Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
PLoS Genet ; 19(1): e1010613, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36652499

RESUMO

Animals alter their behavior in manners that depend on environmental conditions as well as their developmental and metabolic states. For example, C. elegans is quiescent during larval molts or during conditions of satiety. By contrast, worms enter an exploration state when removed from food. Sensory perception influences movement quiescence (defined as a lack of body movement), as well as the expression of additional locomotor states in C. elegans that are associated with increased or reduced locomotion activity, such as roaming (exploration behavior) and dwelling (local search). Here we find that movement quiescence is enhanced, and exploration behavior is reduced in G protein-coupled receptor kinase grk-2 mutant animals. grk-2 was previously shown to act in chemosensation, locomotion, and egg-laying behaviors. Using neuron-specific rescuing experiments, we show that GRK-2 acts in multiple ciliated chemosensory neurons to control exploration behavior. grk-2 acts in opposite ways from the cGMP-dependent protein kinase gene egl-4 to control movement quiescence and exploration behavior. Analysis of mutants with defects in ciliated sensory neurons indicates that grk-2 and the cilium-structure mutants act in the same pathway to control exploration behavior. We find that GRK-2 controls exploration behavior in an opposite manner from the neuropeptide receptor NPR-1 and the neuropeptides FLP-1 and FLP-18. Finally, we show that secretion of the FLP-1 neuropeptide is negatively regulated by GRK-2 and that overexpression of FLP-1 reduces exploration behavior. These results define neurons and molecular pathways that modulate movement quiescence and exploration behavior.


Assuntos
Proteínas de Caenorhabditis elegans , Neuropeptídeos , Animais , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Células Receptoras Sensoriais/metabolismo , Locomoção/genética , Receptores Acoplados a Proteínas G/genética , Proteínas Quinases Dependentes de GMP Cíclico/genética , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo
2.
Hum Mol Genet ; 32(21): 3063-3077, 2023 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-37552066

RESUMO

Rab GTPases are important regulators of intracellular vesicular trafficking. RAB5C is a member of the Rab GTPase family that plays an important role in the endocytic pathway, membrane protein recycling and signaling. Here we report on 12 individuals with nine different heterozygous de novo variants in RAB5C. All but one patient with missense variants (n = 9) exhibited macrocephaly, combined with mild-to-moderate developmental delay. Patients with loss of function variants (n = 2) had an apparently more severe clinical phenotype with refractory epilepsy and intellectual disability but a normal head circumference. Four missense variants were investigated experimentally. In vitro biochemical studies revealed that all four variants were damaging, resulting in increased nucleotide exchange rate, attenuated responsivity to guanine exchange factors and heterogeneous effects on interactions with effector proteins. Studies in C. elegans confirmed that all four variants were damaging in vivo and showed defects in endocytic pathway function. The variant heterozygotes displayed phenotypes that were not observed in null heterozygotes, with two shown to be through a dominant negative mechanism. Expression of the human RAB5C variants in zebrafish embryos resulted in defective development, further underscoring the damaging effects of the RAB5C variants. Our combined bioinformatic, in vitro and in vivo experimental studies and clinical data support the association of RAB5C missense variants with a neurodevelopmental disorder characterized by macrocephaly and mild-to-moderate developmental delay through disruption of the endocytic pathway.


Assuntos
Deficiência Intelectual , Megalencefalia , Transtornos do Neurodesenvolvimento , Animais , Humanos , Criança , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Caenorhabditis elegans/metabolismo , Transtornos do Neurodesenvolvimento/genética , Deficiência Intelectual/genética , Fenótipo , Proteínas rab de Ligação ao GTP/genética , Proteínas rab de Ligação ao GTP/metabolismo , Megalencefalia/genética , Deficiências do Desenvolvimento/genética , Mutação de Sentido Incorreto/genética , Proteínas rab5 de Ligação ao GTP/genética , Proteínas rab5 de Ligação ao GTP/metabolismo
3.
Nat Rev Neurosci ; 20(2): 109-116, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30573905

RESUMO

During sleep, animals do not eat, reproduce or forage. Sleeping animals are vulnerable to predation. Yet, the persistence of sleep despite evolutionary pressures, and the deleterious effects of sleep deprivation, indicate that sleep serves a function or functions that cannot easily be bypassed. Recent research demonstrates sleep to be phylogenetically far more pervasive than previously appreciated; it is possible that the very first animals slept. Here, we give an overview of sleep across various species, with the aim of determining its original purpose. Sleep exists in animals without cephalized nervous systems and can be influenced by non-neuronal signals, including those associated with metabolic rhythms. Together, these observations support the notion that sleep serves metabolic functions in neural and non-neural tissues.


Assuntos
Filogenia , Sono/fisiologia , Animais , Evolução Biológica , Humanos , Fases do Sono/fisiologia , Especificidade da Espécie
4.
PLoS Biol ; 18(4): e3000220, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32315298

RESUMO

Many lines of evidence point to links between sleep regulation and energy homeostasis, but mechanisms underlying these connections are unknown. During Caenorhabditis elegans sleep, energetic stores are allocated to nonneural tasks with a resultant drop in the overall fat stores and energy charge. Mutants lacking KIN-29, the C. elegans homolog of a mammalian Salt-Inducible Kinase (SIK) that signals sleep pressure, have low ATP levels despite high-fat stores, indicating a defective response to cellular energy deficits. Liberating energy stores corrects adiposity and sleep defects of kin-29 mutants. kin-29 sleep and energy homeostasis roles map to a set of sensory neurons that act upstream of fat regulation as well as of central sleep-controlling neurons, suggesting hierarchical somatic/neural interactions regulating sleep and energy homeostasis. Genetic interaction between kin-29 and the histone deacetylase hda-4 coupled with subcellular localization studies indicate that KIN-29 acts in the nucleus to regulate sleep. We propose that KIN-29/SIK acts in nuclei of sensory neuroendocrine cells to transduce low cellular energy charge into the mobilization of energy stores, which in turn promotes sleep.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/fisiologia , Proteínas Serina-Treonina Quinases/metabolismo , Sono/fisiologia , Trifosfato de Adenosina/metabolismo , Animais , Animais Geneticamente Modificados , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Núcleo Celular/metabolismo , Metabolismo Energético/genética , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Mutação , Células Neuroendócrinas/metabolismo , Proteínas Serina-Treonina Quinases/genética , Células Receptoras Sensoriais/metabolismo
5.
J Neurophysiol ; 128(2): 302-309, 2022 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-35730757

RESUMO

The nematode Caenorhabditis elegans uses rhythmic muscle contractions (pumps) of the pharynx, a tubular feeding organ, to filter, transport, and crush food particles. A number of feeding mutants have been identified, including those with slow pharyngeal pumping rate, weak muscle contraction, defective muscle relaxation, and defective grinding of bacteria. Many aspects of these pharyngeal behavioral defects and how they affect pharyngeal function are not well understood. For example, the behavioral deficits underlying inefficient particle transport in "slippery" mutants have been unclear. Here we use high-speed video microscopy to describe pharyngeal pumping behaviors and particle transport in wild-type animals and in feeding mutants. Different "slippery" mutants exhibit distinct defects including weak isthmus contraction, failure to trap particles in the anterior isthmus, and abnormal timing of contraction and relaxation in pharyngeal compartments. Our results show that multiple deficits in pharyngeal timing or contraction can cause defects in particle transport. NEW & NOTEWORTHY The nematode C. elegans uses rhythmic contractions of its pharynx (feeding organ) to filter, transport, and crush food bacteria. Genetic analyses have identified mutants with defective pharyngeal motions, but many details of these movements and how they affect feeding are poorly understood. We use high-speed video microscopy to describe pharyngeal pumping behaviors and particle transport in feeding mutants. We find that multiple deficits in pharyngeal timing or contraction can cause defects in particle transport.


Assuntos
Caenorhabditis elegans , Faringe , Animais , Caenorhabditis elegans/fisiologia , Comportamento Alimentar/fisiologia , Microscopia de Vídeo , Contração Muscular/fisiologia
6.
BMC Biol ; 19(1): 186, 2021 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-34517863

RESUMO

BACKGROUND: Gravity plays an important role in most life forms on Earth. Yet, a complete molecular understanding of sensing and responding to gravity is lacking. While there are anatomical differences among animals, there is a remarkable conservation across phylogeny at the molecular level. Caenorhabditis elegans is suitable for gene discovery approaches that may help identify molecular mechanisms of gravity sensing. It is unknown whether C. elegans can sense the direction of gravity. RESULTS: In aqueous solutions, motile C. elegans nematodes align their swimming direction with the gravity vector direction while immobile worms do not. The worms orient downward regardless of whether they are suspended in a solution less dense (downward sedimentation) or denser (upward sedimentation) than themselves. Gravitaxis is minimally affected by the animals' gait but requires sensory cilia and dopamine neurotransmission, as well as motility; it does not require genes that function in the body touch response. CONCLUSIONS: Gravitaxis is not mediated by passive forces such as non-uniform mass distribution or hydrodynamic effects. Rather, it is mediated by active neural processes that involve sensory cilia and dopamine. C. elegans provides a genetically tractable system to study molecular and neural mechanisms of gravity sensing.


Assuntos
Caenorhabditis elegans , Animais , Caenorhabditis elegans/genética , Dopamina , Gravitação , Sensação Gravitacional , Natação
7.
J Neurogenet ; 34(3-4): 427-429, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33446018

RESUMO

I review the history of sleep research in Caenorhabditis elegans, briefly introduce the four articles in this issue focused on worm sleep and propose future directions our field might take.


Assuntos
Caenorhabditis elegans/fisiologia , Sono/fisiologia , Animais , Caenorhabditis elegans/crescimento & desenvolvimento , Proteínas de Caenorhabditis elegans/fisiologia , Larva , Atividade Motora , Fatores de Transcrição/fisiologia
8.
Proc Natl Acad Sci U S A ; 112(12): 3606-11, 2015 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-25775552

RESUMO

The ability to orient oneself in response to environmental cues is crucial to the survival and function of diverse organisms. One such orientation behavior is the alignment of aquatic organisms with (negative rheotaxis) or against (positive rheotaxis) fluid current. The questions of whether low-Reynolds-number, undulatory swimmers, such as worms, rheotax and whether rheotaxis is a deliberate or an involuntary response to mechanical forces have been the subject of conflicting reports. To address these questions, we use Caenorhabditis elegans as a model undulatory swimmer and examine, in experiment and theory, the orientation of C. elegans in the presence of flow. We find that when close to a stationary surface the animal aligns itself against the direction of the flow. We elucidate for the first time to our knowledge the mechanisms of rheotaxis in worms and show that rheotaxis can be explained solely by mechanical forces and does not require sensory input or deliberate action. The interaction between the flow field induced by the swimmer and a nearby surface causes the swimmer to tilt toward the surface and the velocity gradient associated with the flow rotates the animal to face upstream. Fluid mechanical computer simulations faithfully mimic the behavior observed in experiments, supporting the notion that rheotaxis behavior can be fully explained by hydrodynamics. Our study highlights the important role of hydrodynamics in the behavior of small undulating swimmers and may assist in developing control strategies to affect the animals' life cycles.


Assuntos
Comportamento Animal , Caenorhabditis elegans/fisiologia , Orientação , Animais , Ecossistema , Hidrodinâmica , Microfluídica , Reologia , Natação , Temperatura
9.
J Physiol ; 595(16): 5415-5424, 2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28028818

RESUMO

During health, animal sleep is regulated by an internal clock and by the duration of prior wakefulness. During sickness, sleep is regulated by cytokines released from either peripheral cells or from cells within the nervous system. These cytokines regulate central nervous system neurons to induce sleep. Recent research in the invertebrates Caenorhabditis elegans and Drosophila melanogaster has led to new insights into the mechanism of sleep during sickness. Sickness is triggered by exposure to environments such as infection, heat, or ultraviolet light irradiation, all of which cause cellular stress. Epidermal growth factor is released from stressed cells and signals to activate central neuroendocrine cell(s). These neuron(s) release neuropeptides including those containing an amidated arginine(R)-phenylalanine(F) motif at their C-termini (RFamide peptides). Importantly, mechanisms regulating sickness sleep are partially distinct from those regulating healthy sleep. We will here review key findings that have elucidated the central neuroendocrine mechanism of sleep during sickness. Adaptive mechanisms employed in the control of sickness sleep may play a role in correcting cellular homeostasis after various insults. We speculate that these mechanisms may play a maladaptive role in human pathological conditions such as in the fatigue and anorexia associated with autoimmune diseases, with major depression, and with unexplained chronic fatigue.


Assuntos
Doença , Invertebrados , Sono/fisiologia , Animais , Citocinas/metabolismo , Humanos
10.
Proc Natl Acad Sci U S A ; 111(19): 6865-70, 2014 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-24778261

RESUMO

Collective motion is observed in swarms of swimmers of various sizes, ranging from self-propelled nanoparticles to fish. The mechanisms that govern interactions among individuals are debated, and vary from one species to another. Although the interactions among relatively large animals, such as fish, are controlled by their nervous systems, the interactions among microorganisms, which lack nervous systems, are controlled through physical and chemical pathways. Little is known, however, regarding the mechanism of collective movements in microscopic organisms with nervous systems. To attempt to remedy this, we studied collective swimming behavior in the nematode Caenorhabditis elegans, a microorganism with a compact nervous system. We evaluated the contributions of hydrodynamic forces, contact forces, and mechanosensory input to the interactions among individuals. We devised an experiment to examine pair interactions as a function of the distance between the animals and observed that gait synchronization occurred only when the animals were in close proximity, independent of genes required for mechanosensation. Our measurements and simulations indicate that steric hindrance is the dominant factor responsible for motion synchronization in C. elegans, and that hydrodynamic interactions and genotype do not play a significant role. We infer that a similar mechanism may apply to other microscopic swimming organisms and self-propelled particles.


Assuntos
Comportamento Animal/fisiologia , Caenorhabditis elegans/fisiologia , Marcha , Modelos Biológicos , Natação/fisiologia , Animais , Fenômenos Biomecânicos , Hidrodinâmica
11.
Proc Natl Acad Sci U S A ; 111(28): 10167-72, 2014 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-24982160

RESUMO

Bacteriophytochromes sense light in the near-infrared window, the spectral region where absorption by mammalian tissues is minimal, and their chromophore, biliverdin IXα, is naturally present in animal cells. These properties make bacteriophytochromes particularly attractive for optogenetic applications. However, the lack of understanding of how light-induced conformational changes control output activities has hindered engineering of bacteriophytochrome-based optogenetic tools. Many bacteriophytochromes function as homodimeric enzymes, in which light-induced conformational changes are transferred via α-helical linkers to the rigid output domains. We hypothesized that heterologous output domains requiring homodimerization can be fused to the photosensory modules of bacteriophytochromes to generate light-activated fusions. Here, we tested this hypothesis by engineering adenylate cyclases regulated by light in the near-infrared spectral window using the photosensory module of the Rhodobacter sphaeroides bacteriophytochrome BphG1 and the adenylate cyclase domain from Nostoc sp. CyaB1. We engineered several light-activated fusion proteins that differed from each other by approximately one or two α-helical turns, suggesting that positioning of the output domains in the same phase of the helix is important for light-dependent activity. Extensive mutagenesis of one of these fusions resulted in an adenylate cyclase with a sixfold photodynamic range. Additional mutagenesis produced an enzyme with a more stable photoactivated state. When expressed in cholinergic neurons in Caenorhabditis elegans, the engineered adenylate cyclase affected worm behavior in a light-dependent manner. The insights derived from this study can be applied to the engineering of other homodimeric bacteriophytochromes, which will further expand the optogenetic toolset.


Assuntos
Adenilil Ciclases/biossíntese , Proteínas de Bactérias/biossíntese , Caenorhabditis elegans/metabolismo , Expressão Gênica , Raios Infravermelhos , Engenharia de Proteínas , Proteínas Recombinantes de Fusão/biossíntese , Adenilil Ciclases/genética , Animais , Animais Geneticamente Modificados , Proteínas de Bactérias/genética , Caenorhabditis elegans/genética , Escherichia coli/genética , Escherichia coli/metabolismo , Nostoc/genética , Nostoc/metabolismo , Estrutura Secundária de Proteína , Proteínas Recombinantes de Fusão/genética , Rhodobacter sphaeroides/genética , Rhodobacter sphaeroides/metabolismo , Synechocystis/genética , Synechocystis/metabolismo
12.
J Neurosci ; 35(43): 14571-84, 2015 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-26511247

RESUMO

Electrophysiological recordings have enabled identification of physiologically distinct yet behaviorally similar states of mammalian sleep. In contrast, sleep in nonmammals has generally been identified behaviorally and therefore regarded as a physiologically uniform state characterized by quiescence of feeding and locomotion, reduced responsiveness, and rapid reversibility. The nematode Caenorhabditis elegans displays sleep-like quiescent behavior under two conditions: developmentally timed quiescence (DTQ) occurs during larval transitions, and stress-induced quiescence (SIQ) occurs in response to exposure to cellular stressors. Behaviorally, DTQ and SIQ appear identical. Here, we use optogenetic manipulations of neuronal and muscular activity, pharmacology, and genetic perturbations to uncover circuit and molecular mechanisms of DTQ and SIQ. We find that locomotion quiescence induced by DTQ- and SIQ-associated neuropeptides occurs via their action on the nervous system, although their neuronal target(s) and/or molecular mechanisms likely differ. Feeding quiescence during DTQ results from a loss of pharyngeal muscle excitability, whereas feeding quiescence during SIQ results from a loss of excitability in the nervous system. Together these results indicate that, as in mammals, quiescence is subserved by different mechanisms during distinct sleep-like states in C. elegans.


Assuntos
Caenorhabditis elegans/fisiologia , Sono/fisiologia , Torpor/fisiologia , Animais , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/fisiologia , Comportamento Alimentar/fisiologia , Larva/crescimento & desenvolvimento , Larva/fisiologia , Locomoção/fisiologia , Músculos/fisiologia , Rede Nervosa/fisiologia , Vias Neurais/crescimento & desenvolvimento , Vias Neurais/fisiologia , Neurônios/fisiologia , Neuropeptídeos/fisiologia , Optogenética , Músculos Faríngeos/inervação , Músculos Faríngeos/fisiologia , Estresse Fisiológico
14.
Brain Behav Immun ; 47: 141-8, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25668617

RESUMO

Enhanced sleep in response to cellular stress is a conserved adaptive behavior across multiple species, but the mechanism of this process is poorly understood. Drosophila melanogaster increases sleep following exposure to septic or aseptic injury, and Caenorhabditis elegans displays sleep-like quiescence following exposure to high temperatures that stress cells. We show here that, similar to C. elegans, Drosophila responds to heat stress with an increase in sleep. In contrast to Drosophila infection-induced sleep, heat-induced sleep is not sensitive to the time-of-day of the heat pulse. Moreover, the sleep response to heat stress does not require Relish, the NFκB transcription factor that is necessary for infection-induced sleep, indicating that sleep is induced by multiple mechanisms from different stress modalities. We identify a sleep-regulating role for a signaling pathway involving FMRFamide neuropeptides and their receptor FR. Animals mutant for either FMRFamide or for the FMRFamide receptor (FR) have a reduced recovery sleep in response to heat stress. FR mutants, in addition, show reduced sleep responses following infection with Serratia marcescens, and succumb to infection at a faster rate than wild-type controls. Together, these findings support the hypothesis that FMRFamide and its receptor promote an adaptive increase in sleep following stress. Because an FMRFamide-like neuropeptide plays a similar role in C. elegans, we propose that FRMFamide neuropeptide signaling is an ancient regulator of recovery sleep which occurs in response to cellular stress.


Assuntos
FMRFamida/metabolismo , Receptores de Peptídeos de Invertebrados/metabolismo , Sono/fisiologia , Estresse Fisiológico/fisiologia , Animais , Animais Geneticamente Modificados , Drosophila , FMRFamida/genética , Temperatura Alta , Receptores de Peptídeos de Invertebrados/genética , Transdução de Sinais
15.
Methods ; 68(3): 500-7, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-24642199

RESUMO

The nematode Caenorhabditis (C.) elegans, a long time work horse for behavioral genetic studies of locomotion, has recently been studied for quiescent behavior. Methods previously established for the study of C. elegans locomotion are not well-suited for the study of quiescent behavior. We describe in detail two computer vision approaches to distinguish quiescent from movement bouts focusing on the behavioral quiescence that occurs during fourth larval stage lethargus, a transition stage between the larva and the adult. The first is the frame subtraction method, which consists of subtraction of temporally adjacent images as a sensitive way to detect motion. The second, which is more computationally intensive, is the posture analysis method, which consists of analysis of the rate of local angle change of the animal's body. Quiescence measurements should be done continuously while minimizing sensory perturbation of the animal.


Assuntos
Comportamento Animal/fisiologia , Caenorhabditis elegans/genética , Locomoção/fisiologia , Animais , Caenorhabditis elegans/fisiologia , Larva/genética , Larva/fisiologia , Locomoção/genética
16.
J Neurophysiol ; 112(4): 951-61, 2014 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-24872529

RESUMO

Degenerate networks, in which structurally distinct elements can perform the same function or yield the same output, are ubiquitous in biology. Degeneracy contributes to the robustness and adaptability of networks in varied environmental and evolutionary contexts. However, how degenerate neural networks regulate behavior in vivo is poorly understood, especially at the genetic level. Here, we identify degenerate neural and genetic mechanisms that underlie excitation of the pharynx (feeding organ) in the nematode Caenorhabditis elegans using cell-specific optogenetic excitation and inhibition. We show that the pharyngeal neurons MC, M2, M4, and I1 form multiple direct and indirect excitatory pathways in a robust network for control of pharyngeal pumping. I1 excites pumping via MC and M2 in a state-dependent manner. We identify nicotinic and muscarinic receptors through which the pharyngeal network regulates feeding rate. These results identify two different mechanisms by which degeneracy is manifest in a neural circuit in vivo.


Assuntos
Caenorhabditis elegans/fisiologia , Comportamento Alimentar , Rede Nervosa/citologia , Neurônios/fisiologia , Animais , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Gânglios dos Invertebrados/citologia , Gânglios dos Invertebrados/fisiologia , Rede Nervosa/fisiologia , Neurônios/metabolismo , Optogenética , Faringe/inervação , Faringe/fisiologia , Receptores Muscarínicos/metabolismo , Receptores Nicotínicos/metabolismo
17.
Nature ; 451(7178): 569-72, 2008 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-18185515

RESUMO

There are fundamental similarities between sleep in mammals and quiescence in the arthropod Drosophila melanogaster, suggesting that sleep-like states are evolutionarily ancient. The nematode Caenorhabditis elegans also has a quiescent behavioural state during a period called lethargus, which occurs before each of the four moults. Like sleep, lethargus maintains a constant temporal relationship with the expression of the C. elegans Period homologue LIN-42 (ref. 5). Here we show that quiescence associated with lethargus has the additional sleep-like properties of reversibility, reduced responsiveness and homeostasis. We identify the cGMP-dependent protein kinase (PKG) gene egl-4 as a regulator of sleep-like behaviour, and show that egl-4 functions in sensory neurons to promote the C. elegans sleep-like state. Conserved effects on sleep-like behaviour of homologous genes in C. elegans and Drosophila suggest a common genetic regulation of sleep-like states in arthropods and nematodes. Our results indicate that C. elegans is a suitable model system for the study of sleep regulation. The association of this C. elegans sleep-like state with developmental changes that occur with larval moults suggests that sleep may have evolved to allow for developmental changes.


Assuntos
Caenorhabditis elegans/fisiologia , Sono/fisiologia , Animais , Nível de Alerta/genética , Nível de Alerta/fisiologia , Evolução Biológica , Caenorhabditis elegans/enzimologia , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Sequência Conservada/genética , Proteínas Quinases Dependentes de GMP Cíclico/genética , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/fisiologia , Homeostase/fisiologia , Larva/fisiologia , Letargia , Muda/fisiologia , Sono/genética
18.
iScience ; 27(4): 109477, 2024 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-38551003

RESUMO

Structural neuroplasticity (changes in the size, strength, number, and targets of synaptic connections) can be modified by sleep and sleep disruption. However, the causal relationships between genetic perturbations, sleep loss, neuroplasticity, and behavior remain unclear. The C. elegans GABAergic DVB neuron undergoes structural plasticity in adult males in response to adolescent stress, which rewires synaptic connections, alters behavior, and is dependent on conserved autism-associated genes NRXN1/nrx-1 and NLGN3/nlg-1. We find that four methods of sleep deprivation transiently induce DVB neurite extension in day 1 adults and increase the time to spicule protraction, which is the functional and behavioral output of the DVB neuron. Loss of nrx-1 and nlg-1 prevent DVB structural plasticity and behavioral changes at day 1 caused by adolescent sleep loss. Therefore, nrx-1 and nlg-1 mediate the morphologic and behavioral consequences of sleep loss, providing insight into the relationship between sleep, neuroplasticity, behavior, and neurologic disease.

19.
Cell Metab ; 7(3): 249-57, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18316030

RESUMO

Despite the prevalence of obesity and its related diseases, the signaling pathways for appetite control and satiety are not clearly understood. Here we report C. elegans quiescence behavior, a cessation of food intake and movement that is possibly a result of satiety. C. elegans quiescence shares several characteristics of satiety in mammals. It is induced by high-quality food, it requires nutritional signals from the intestine, and it depends on prior feeding history: fasting enhances quiescence after refeeding. During refeeding after fasting, quiescence is evoked, causing gradual inhibition of food intake and movement, mimicking the behavioral sequence of satiety in mammals. Based on these similarities, we propose that quiescence results from satiety. This hypothesized satiety-induced quiescence is regulated by peptide signals such as insulin and TGF-beta. The EGL-4 cGMP-dependent protein kinase functions downstream of insulin and TGF-beta in sensory neurons including ASI to control quiescence in response to food intake.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , GMP Cíclico/metabolismo , Ingestão de Alimentos , Insulina/metabolismo , Locomoção , Resposta de Saciedade , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Fenômenos Fisiológicos da Nutrição Animal , Animais , Regulação do Apetite , Caenorhabditis elegans/enzimologia , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Jejum/metabolismo , Canais Iônicos/metabolismo , Modelos Animais , Mutação , Neurônios/metabolismo , Receptor de Insulina/metabolismo
20.
Front Neurol ; 14: 1135462, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37576014

RESUMO

Fatigue is a common symptom associated with cancer treatments. Brain mechanisms underlying cancer-related fatigue (CRF) and its progression following therapy are poorly understood. Previous studies have suggested a role of the default mode network (DMN) in fatigue. In this study we used arterial spin labeling (ASL) perfusion functional magnetic resonance imaging (fMRI) and compared resting cerebral blood flow (CBF) differences in the posterior cingulate cortex (PCC), a core hub of the DMN, between 16 patients treated with radiation therapy (RAT) for prostate (9 males) or breast (7 females) cancer and 18 healthy controls (HC). Resting CBF in patients was also measured immediately after the performance of a fatiguing 20-min psychomotor vigilance task (PVT). Twelve of 16 cancer patients were further followed between 3 and 7 months after completion of the RAT (post-RAT). Patients reported elevated fatigue on RAT in comparison to post-RAT, but no change in sleepiness, suggesting that the underlying neural mechanisms of CRF progression are distinct from those regulating sleep drive progression. Compared to HC, patients showed significantly increased resting CBF in the PCC and the elevated PCC CBF persisted during the follow up visit. Post-PVT, but not pre-PVT, resting CBF changes in the PCC correlated with fatigue changes after therapy in patients with CRF, suggesting that PCC CBF following a fatiguing cognitive task may be a biomarker for CRF recovery.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA