Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Immunity ; 57(2): 364-378.e9, 2024 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-38301651

RESUMO

Mutations of the CBP/p300 histone acetyltransferase (HAT) domain can be linked to leukemic transformation in humans, suggestive of a checkpoint of leukocyte compartment sizes. Here, we examined the impact of reversible inhibition of this domain by the small-molecule A485. We found that A485 triggered acute and transient mobilization of leukocytes from the bone marrow into the blood. Leukocyte mobilization by A485 was equally potent as, but mechanistically distinct from, granulocyte colony-stimulating factor (G-CSF), which allowed for additive neutrophil mobilization when both compounds were combined. These effects were maintained in models of leukopenia and conferred augmented host defenses. Mechanistically, activation of the hypothalamus-pituitary-adrenal gland (HPA) axis by A485 relayed shifts in leukocyte distribution through corticotropin-releasing hormone receptor 1 (CRHR1) and adrenocorticotropic hormone (ACTH), but independently of glucocorticoids. Our findings identify a strategy for rapid expansion of the blood leukocyte compartment via a neuroendocrine loop, with implications for the treatment of human pathologies.


Assuntos
Medula Óssea , Histona Acetiltransferases , Humanos , Histona Acetiltransferases/metabolismo , Medula Óssea/metabolismo , Histonas/metabolismo , Neutrófilos/metabolismo , Sistema Hipotálamo-Hipofisário/metabolismo
2.
PLoS Genet ; 19(3): e1010665, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36917595

RESUMO

Sleep is controlled by neurons that induce behavioral quiescence and physiological restoration. It is not known, however, how sleep neurons link sleep behavior and survival. In Caenorhabditis elegans, the sleep-active RIS neuron induces sleep behavior and is required for survival of starvation and wounding. Sleep-active neurons such as RIS might hypothetically promote survival primarily by causing sleep behavior and associated conservation of energy. Alternatively, RIS might provide a survival benefit that does not depend on behavioral sleep. To probe these hypotheses, we tested how activity of the sleep-active RIS neuron in Caenorhabditis elegans controls sleep behavior and survival during larval starvation. To manipulate the activity of RIS, we expressed constitutively active potassium channel (twk-18gf and egl-23gf) or sodium channel (unc-58gf) mutant alleles in this neuron. Low levels of unc-58gf expression in RIS increased RIS calcium transients and sleep. High levels of unc-58gf expression in RIS elevated baseline calcium activity and inhibited calcium activation transients, thus locking RIS activity at a high but constant level. This manipulation caused a nearly complete loss of sleep behavior but increased survival. Long-term optogenetic activation also caused constantly elevated RIS activity and a small trend towards increased survival. Disturbing sleep by lethal blue-light stimulation also overactivated RIS, which again increased survival. FLP-11 neuropeptides were important for both, induction of sleep behavior and starvation survival, suggesting that FLP-11 might have divergent roles downstream of RIS. These results indicate that promotion of sleep behavior and survival are separable functions of RIS. These two functions may normally be coupled but can be uncoupled during conditions of strong RIS activation or when sleep behavior is impaired. Through this uncoupling, RIS can provide survival benefits under conditions when behavioral sleep is disturbed. Promoting survival in the face of impaired sleep might be a general function of sleep neurons.


Assuntos
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Animais , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Cálcio/metabolismo , Neurônios/metabolismo , Sono/genética
3.
PLoS Biol ; 18(2): e3000361, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-32078631

RESUMO

Sleep-active neurons depolarize during sleep to suppress wakefulness circuits. Wake-active wake-promoting neurons in turn shut down sleep-active neurons, thus forming a bipartite flip-flop switch. However, how sleep is switched on is unclear because it is not known how wakefulness is translated into sleep-active neuron depolarization when the system is set to sleep. Using optogenetics in Caenorhabditis elegans, we solved the presynaptic circuit for depolarization of the sleep-active RIS neuron during developmentally regulated sleep, also known as lethargus. Surprisingly, we found that RIS activation requires neurons that have known roles in wakefulness and locomotion behavior. The RIM interneurons-which are active during and can induce reverse locomotion-play a complex role and can act as inhibitors of RIS when they are strongly depolarized and as activators of RIS when they are modestly depolarized. The PVC command interneurons, which are known to promote forward locomotion during wakefulness, act as major activators of RIS. The properties of these locomotion neurons are modulated during lethargus. The RIMs become less excitable. The PVCs become resistant to inhibition and have an increased capacity to activate RIS. Separate activation of neither the PVCs nor the RIMs appears to be sufficient for sleep induction; instead, our data suggest that they act in concert to activate RIS. Forward and reverse circuit activity is normally mutually exclusive. Our data suggest that RIS may be activated at the transition between forward and reverse locomotion states, perhaps when both forward (PVC) and reverse (including RIM) circuit activity overlap. While RIS is not strongly activated outside of lethargus, altered activity of the locomotion interneurons during lethargus favors strong RIS activation and thus sleep. The control of sleep-active neurons by locomotion circuits suggests that sleep control may have evolved from locomotion control. The flip-flop sleep switch in C. elegans thus requires an additional component, wake-active sleep-promoting neurons that translate wakefulness into the depolarization of a sleep-active neuron when the worm is sleepy. Wake-active sleep-promoting circuits may also be required for sleep state switching in other animals, including in mammals.


Assuntos
Locomoção/fisiologia , Neurônios/fisiologia , Fases do Sono/fisiologia , Vigília/fisiologia , Animais , Nível de Alerta/fisiologia , Comportamento Animal/fisiologia , Caenorhabditis elegans/fisiologia , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Cálcio/metabolismo , Homeostase , Interneurônios/metabolismo , Interneurônios/fisiologia , Larva/fisiologia , Vias Neurais/fisiologia , Neurônios/metabolismo , Optogenética
4.
EMBO Rep ; 20(3)2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30804011

RESUMO

Sleep is a fundamental conserved physiological state in animals and humans. It may serve multiple functions, ranging from energy conservation to higher brain operation. Understanding sleep functions and the underlying mechanisms requires the study of sleeplessness and its consequences. The traditional approach to remove sleep is sleep deprivation (SD) by sensory stimulation. However, stimulation-induced SD can be stressful and can cause non-specific side effects. An emerging alternative method is "genetic SD", which removes sleep using genetics or optogenetics. Sleep requires sleep-active neurons and their regulators. Thus, genetic impairment of sleep circuits might lead to more specific and comprehensive sleep loss. Here, I discuss the advantages and limits of genetic SD in key genetic sleep model animals: rodents, zebrafish, fruit flies and roundworms, and how the study of genetic SD alters our view of sleep functions. Genetic SD typically causes less severe phenotypes compared with stimulation-induced SD, suggesting that sensory stimulation-induced SD may have overestimated the role of sleep, calling for a re-investigation of sleep functions.


Assuntos
Mutação/genética , Privação do Sono/genética , Privação do Sono/fisiopatologia , Sono/fisiologia , Animais , Humanos , Modelos Animais , Neurônios/fisiologia , Optogenética
5.
J Neurogenet ; 34(3-4): 466-474, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32543249

RESUMO

Optogenetics controls neural activity and behavior in living organisms through genetically targetable actuators and light. This method has revolutionized biology and medicine as it allows controlling cells with high temporal and spatial precision. Optogenetics is typically applied only at short time scales, for instance to study specific behaviors. Optogenetically manipulating behavior also gives insights into physiology, as behavior controls systemic physiological processes. For example, arousal and sleep affect aging and health span. To study how behavior controls key physiological processes, behavioral manipulations need to occur at extended time scales. However, methods for long-term optogenetics are scarce and typically require expensive compound microscope setups. Optogenetic experiments can be conducted in many species. Small model animals such as the nematode C. elegans have been instrumental in solving the mechanistic basis of medically important biological processes. We developed the OptoGenBox, an affordable stand-alone and simple-to-use device for long-term optogenetic manipulation of C. elegans. The OptoGenBox provides a controlled environment and is programmable to allow the execution of complex optogenetic manipulations over long experimental times of many days to weeks. To test our device, we investigated how optogenetically increased arousal and optogenetic sleep deprivation affect survival of arrested first larval stage C. elegans. We optogenetically activated the nociceptive ASH sensory neurons using ReaChR, thus triggering an escape response and increase in arousal. In addition, we optogenetically inhibited the sleep neuron RIS using ArchT, a condition known to impair sleep. Both optogenetic manipulations reduced survival. Thus, the OptoGenBox presents an affordable system to study the long-term consequences of optogenetic manipulations of key biological processes in C. elegans and perhaps other small animals.


Assuntos
Caenorhabditis elegans/fisiologia , Optogenética/instrumentação , Animais , Nível de Alerta/fisiologia , Caenorhabditis elegans/crescimento & desenvolvimento , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/fisiologia , Desenho de Equipamento , Reação de Fuga/fisiologia , Larva , Longevidade , Neurônios/fisiologia , Nociceptores/fisiologia , Optogenética/métodos , Projetos de Pesquisa , Retinaldeído/farmacologia , Sono/fisiologia , Privação do Sono/fisiopatologia
6.
Genesis ; 54(4): 212-9, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26833569

RESUMO

Longitudinal analyses are crucial for understanding long-term processes such as development and behavioral rhythms. For a complete understanding of such processes, both organism-level observations as well as single-cell observations are necessary. Sleep is an example for a long-term process that is under developmental control. This behavioral state is induced by conserved sleep-active neurons, but little is known about how sleep neurons control the physiology of an animal systemically. In the nematode C. elegans, sleep induction crucially requires the single RIS interneuron to actively induce a developmentally regulated sleep behavior. Here, we used RIS-induced sleep as an example of how longitudinal analyses can be automated. We developed methods to analyze both behavior and neural activity in larva across the sleep-wake cycle. To image behavior, we used an improved DIC contrast to extract the head and detect the nose. To image neural activity, we used GCaMP3 expression in a small number of neurons including RIS combined with a neuron discrimination algorithm. Thus, we present a comprehensive platform for automatically analyzing behavior and neural activity in C. elegans exemplified by using RIS-induced sleep during C. elegans development.


Assuntos
Caenorhabditis elegans/fisiologia , Neurônios/fisiologia , Análise de Célula Única/métodos , Sono/fisiologia , Algoritmos , Animais , Automação Laboratorial , Comportamento Animal , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Interneurônios/metabolismo , Larva/fisiologia
7.
BMC Neurosci ; 17(1): 72, 2016 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-27829368

RESUMO

BACKGROUND: The AP-2 transcription factor APTF-1 is crucially required for developmentally controlled sleep behavior in Caenorhabditis elegans larvae. Its human ortholog, TFAP-2beta, causes Char disease and has also been linked to sleep disorders. These data suggest that AP-2 transcription factors may be highly conserved regulators of various types of sleep behavior. Here, we tested the idea that AP-2 controls adult sleep in Drosophila. RESULTS: Drosophila has one AP-2 ortholog called TfAP-2, which is essential for viability. To investigate its potential role in sleep behavior and neural development, we specifically downregulated TfAP-2 in the nervous system. We found that neuronal TfAP-2 knockdown almost completely abolished night sleep but did not affect day sleep. TfAP-2 insufficiency affected nervous system development. Conditional TfAP-2 knockdown in the adult also produced a modest sleep phenotype, suggesting that TfAP-2 acts both in larval as well as in differentiated neurons. CONCLUSIONS: Thus, our results show that AP-2 transcription factors are highly conserved regulators of development and sleep.


Assuntos
Proteínas de Drosophila/metabolismo , Sono/fisiologia , Fator de Transcrição AP-2/metabolismo , Animais , Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Encéfalo/patologia , Proteínas de Drosophila/genética , Drosophila melanogaster , Técnicas de Silenciamento de Genes , Imuno-Histoquímica , Masculino , Neurônios/metabolismo , Neurônios/patologia , Fotoperíodo , Filogenia , Reação em Cadeia da Polimerase em Tempo Real , Fator de Transcrição AP-2/genética , Gravação em Vídeo
8.
Nat Methods ; 8(3): 250-2, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21278743

RESUMO

We present a method to control protein levels under native genetic regulation in Caenorhabditis elegans by using synthetic genes with adapted codons. We found that the force acting on the spindle in C. elegans embryos was related to the amount of the G-protein regulator GPR-1/2. Codon-adapted versions of any C. elegans gene can be designed using our web tool, C. elegans codon adapter.


Assuntos
Proteínas de Caenorhabditis elegans/genética , Caenorhabditis elegans/genética , Códon/genética , Regulação da Expressão Gênica , Técnicas Genéticas , Biossíntese de Proteínas/genética , Animais , Divisão Celular/genética
9.
Development ; 137(10): 1743-53, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20430749

RESUMO

In asymmetrically dividing cells, a failure to coordinate cell polarity with the site of cell division can lead to cell fate transformations and tumorigenesis. Cell polarity in C. elegans embryos is defined by PAR proteins, which occupy reciprocal halves of the cell cortex. During asymmetric division, the boundary between the anterior and posterior PAR domains precisely matches the site of cell division, ensuring exclusive segregation of cell fate. The PAR domains determine the site of cell division by positioning the mitotic spindle, suggesting one means by which cell polarity and cell division might be coordinated. Here, we report that cell polarity and cell division are coordinated through an additional mechanism: the site of cell division repositions the PAR-2 boundary. Galpha-mediated microtubule-cortex interactions appear to direct cortical flows of PAR-2 and myosin toward the site of cell division, which acts as a PAR-2 and myosin sink. Embryos with defects in PAR-2 boundary correction undergo mis-segregation of cortical polarity and cytoplasmic determinants, suggesting that PAR domain correction might help prevent cell fate transformation.


Assuntos
Padronização Corporal/fisiologia , Caenorhabditis elegans/embriologia , Divisão Celular/fisiologia , Polaridade Celular/fisiologia , Citocinese/fisiologia , Animais , Animais Geneticamente Modificados , Caenorhabditis elegans/fisiologia , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/fisiologia , Fase de Clivagem do Zigoto/metabolismo , Fase de Clivagem do Zigoto/fisiologia , Corrente Citoplasmática/fisiologia , Embrião não Mamífero , Complexos Multiproteicos/metabolismo , Complexos Multiproteicos/fisiologia , Proteína Quinase C/metabolismo , Proteína Quinase C/fisiologia , Proteínas Serina-Treonina Quinases , Fuso Acromático/metabolismo
10.
Sci Rep ; 13(1): 8026, 2023 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-37198238

RESUMO

Sleep is a universal state of behavioral quiescence in both vertebrates and invertebrates that is controlled by conserved genes. We previously found that AP2 transcription factors control sleep in C. elegans, Drosophila, and mice. Heterozygous deletion of Tfap2b, one of the mammalian AP2 paralogs, reduces sleep in mice. The cell types and mechanisms through which Tfap2b controls sleep in mammals are, however, not known. In mice, Tfap2b acts during early embryonic stages. In this study, we used RNA-seq to measure the gene expression changes in brains of Tfap2b-/- embryos. Our results indicated that genes related to brain development and patterning were differentially regulated. As many sleep-promoting neurons are known to be GABAergic, we measured the expression of GAD1, GAD2 and Vgat genes in different brain areas of adult Tfap2b+/- mice using qPCR. These experiments suggested that GABAergic genes are downregulated in the cortex, brainstem and cerebellum areas, but upregulated in the striatum. To investigate whether Tfap2b controls sleep through GABAergic neurons, we specifically deleted Tfap2b in GABAergic neurons. We recorded the EEG and EMG before and after a 6-h period of sleep deprivation and extracted the time spent in NREM and in REM sleep as well as delta and theta power to assess NREM and REM sleep, respectively. During baseline conditions, Vgat-tfap2b-/- mice exhibited both shortened NREM and REM sleep time and reduced delta and theta power. Consistently, weaker delta and theta power were observed during rebound sleep in the Vgat-tfap2b-/- mice after sleep deprivation. Taken together, the results indicate that Tfap2b in GABAergic neurons is required for normal sleep.


Assuntos
Privação do Sono , Animais , Camundongos , Eletroencefalografia , Neurônios GABAérgicos , Mamíferos , Sono/fisiologia , Privação do Sono/genética , Fases do Sono/fisiologia
11.
MicroPubl Biol ; 20232023.
Artigo em Inglês | MEDLINE | ID: mdl-37038480

RESUMO

Phosphomimetic substitutions have been instrumental in understanding the role of phosphorylation in protein function. Here we describe the design and construction of a predicted phosphomimetic allele of the JUN kinase kinase gene jkk-1 in C. elegans. To generate the phosphomimetic kinase mutant JKK-1(3E), we edited jkk-1 to introduce three amino acid substitutions, S274E, S278E and S280E. The resulting strain is homozygous viable and extends the survival of L1-arrested larvae. This survival-extending phenotype suggests that the phosphomimetic mutations might promote activation of JKK-1 during the arrest. This jkk-1 potential gain-of-function allele might be useful for studying the regulation and functions of JKK-1.

12.
Genetics ; 225(2)2023 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-37682641

RESUMO

Sleep is a fundamental state of behavioral quiescence and physiological restoration. Sleep is controlled by environmental conditions, indicating a complex regulation of sleep by multiple processes. Our knowledge of the genes and mechanisms that control sleep during various conditions is, however, still incomplete. In Caenorhabditis elegans, sleep is increased when development is arrested upon starvation. Here, we performed a reverse genetic sleep screen in arrested L1 larvae for genes that are associated with metabolism. We found over 100 genes that are associated with a reduced sleep phenotype. Enrichment analysis revealed sphingolipid metabolism as a key pathway that controls sleep. A strong sleep loss was caused by the loss of function of the diacylglycerol kinase 1 gene, dgk-1, a negative regulator of synaptic transmission. Rescue experiments indicated that dgk-1 is required for sleep in cholinergic and tyraminergic neurons. The Ring Interneuron S (RIS) neuron is crucial for sleep in C. elegans and activates to induce sleep. RIS activation transients were abolished in dgk-1 mutant animals. Calcium transients were partially rescued by a reduction-of-function mutation of unc-13, suggesting that dgk-1 might be required for RIS activation by limiting synaptic vesicle release. dgk-1 mutant animals had impaired L1 arrest survival and dampened expression of the protective heat shock factor gene hsp-12.6. These data suggest that dgk-1 impairment causes broad physiological deficits. Microcalorimetry and metabolomic analyses of larvae with impaired RIS showed that RIS is broadly required for energy conservation and metabolic control, including for the presence of sphingolipids. Our data support the notion that metabolism broadly influences sleep and that sleep is associated with profound metabolic changes. We thus provide novel insights into the interplay of lipids and sleep and provide a rich resource of mutants and metabolic pathways for future sleep studies.

13.
Curr Biol ; 32(10): 2248-2262.e9, 2022 05 23.
Artigo em Inglês | MEDLINE | ID: mdl-35504281

RESUMO

Sleep is an essential state that allows for recuperation and survival processes. Disturbing sleep triggers stress responses that promote protective gene expression. Sleep and its deprivation grossly impact gene expression, but little is known about how normal or disturbed sleep control gene expression. Central to the induction of sleep are sleep-active neurons, which inhibit wakefulness and promote survival. Sleep and sleep-active neurons are highly conserved. In Caenorhabditis elegans, the sleep-active RIS neuron is crucial for sleep and survival. Here, we show that RIS depolarization promotes the protective gene expression response that occurs during developmental arrest. This response includes the activation of FOXO/DAF-16 and expression of DAF-16 target genes such as HSP-12.6, a small heat-shock protein that is required for starvation survival. Disturbing sleep by mechanical stimulation increases RIS depolarization. RIS activation in turn activates DAF-16 and other genes required for survival. Hence, during normal sleep, RIS depolarization promotes protective gene expression. When sleep is disturbed, protective gene expression gets further increased by raised RIS depolarization. We thus link sleep-active neuron depolarization to protective gene expression changes and suggest that the cellular stress response following sleep deprivation could be understood as a safeguarding process that is caused by the overactivation of sleep-active neurons.


Assuntos
Proteínas de Caenorhabditis elegans , Animais , Caenorhabditis elegans/fisiologia , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Expressão Gênica , Neurônios/fisiologia , Sono/genética
14.
Nature ; 436(7051): 731-4, 2005 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-16079852

RESUMO

The position of the cytokinesis furrow in a cell determines the relative sizes of its two daughter cells as well as the distribution of their contents. In animal cells, the position of the cytokinesis furrow is specified by the position of the mitotic spindle. The cytokinesis furrow bisects the spindle midway between the microtubule asters, at the site of the microtubule-based midzone, producing two daughter cells. Experiments in some cell types have suggested that the midzone positions the furrow, but experiments in other cells have suggested that the asters position the furrow. One possibility is that different organisms and cell types use different mechanisms to position the cytokinesis furrow. An alternative possibility is that both asters and the midzone contribute to furrow positioning. Recent work in C. elegans has suggested that centrosome separation and the midzone are implicated in cytokinesis. Here we examine the relative contributions of different parts of the mitotic spindle to positioning of the cytokinesis furrow in the C. elegans zygote. By spatially separating the spindle midzone from one of the asters using an ultraviolet laser, we show that the cytokinesis furrow is first positioned by a signal determined by microtubule asters, and then by a second signal that is derived from the spindle midzone. Thus, the position of the cytokinesis furrow is specified by two consecutive furrowing activities.


Assuntos
Caenorhabditis elegans/citologia , Compartimento Celular/fisiologia , Citocinese/fisiologia , Fuso Acromático/fisiologia , Actomiosina/metabolismo , Animais , Lasers , Microtúbulos/metabolismo , Raios Ultravioleta , Zigoto/citologia
15.
Curr Biol ; 31(3): 564-577.e12, 2021 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-33259791

RESUMO

Wounding and infection trigger a protective innate immune response that includes the production of antimicrobial peptides in the affected tissue as well as increased sleep. Little is known, however, how peripheral wounds or innate immunity signal to the nervous system to increase sleep. We found that, during C. elegans larval molting, an epidermal tolloid/bone morphogenic protein (BMP)-1-like protein called NAS-38 promotes sleep. NAS-38 is negatively regulated by its thrombospondin domain and acts through its astacin protease domain to activate p38 mitogen-activated protein (MAP)/PMK-1 kinase and transforming growth factor ß (TGF-ß)-SMAD/SMA-3-dependent innate immune pathways in the epidermis that cause STAT/STA-2 and SLC6 (solute carrier)/SNF-12-dependent expression of antimicrobial peptide (AMP) genes. We show that more than a dozen epidermal AMPs act as somnogens, signaling across tissues to promote sleep through the sleep-active RIS neuron. In the adult, epidermal injury activates innate immunity and turns up AMP production to trigger sleep, a process that requires epidermal growth factor receptor (EGFR) signaling that is known to promote sleep following cellular stress. We show for one AMP, neuropeptide-like protein (NLP)-29, that it acts through the neuropeptide receptor NPR-12 in locomotion-controlling neurons that are presynaptic to RIS and that depolarize this neuron to induce sleep. Sleep in turn increases the chance of surviving injury. Thus, we found a novel mechanism by which peripheral wounds signal to the nervous system to increase protective sleep. Such a cross-tissue somnogen-signaling function of AMPs might also boost sleep in other animals, including humans.


Assuntos
Imunidade Inata , Animais , Humanos , Peptídeos Antimicrobianos , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans , Epiderme , Sono
16.
Curr Biol ; 17(2): 185-91, 2007 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-17189697

RESUMO

At anaphase, the mitotic spindle positions the cytokinesis furrow [1]. Two populations of spindle microtubules are implicated in cytokinesis: radial microtubule arrays called asters and bundled nonkinetochore microtubules called the spindle midzone [2-4]. In C. elegans embryos, these two populations of microtubules provide two consecutive signals that position the cytokinesis furrow: The first signal is positioned midway between the microtubule asters; the second signal is positioned over the spindle midzone [5]. Evidence for two cytokinesis signals came from the identification of molecules that block midzone-positioned cytokinesis [5-7]. However, no molecules that are only required for, and thus define, the molecular pathway of aster-positioned cytokinesis have been identified. With RNAi screening, we identify LET-99 and the heterotrimeric G proteins GOA-1/GPA-16 and their regulator GPR-1/2 [10-12] in aster-positioned cytokinesis. By using mechanical spindle displacement, we show that the anaphase spindle positions cortical LET-99, at the site of the presumptive cytokinesis furrow. LET-99 enrichment at the furrow depends on the G proteins. GPR-1 is locally reduced at the site of cytokinesis-furrow formation by LET-99, which prevents accumulation of GPR-1 at this site. We conclude that LET-99 and the G proteins define a molecular pathway required for aster-positioned cytokinesis.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Citocinese/fisiologia , Proteínas Heterotriméricas de Ligação ao GTP/metabolismo , Anáfase/fisiologia , Animais , Caenorhabditis elegans , Citocinese/genética , Embrião não Mamífero , Subunidades alfa de Proteínas de Ligação ao GTP/metabolismo , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP , Proteínas de Ligação ao GTP/metabolismo , Interferência de RNA , Fuso Acromático/metabolismo , Fatores de Tempo
17.
Genetics ; 216(3): 735-752, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32769099

RESUMO

Sleep is a conserved behavioral state. Invertebrates typically show quiet sleep, whereas in mammals, sleep consists of periods of nonrapid-eye-movement sleep (NREMS) and REM sleep (REMS). We previously found that the transcription factor AP-2 promotes sleep in Caenorhabditiselegans and Drosophila In mammals, several paralogous AP-2 transcription factors exist. Sleep-controlling genes are often conserved. However, little is known about how sleep genes evolved from controlling simpler types of sleep to govern complex mammalian sleep. Here, we studied the roles of Tfap2a and Tfap2b in sleep control in mice. Consistent with our results from C. elegans and Drosophila, the AP-2 transcription factors Tfap2a and Tfap2b also control sleep in mice. Surprisingly, however, the two AP-2 paralogs play contrary roles in sleep control. Tfap2a reduction of function causes stronger delta and theta power in both baseline and homeostasis analysis, thus indicating increased sleep quality, but did not affect sleep quantity. By contrast, Tfap2b reduction of function decreased NREM sleep time specifically during the dark phase, reduced NREMS and REMS power, and caused a weaker response to sleep deprivation. Consistent with the observed signatures of decreased sleep quality, stress resistance and memory were impaired in Tfap2b mutant animals. Also, the circadian period was slightly shortened. Taken together, AP-2 transcription factors control sleep behavior also in mice, but the role of the AP-2 genes functionally diversified to allow for a bidirectional control of sleep quality. Divergence of AP-2 transcription factors might perhaps have supported the evolution of more complex types of sleep.


Assuntos
Privação do Sono/genética , Fases do Sono , Fator de Transcrição AP-2/metabolismo , Animais , Ritmo Circadiano , Ritmo Delta , Memória , Camundongos , Privação do Sono/fisiopatologia , Ritmo Teta , Fator de Transcrição AP-2/genética
18.
Curr Biol ; 30(1): 1-16.e13, 2020 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-31839447

RESUMO

Sleep requires sleep-active neurons that depolarize to inhibit wake circuits. Sleep-active neurons are under the control of homeostatic mechanisms that determine sleep need. However, little is known about the molecular and circuit mechanisms that translate sleep need into the depolarization of sleep-active neurons. During many stages and conditions in C. elegans, sleep requires a sleep-active neuron called RIS. Here, we defined the transcriptome of RIS and discovered that genes of the epidermal growth factor receptor (EGFR) signaling pathway are expressed in RIS. Because of cellular stress, EGFR directly activates RIS. Activation of EGFR signaling in the ALA neuron has previously been suggested to promote sleep independently of RIS. Unexpectedly, we found that ALA activation promotes RIS depolarization. Our results suggest that ALA is a drowsiness neuron with two separable functions: (1) it inhibits specific behaviors, such as feeding, independently of RIS, (2) and it activates RIS. Whereas ALA plays a strong role in surviving cellular stress, surprisingly, RIS does not. In summary, EGFR signaling can depolarize RIS by an indirect mechanism through activation of the ALA neuron that acts upstream of the sleep-active RIS neuron and through a direct mechanism using EGFR signaling in RIS. ALA-dependent drowsiness, rather than RIS-dependent sleep bouts, appears to be important for increasing survival after cellular stress, suggesting that different types of behavioral inhibition play different roles in restoring health. VIDEO ABSTRACT.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/fisiologia , Fator de Crescimento Epidérmico/metabolismo , Neurônios/fisiologia , Transdução de Sinais , Sono/fisiologia , Animais
19.
Nat Commun ; 10(1): 4095, 2019 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-31506439

RESUMO

Animals must slow or halt locomotion to integrate sensory inputs or to change direction. In Caenorhabditis elegans, the GABAergic and peptidergic neuron RIS mediates developmentally timed quiescence. Here, we show RIS functions additionally as a locomotion stop neuron. RIS optogenetic stimulation caused acute and persistent inhibition of locomotion and pharyngeal pumping, phenotypes requiring FLP-11 neuropeptides and GABA. RIS photoactivation allows the animal to maintain its body posture by sustaining muscle tone, yet inactivating motor neuron oscillatory activity. During locomotion, RIS axonal Ca2+ signals revealed functional compartmentalization: Activity in the nerve ring process correlated with locomotion stop, while activity in a branch correlated with induced reversals. GABA was required to induce, and FLP-11 neuropeptides were required to sustain locomotion stop. RIS attenuates neuronal activity and inhibits movement, possibly enabling sensory integration and decision making, and exemplifies dual use of one cell across development in a compact nervous system.


Assuntos
Caenorhabditis elegans/fisiologia , Cálcio/metabolismo , Neurônios GABAérgicos/metabolismo , Locomoção/fisiologia , Neuropeptídeos/metabolismo , Sono/fisiologia , Animais , Axônios/metabolismo , Caenorhabditis elegans/citologia , Neurônios Colinérgicos/fisiologia , Junções Comunicantes/metabolismo , Luz , Modelos Biológicos , Neurônios Motores/fisiologia , Músculos/citologia , Fenótipo , Transdução de Sinais , Ácido gama-Aminobutírico/metabolismo
20.
Genetics ; 208(4): 1279-1289, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29618588

RESUMO

Sleep is crucial for survival and well-being. This behavioral and physiological state has been studied in all major genetically accessible model animals, including rodents, fish, flies, and worms. Genetic and optogenetic studies have identified several neurons that control sleep, making it now possible to compare circuit mechanisms across species. The "motor" of sleep across animal species is formed by neurons that depolarize at the onset of sleep to actively induce this state by directly inhibiting wakefulness. These sleep-inducing neurons are themselves controlled by inhibitory or activating upstream pathways, which act as the "drivers" of the sleep motor: arousal inhibits "sleep-active" neurons whereas various sleep-promoting "tiredness" pathways converge onto sleep-active neurons to depolarize them. This review provides the first overview of sleep-active neurons across the major model animals. The occurrence of sleep-active neurons and their regulation by upstream pathways in both vertebrate and invertebrate species suggests that these neurons are general and ancient components that evolved early in the history of nervous systems.


Assuntos
Evolução Biológica , Neurônios/fisiologia , Sono/fisiologia , Animais , Humanos , Mamíferos , Modelos Animais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA