1 in the cytoplasm at rest (20°C) and showed that Ca2+/CaM binding is sufficient to enhance CMK-1 affinity for IMA-3 via a specific nuclear localization signal (NLS) in order to promote nuclear entry after persistent heat stimulation (90 min at 28°C) (Ippolito et al., 2021). Here, we identified additional functional NES and NLS on CMK-1, whose activity can counteract previously identified elements. Furthermore, we clarify the relationship between the CaM-binding-dependent and T179-dependent effects. T179 phosphorylation can promote nuclear entry both downstream of CaM binding and as part of an independent/parallel pathway. Moreover, T179 phosphorylation can also produce the opposite effect by promoting nuclear export. Taken together, our studies suggest that multiple calcium-dependent regulatory mechanisms converge to bias the activity pattern across a network of NES/NLS elements, in order to control CMK-1 nucleo-cytoplasmic shuttling, and actuate stimulation-dependent nociceptive plasticity.


Asunto(s)
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina , Calcio , Animales , Transporte Activo de Núcleo Celular , Caenorhabditis elegans/metabolismo , Calcio/metabolismo , Núcleo Celular/metabolismo , Nociceptores/metabolismo , Señales de Localización Nuclear/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo
13.
Genetics ; 224(1)2023 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-36947448

RESUMEN

The detection and avoidance of harmful stimuli are essential animal capabilities. The molecular and cellular mechanisms controlling nociception and its plasticity are conserved, genetically controlled processes of broad biomedical interest given their relevance to understand and treat pain conditions that represent a major health burden. Recent genome-wide association studies (GWAS) have identified a rich set of polymorphisms related to different pain conditions and pointed to many human pain gene candidates, whose connection to the pain pathways is however often poorly understood. Here, we used a computer-assisted Caenorhabditis elegans thermal avoidance analysis pipeline to screen for behavioral defects in a set of 109 mutants for genes orthologous to human pain-related genes. We measured heat-evoked reversal thermosensitivity profiles, as well as spontaneous reversal rate, and compared naïve animals with adapted animals submitted to a series of repeated noxious heat stimuli, which in wild type causes a progressive habituation. Mutations affecting 28 genes displayed defects in at least one of the considered parameters and could be clustered based on specific phenotypic footprints, such as high-sensitivity mutants, nonadapting mutants, or mutants combining multiple defects. Collectively, our data reveal the functional architecture of a network of conserved pain-related genes in C. elegans and offer novel entry points for the characterization of poorly understood human pain genes in this genetic model.


Asunto(s)
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Animales , Humanos , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Nocicepción/fisiología , Estudio de Asociación del Genoma Completo , Dolor
14.
Nat Commun ; 14(1): 3052, 2023 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-37236963

RESUMEN

Maintaining or shifting between behavioral states according to context is essential for animals to implement fitness-promoting strategies. How the integration of internal state, past experience and sensory inputs orchestrates persistent multidimensional behavioral changes remains poorly understood. Here, we show that C. elegans integrates environmental temperature and food availability over different timescales to engage in persistent dwelling, scanning, global or glocal search strategies matching thermoregulatory and feeding needs. Transition between states, in each case, involves regulating multiple processes including AFD or FLP tonic sensory neurons activity, neuropeptide expression and downstream circuit responsiveness. State-specific FLP-6 or FLP-5 neuropeptide signaling acts on a distributed set of inhibitory GPCR(s) to promote scanning or glocal search, respectively, bypassing dopamine and glutamate-dependent behavioral state control. Integration of multimodal context via multisite regulation in sensory circuits might represent a conserved regulatory logic for a flexible prioritization on the valence of multiple inputs when operating persistent behavioral state transitions.


Asunto(s)
Proteínas de Caenorhabditis elegans , Neuropéptidos , Animales , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Células Receptoras Sensoriales/metabolismo , Neuropéptidos/metabolismo , Transducción de Señal
15.
Curr Opin Neurobiol ; 73: 102525, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35307612

RESUMEN

As small ectotherms, whose temperature equilibrates almost instantly with that of their environment, free-living nematodes rely on their behavior for thermoregulation. Caenorhabditis elegans has been extensively used as a model to address the fundamental mechanisms involved in thermosensation and the production of temperature-dependent behaviors. Behavioral responses include avoidance of acute noxious heat or cold stimuli and thermotactic responses to innocuous temperatures to produce oriented navigation in spatial thermogradients. In order to produce these behaviors, C. elegans relies on its ability to detect thermal cues with exquisite sensitivity, orchestrate a set of specific behavioral responses and adapt these responses in specific contexts, including according to past sensory experience and current internal states. The present review focuses on recent advances in our understanding of the processes occurring at the molecular, cellular, and circuit levels that enable thermosensory information processing and plasticity.


Asunto(s)
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Adaptación Fisiológica , Animales , Conducta Animal/fisiología , Caenorhabditis elegans/fisiología , Temperatura , Sensación Térmica/fisiología
16.
Elife ; 102021 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-34766550

RESUMEN

Sensory and behavioral plasticity are essential for animals to thrive in changing environments. As key effectors of intracellular calcium signaling, Ca2+/calmodulin-dependent protein kinases (CaMKs) can bridge neural activation with the many regulatory processes needed to orchestrate sensory adaptation, including by relaying signals to the nucleus. Here, we elucidate the molecular mechanism controlling the cell activation-dependent nuclear translocation of CMK-1, the Caenorhabditis elegans ortholog of mammalian CaMKI/IV, in thermosensory neurons in vivo. We show that an intracellular Ca2+ concentration elevation is necessary and sufficient to favor CMK-1 nuclear import. The binding of Ca2+/CaM to CMK-1 increases its affinity for IMA-3 importin, causing a redistribution with a relatively slow kinetics, matching the timescale of sensory adaptation. Furthermore, we show that this mechanism enables the encoding of opposite nuclear signals in neuron types with opposite calcium-responses and that it is essential for experience-dependent behavioral plasticity and gene transcription control in vivo. Since CaMKI/IV are conserved regulators of adaptable behaviors, similar mechanisms could exist in other organisms and for other sensory modalities.


Asunto(s)
Caenorhabditis elegans/fisiología , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Células Receptoras Sensoriales/metabolismo , Adaptación Fisiológica , Animales , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Señalización del Calcio , Núcleo Celular/metabolismo , Carioferinas/metabolismo , Sensación Térmica
17.
J Biol Methods ; 8(3): e152, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34514013

RESUMEN

DNA adenine methylation identification (DamID) is a powerful method to determine DNA binding profiles of proteins at a genomic scale. The method leverages the fusion between a protein of interest and the Dam methyltransferase of E. coli, which methylates proximal DNA in vivo. Here, we present an optimized procedure, which was developed for tissue-specific analyses in Caenorhabditis elegans and successfully used to footprint genes actively transcribed by RNA polymerases and to map transcription factor binding in gene regulatory regions. The present protocol details C. elegans-specific steps involved in the preparation of transgenic lines and genomic DNA samples, as well as broadly applicable steps for the DamID procedure, including the isolation of methylated DNA fragments, the preparation of multiplexed libraries, Nanopore sequencing, and data analysis. Two distinctive features of the approach are (i) the use of an efficient recombination-based strategy to selectively analyze rare cell types and (ii) the use of Nanopore sequencing, which streamlines the process. The method allows researchers to go from genomic DNA samples to sequencing results in less than a week, while being sensitive enough to report reliable DNA footprints in cell types as rare as 2 cells per animal.

18.
Sci Rep ; 11(1): 21766, 2021 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-34741086

RESUMEN

Light affects many physiological processes in mammals such as entrainment of the circadian clock, regulation of mood, and relaxation of blood vessels. At the molecular level, a stimulus such as light initiates a cascade of kinases that phosphorylate CREB at various sites, including serine 133 (S133). This modification leads CREB to recruit the co-factor CRCT1 and the histone acetyltransferase CBP to stimulate the transcription of genes containing a CRE element in their promoters, such as Period 1 (Per1). However, the details of this pathway are poorly understood. Here we provide evidence that PER2 acts as a co-factor of CREB to facilitate the formation of a transactivation complex on the CRE element of the Per1 gene regulatory region in response to light or forskolin. Using in vitro and in vivo approaches, we show that PER2 modulates the interaction between CREB and its co-regulator CRTC1 to support complex formation only after a light or forskolin stimulus. Furthermore, the absence of PER2 abolished the interaction between the histone acetyltransferase CBP and CREB. This process was accompanied by a reduction of histone H3 acetylation and decreased recruitment of RNA Pol II to the Per1 gene. Collectively, our data show that PER2 supports the stimulus-dependent induction of the Per1 gene via modulation of the CREB/CRTC1/CBP complex.


Asunto(s)
Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Regulación de la Expresión Génica/efectos de la radiación , Proteínas Circadianas Period/metabolismo , Acetilación , Animales , Cromatina/metabolismo , Masculino , Ratones , Proteínas Proto-Oncogénicas c-fos/metabolismo
19.
J Biol Methods ; 7(1): e129, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32313814

RESUMEN

Nociception and its plasticity are essential biological processes controlling adaptive behavioral responses in animals. These processes are also linked to different pain conditions in human and have received considerable attention, notably via studies in rodent models and the use of heat-evoked withdrawal behavior assays as a readout of unpleasant experience. More recently, invertebrates have also emerged as useful complementary models, with their own set of advantages, including their amenability to genetic manipulations, the accessibility and relative simplicity of their nervous system and ethical concerns linked to animal suffering. Like humans, the nematode Caenorhabditis elegans (C. elegans) can detect noxious heat and produce avoidance responses such as reversals. Here, we present a methodology suitable for the high-throughput analysis of C. elegans heat-evoked reversals and the adaptation to repeated stimuli. We introduce two platforms: the INFERNO (for infrared-evoked reversal analysis platform), allowing the quantification of the thermal sensitivity in a petri dish containing a large population (> 100 animals), and the ThermINATOR (for thermal adaptation multiplexed induction platform), allowing the mass-adaptation of up to 18 worm populations at the same time. We show that wild type animals progressively desensitize in response to repeated noxious heat pulses. Furthermore, analyzing the phenotype of mutant animals, we show that the mechanisms underlying baseline sensitivity and adaptation, respectively, are supported by genetically separable molecular pathways. In conclusion, the presented method enables the high-throughput evaluation of thermal avoidance in C. elegans and will contribute to accelerate studies in the field with this invertebrate model.

20.
Genetics ; 216(4): 931-945, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33037050

RESUMEN

Differential gene expression across cell types underlies development and cell physiology in multicellular organisms. Caenorhabditis elegans is a powerful, extensively used model to address these biological questions. A remaining bottleneck relates to the difficulty to obtain comprehensive tissue-specific gene transcription data, since available methods are still challenging to execute and/or require large worm populations. Here, we introduce the RNA Polymerase DamID (RAPID) approach, in which the Dam methyltransferase is fused to a ubiquitous RNA polymerase subunit to create transcriptional footprints via methyl marks on the DNA of transcribed genes. To validate the method, we determined the polymerase footprints in whole animals, in sorted embryonic blastomeres and in different tissues from intact young adults by driving tissue-specific Dam fusion expression. We obtained meaningful transcriptional footprints in line with RNA-sequencing (RNA-seq) studies in whole animals or specific tissues. To challenge the sensitivity of RAPID and demonstrate its utility to determine novel tissue-specific transcriptional profiles, we determined the transcriptional footprints of the pair of XXX neuroendocrine cells, representing 0.2% of the somatic cell content of the animals. We identified 3901 candidate genes with putatively active transcription in XXX cells, including the few previously known markers for these cells. Using transcriptional reporters for a subset of new hits, we confirmed that the majority of them were expressed in XXX cells and identified novel XXX-specific markers. Taken together, our work establishes RAPID as a valid method for the determination of RNA polymerase footprints in specific tissues of C. elegans without the need for cell sorting or RNA tagging.


Asunto(s)
Proteínas de Caenorhabditis elegans/genética , ARN Polimerasas Dirigidas por ADN/metabolismo , Perfilación de la Expresión Génica/métodos , Huella de Proteína/métodos , RNA-Seq/métodos , Metiltransferasa de ADN de Sitio Específico (Adenina Especifica)/metabolismo , Animales , Blastómeros/metabolismo , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/metabolismo , ARN Polimerasas Dirigidas por ADN/genética , Perfilación de la Expresión Génica/normas , Células Neuroendocrinas/metabolismo , Especificidad de Órganos , Huella de Proteína/normas , RNA-Seq/normas , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Metiltransferasa de ADN de Sitio Específico (Adenina Especifica)/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA