Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Artículo en Inglés | MEDLINE | ID: mdl-38336391

RESUMEN

Sleep is a fundamental feature of life for virtually all multicellular animals, but many questions remain about how sleep is regulated by circadian rhythms, homeostatic sleep drive that builds up with wakefulness, and modifying factors such as hunger or social interactions, as well as about the biological functions of sleep. Substantial headway has been made in the study of both circadian rhythms and sleep in the fruit fly Drosophila melanogaster, much of it through studies of individual fly activity using Drosophila activity monitors (DAMs). Here, we describe approaches for the activation of specific neurons of interest using optogenetics (involving genetic modifications that allow for light-based neuronal activation) and thermogenetics (involving genetic modifications that allow for temperature-based neuronal activation) so that researchers can evaluate the roles of those neurons in controlling rest and activity behavior. In this protocol, we describe how to set up a rig for simultaneous optogenetic or thermogenetic stimulation and activity monitoring for analysis of sleep and circadian rhythms in Drosophila, how to raise appropriate flies, and how to perform the experiment. This protocol will allow researchers to assess the causative role in the regulation of sleep and activity rhythms of any genetically tractable subset of cells.

2.
Artículo en Inglés | MEDLINE | ID: mdl-38336392

RESUMEN

Sleep is a fundamental feature of life for virtually all multicellular animals, but many questions remain about how sleep is regulated and what biological functions it plays. Substantial headway has been made in the study of both circadian rhythms and sleep in the fruit fly Drosophila melanogaster, much of it through studies of individual fly activity using beam break counts from Drosophila activity monitors (DAMs). The number of laboratories worldwide studying sleep in Drosophila has grown from only a few 20 years ago to hundreds today. The utility of these studies is limited by the quality of the metrics that can be extracted from the data. Many software options exist to help analyze DAM data; however, these are often expensive or have significant limitations. Therefore, we describe here a method for analyzing DAM-based data using the sleep and circadian analysis MATLAB program (SCAMP). This user-friendly software has an advantage of combining several analyses of both sleep and circadian rhythms in one package and produces graphical outputs as well as spreadsheets of the outputs for further statistical analysis. The version of SCAMP described here is also the first published software package that can analyze data from multibeam DAM5Ms, enabling determination of positional preference over time.

3.
Artículo en Inglés | MEDLINE | ID: mdl-38336390

RESUMEN

Sleep is important for survival, and the need for sleep is conserved across species. In the past two decades, the fruit fly Drosophila melanogaster has emerged as a promising system in which to study the genetic, neural, and physiological bases of sleep. Through significant advances in our understanding of the regulation of sleep in flies, the field is poised to address several open questions about sleep, such as how the need for sleep is encoded, how molecular regulators of sleep are situated within brain networks, and what the functions of sleep are. Here, we describe key findings, open questions, and commonly used methods that have been used to inform existing theories and develop new ways of thinking about the function, regulation, and adaptability of sleep behavior.

4.
Artículo en Inglés | MEDLINE | ID: mdl-34785379

RESUMEN

Balance between sleep, wakefulness and arousal is important for survival of organisms and species as a whole. While, the benefits of sleep both in terms of quantity and quality is widely recognized across species, sleep has a cost for organismal survival and reproduction. Here we focus on how sleep duration, sleep depth and sleep pressure affect the ability of animals to engage in courtship and egg-laying behaviors critical for reproductive success. Using isogenic lines from the Drosophila Genetic Reference Panel with variable sleep phenotypes we investigated the relationship between sleep and reproductive behaviors, courtship and oviposition. We found that three out of five lines with decreased sleep and increased arousal phenotypes, showed increased courtship and decreased latency to court as compared to normal and long sleeping lines. However, the male courtship phenotype is dependent on context and genotype as some but not all long sleeping-low courting lines elevate their courtship in the presence of short sleeping-high courting flies. We also find that unlike courtship, sleep phenotypes were less variable and minimally susceptible to social experience. In addition to male courtship, we also investigated egg-laying phenotype, a readout of female reproductive output and find oviposition to be less sensitive to sleep length and parameters that are indicative of switch between sleep and wake states. Taken together our extensive behavioral analysis here shows complex bidirectional interactions between genotype and environment and add to the growing evidence linking sleep duration and sleep-wake switch parameters to behavioral decision making critical to reproductive output.


Asunto(s)
Drosophila melanogaster/fisiología , Reproducción/fisiología , Conducta Sexual Animal/fisiología , Sueño/fisiología , Animales , Nivel de Alerta/fisiología , Cortejo , Toma de Decisiones , Femenino , Masculino , Oviposición/fisiología , Fenotipo , Grabación en Video , Vigilia/fisiología
5.
Sci Rep ; 11(1): 20067, 2021 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-34625611

RESUMEN

Sleep is a fundamental behavioral state important for survival and is universal in animals with sufficiently complex nervous systems. As a highly conserved neurobehavioral state, sleep has been described in species ranging from jellyfish to humans. Biogenic amines like dopamine, serotonin and norepinephrine have been shown to be critical for sleep regulation across species but the precise circuit mechanisms underlying how amines control persistence of sleep, arousal and wakefulness remain unclear. The fruit fly, Drosophila melanogaster, provides a powerful model system for the study of sleep and circuit mechanisms underlying state transitions and persistence of states to meet the organisms motivational and cognitive needs. In Drosophila, two neuropils in the central brain, the mushroom body (MB) and the central complex (CX) have been shown to influence sleep homeostasis and receive aminergic neuromodulator input critical to sleep-wake switch. Dopamine neurons (DANs) are prevalent neuromodulator inputs to the MB but the mechanisms by which they interact with and regulate sleep- and wake-promoting neurons within MB are unknown. Here we investigate the role of subsets of PAM-DANs that signal wakefulness and project to wake-promoting compartments of the MB. We find that PAM-DANs are GABA responsive and require GABAA-Rdl receptor in regulating sleep. In mapping the pathways downstream of PAM neurons innervating γ5 and ß'2 MB compartments we find that wakefulness is regulated by both DopR1 and DopR2 receptors in downstream Kenyon cells (KCs) and mushroom body output neurons (MBONs). Taken together, we have identified and characterized a dopamine modulated sleep microcircuit within the mushroom body that has previously been shown to convey information about positive and negative valence critical for memory formation. These studies will pave way for understanding how flies balance sleep, wakefulness and arousal.


Asunto(s)
Neuronas Dopaminérgicas/fisiología , Drosophila melanogaster/fisiología , Cuerpos Pedunculados/fisiología , Sueño/fisiología , Vigilia/fisiología , Ácido gamma-Aminobutírico/metabolismo , Animales , Femenino , Masculino , Transducción de Señal
7.
Artículo en Inglés | MEDLINE | ID: mdl-33779562

RESUMEN

This review serves as an introduction to a Special Issue of Comparative Biochemistry and Physiology, focused on using non-human models to study biomedical physiology. The concept of a model differs across disciplines. For example, several models are used primarily to gain an understanding of specific human pathologies and disease states, whereas other models may be focused on gaining insight into developmental or evolutionary mechanisms. It is often the case that animals initially used to gain knowledge of some unique biochemical or physiological process finds foothold in the biomedical community and becomes an established model. The choice of a particular model for biomedical research is an ongoing process and model validation must keep pace with existing and emerging technologies. While the importance of non-mammalian models, such as Caenorhabditis elegans, Drosophila melanogaster, Danio rerio and Xenopus laevis, is well known, we also seek to bring attention to emerging alternative models of both invertebrates and vertebrates, which are less established but of interest to the comparative biochemistry and physiology community.


Asunto(s)
Investigación Biomédica , Modelos Biológicos , Animales , Caenorhabditis elegans , Drosophila melanogaster , Xenopus laevis , Pez Cebra
8.
Artículo en Inglés | MEDLINE | ID: mdl-33737041

RESUMEN

The Publisher regrets that this article is an accidental duplication of an article that has already been published in Comparative Biochemistry and Physiology Part B: Biochemistry and Molecular Biology, Volume 255, 2021, 110593, https://doi.org/10.1016/j.cbpb.2021.110593. The duplicate article has therefore been withdrawn. The full Elsevier Policy on Article Withdrawal can be found at https://www.elsevier.com/about/our-business/policies/article-withdrawal.

9.
J Neurogenet ; 34(1): 2-4, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32233836

RESUMEN

Troy D. Zars (1967-2018) was an American biologist. He studied the relationships between genes, neuronal circuits and behavior in the fruit fly Drosophila melanogaster. Zars co-pioneered the use of transgene expression to locally restore gene function in memory-defective fly mutants, an approach that provided breakthrough insights into the localization of memory traces in the fly brain. With ensuing refinements of the methods of transgene expression and the broadening in the range of transgenes to be expressed, this shaped the field of modern behavioral neurogenetics.


Asunto(s)
Genética/historia , Neurología/historia , Animales , Conducta/fisiología , Encéfalo/fisiología , Drosophila melanogaster , Historia del Siglo XX , Historia del Siglo XXI , Humanos
10.
J Neurogenet ; 34(1): 21-27, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31878832

RESUMEN

Preference for spatial locations to maximize favorable outcomes and minimize aversive experiences helps animals survive and adapt to the changing environment. Both visual and non-visual cues play a critical role in spatial navigation and memory of a place supports and guides these strategies. Here we present the neural, genetic and behavioral processes involved in place memory formation using Drosophila melanogaster with a focus on non-visual cue based spatial memories. The work presented here highlights the work done by Dr. Troy Zars and his colleagues with an emphasis on role of biogenic amines in learning, cell biological mechanisms of neural systems and behavioral plasticity of place conditioning.


Asunto(s)
Conducta Animal/fisiología , Drosophila melanogaster/fisiología , Aprendizaje/fisiología , Memoria/fisiología , Refuerzo en Psicología , Animales
11.
Bio Protoc ; 9(12)2019.
Artículo en Inglés | MEDLINE | ID: mdl-33225013

RESUMEN

Sleep is a conserved neurobehavioral state observed in animals with sufficiently complex nervous systems and is critical for survival. While the exact function of sleep remains unknown, the lack of sleep can have a range of physiological and behavioral effects. Studies in invertebrates and vertebrates have identified conserved neural mechanisms and cellular pathways in control of sleep, wakefulness and arousal. Methodologies to measure sleep have ranged from EEG recordings in humans and rodents to in-depth analysis of locomotor patterns in flies, fish and worms. Here we focus on sleep measurements using activity monitoring in the highly versatile experimental model system, Drosophila melanogaster, which is amenable to a number of genetic, physiological and behavioral manipulations. Further, we also describe methods used to manipulate sleep and wakefulness to understand the neural regulation of sleep and how organisms balance sleep, wakefulness and behavioral arousal. Sleep as a behavioral state is regulated by a number of factors including food, environmental conditions, and genetic background. The methodologies described here provide, a high-throughput approach to study neural regulation of sleep and factors that affect this complex behavior.

12.
J Undergrad Neurosci Educ ; 16(2): A177-A185, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30057500

RESUMEN

A key element of laboratory courses introducing students to neuroscience includes behavioral exercises. Associative learning experiments often conducted in research laboratories are difficult to perform and time consuming. Commonly, these experiments cannot be performed without extensive instrumentation or animal care facilities. Here, we describe three distinct laboratory modules that build on simple chemosensory and memory assays in Drosophila larvae. Additionally, we describe open-ended research projects using these assays that can be developed into semester long independent research experiences. Given that Drosophila is a genetic model organism, these simple behavioral assays can be used to generate multiple hypothesis driven projects aimed at identifying a gene or class of neurons involved in appetitive and aversive learning. These lab modules are ideally suited for undergraduates at all levels to experience and can be incorporated in a lower/upper level neuroscience course or as a high school outreach exercise. Further, these modules enable students to collect their own data sets, work in groups in collating large data sets, performing statistical comparisons, and presenting results in the form of short research papers or traditional laboratory reports that include a short literature review.

13.
Curr Biol ; 27(13): 1915-1927.e5, 2017 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-28669757

RESUMEN

The mechanisms by which clock neurons in the Drosophila brain confer an ∼24-hr rhythm onto locomotor activity are unclear, but involve the neuropeptide diuretic hormone 44 (DH44), an ortholog of corticotropin-releasing factor. Here we identified DH44 receptor 1 as the relevant receptor for rest:activity rhythms and mapped its site of action to hugin-expressing neurons in the subesophageal zone (SEZ). We traced a circuit that extends from Dh44-expressing neurons in the pars intercerebralis (PI) through hugin+ SEZ neurons to the ventral nerve cord. Hugin neuropeptide, a neuromedin U ortholog, also regulates behavioral rhythms. The DH44 PI-Hugin SEZ circuit controls circadian locomotor activity in a daily cycle but has minimal effect on feeding rhythms, suggesting that the circadian drive to feed can be separated from circadian locomotion. These findings define a linear peptidergic circuit that links the clock to motor outputs to modulate circadian control of locomotor activity.


Asunto(s)
Relojes Circadianos/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/fisiología , Locomoción/genética , Neuropéptidos/genética , Receptores de Superficie Celular/genética , Animales , Animales Modificados Genéticamente/genética , Animales Modificados Genéticamente/fisiología , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Masculino , Neuropéptidos/metabolismo , Receptores de Superficie Celular/metabolismo
14.
Nat Commun ; 8(1): 154, 2017 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-28754889

RESUMEN

Animals execute one particular behavior among many others in a context-dependent manner, yet the mechanisms underlying such behavioral choice remain poorly understood. Here we studied how two fundamental behaviors, sex and sleep, interact at genetic and neuronal levels in Drosophila. We show that an increased need for sleep inhibits male sexual behavior by decreasing the activity of the male-specific P1 neurons that coexpress the sex determination genes fru M and dsx, but does not affect female sexual behavior. Further, we delineate a sex-specific neuronal circuit wherein the P1 neurons encoding increased courtship drive suppressed male sleep by forming mutually excitatory connections with the fru M -positive sleep-controlling DN1 neurons. In addition, we find that FRUM regulates male courtship and sleep through distinct neural substrates. These studies reveal the genetic and neuronal basis underlying the sex-specific interaction between sleep and sexual behaviors in Drosophila, and provide insights into how competing behaviors are co-regulated.Genes and circuits involved in sleep and sexual arousal have been extensively studied in Drosophila. Here the authors identify the sex determination genes fruitless and doublesex, and a sex-specific P1-DN1 neuronal feedback that governs the interaction between these competing behaviors.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Proteínas del Tejido Nervioso/genética , Neuronas/metabolismo , Conducta Sexual Animal/fisiología , Sueño/genética , Factores de Transcripción/genética , Animales , Cortejo , Drosophila melanogaster/fisiología , Femenino , Masculino , Neuronas/fisiología , Caracteres Sexuales , Sueño/fisiología
15.
Front Syst Neurosci ; 11: 92, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29321732

RESUMEN

Feedback mechanisms in operant learning are critical for animals to increase reward or reduce punishment. However, not all conditions have a behavior that can readily resolve an event. Animals must then try out different behaviors to better their situation through outcome learning. This form of learning allows for novel solutions and with positive experience can lead to unexpected behavioral routines. Learned helplessness, as a type of outcome learning, manifests in part as increases in escape latency in the face of repeated unpredicted shocks. Little is known about the mechanisms of outcome learning. When fruit fly Drosophilamelanogaster are exposed to unpredicted high temperatures in a place learning paradigm, flies both increase escape latencies and have a higher memory when given control of a place/temperature contingency. Here we describe discrete serotonin neuronal circuits that mediate aversive reinforcement, escape latencies, and memory levels after place learning in the presence and absence of unexpected aversive events. The results show that two features of learned helplessness depend on the same modulatory system as aversive reinforcement. Moreover, changes in aversive reinforcement and escape latency depend on local neural circuit modulation, while memory enhancement requires larger modulation of multiple behavioral control circuits.

16.
Artículo en Inglés | MEDLINE | ID: mdl-26617493

RESUMEN

The Drosophila mushroom body (MB) is an associative learning network that is important for the control of sleep. We have recently identified particular intrinsic MB Kenyon cell (KC) classes that regulate sleep through synaptic activation of particular MB output neurons (MBONs) whose axons convey sleep control signals out of the MB to downstream target regions. Specifically, we found that sleep-promoting KCs increase sleep by preferentially activating cholinergic sleep-promoting MBONs, while wake-promoting KCs decrease sleep by preferentially activating glutamatergic wake-promoting MBONs. Here we use a combination of genetic and physiological approaches to identify wake-promoting dopaminergic neurons (DANs) that innervate the MB, and show that they activate wake-promoting MBONs. These studies reveal a dopaminergic sleep control mechanism that likely operates by modulation of KC-MBON microcircuits.


Asunto(s)
Neuronas Dopaminérgicas/fisiología , Cuerpos Pedunculados/fisiología , Sueño/fisiología , Animales , Animales Modificados Genéticamente , Drosophila melanogaster , Inmunohistoquímica , Modelos Animales , Vías Nerviosas/fisiología
17.
Curr Biol ; 25(22): 2915-27, 2015 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-26455303

RESUMEN

The Drosophila mushroom body (MB) is a key associative memory center that has also been implicated in the control of sleep. However, the identity of MB neurons underlying homeostatic sleep regulation, as well as the types of sleep signals generated by specific classes of MB neurons, has remained poorly understood. We recently identified two MB output neuron (MBON) classes whose axons convey sleep control signals from the MB to converge in the same downstream target region: a cholinergic sleep-promoting MBON class and a glutamatergic wake-promoting MBON class. Here, we deploy a combination of neurogenetic, behavioral, and physiological approaches to identify and mechanistically dissect sleep-controlling circuits of the MB. Our studies reveal the existence of two segregated excitatory synaptic microcircuits that propagate homeostatic sleep information from different populations of intrinsic MB "Kenyon cells" (KCs) to specific sleep-regulating MBONs: sleep-promoting KCs increase sleep by preferentially activating the cholinergic MBONs, while wake-promoting KCs decrease sleep by preferentially activating the glutamatergic MBONs. Importantly, activity of the sleep-promoting MB microcircuit is increased by sleep deprivation and is necessary for homeostatic rebound sleep (i.e., the increased sleep that occurs after, and in compensation for, sleep lost during deprivation). These studies reveal for the first time specific functional connections between subsets of KCs and particular MBONs and establish the identity of synaptic microcircuits underlying transmission of homeostatic sleep signals in the MB.


Asunto(s)
Drosophila melanogaster/fisiología , Cuerpos Pedunculados/fisiología , Sueño/fisiología , Animales , Femenino , Homeostasis/fisiología , Masculino , Memoria/fisiología , Neuronas/fisiología , Reproducción/fisiología , Olfato/fisiología , Relación Estructura-Actividad
18.
Artículo en Inglés | MEDLINE | ID: mdl-26733821

RESUMEN

[This corrects the article on p. 73 in vol. 9, PMID: 26617493.].

19.
Elife ; 3: e04580, 2014 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-25535794

RESUMEN

Animals discriminate stimuli, learn their predictive value and use this knowledge to modify their behavior. In Drosophila, the mushroom body (MB) plays a key role in these processes. Sensory stimuli are sparsely represented by ∼2000 Kenyon cells, which converge onto 34 output neurons (MBONs) of 21 types. We studied the role of MBONs in several associative learning tasks and in sleep regulation, revealing the extent to which information flow is segregated into distinct channels and suggesting possible roles for the multi-layered MBON network. We also show that optogenetic activation of MBONs can, depending on cell type, induce repulsion or attraction in flies. The behavioral effects of MBON perturbation are combinatorial, suggesting that the MBON ensemble collectively represents valence. We propose that local, stimulus-specific dopaminergic modulation selectively alters the balance within the MBON network for those stimuli. Our results suggest that valence encoded by the MBON ensemble biases memory-based action selection.


Asunto(s)
Conducta de Elección , Drosophila melanogaster/citología , Drosophila melanogaster/fisiología , Memoria , Cuerpos Pedunculados/citología , Cuerpos Pedunculados/inervación , Neuronas/fisiología , Animales , Conducta Apetitiva/efectos de la radiación , Aprendizaje por Asociación/efectos de la radiación , Reacción de Prevención/efectos de la radiación , Conducta Animal/efectos de la radiación , Conducta de Elección/efectos de la radiación , Luz , Memoria/efectos de la radiación , Modelos Neurológicos , Cuerpos Pedunculados/efectos de la radiación , Neuronas/efectos de la radiación , Odorantes , Sueño/efectos de la radiación , Factores de Tiempo , Visión Ocular
20.
J Neurogenet ; 26(2): 238-44, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22436011

RESUMEN

The biogenic amines dopamine, octopamine, and serotonin are critical in establishing normal memories. A common view for the amines in insect memory performance has emerged in which dopamine and octopamine are largely responsible for aversive and appetitive memories. Examination of the function of serotonin begins to challenge the notion of one amine type per memory because altering serotonin function also reduces aversive olfactory memory and place memory levels. Could the function of serotonin be restricted to the aversive domain, suggesting a more specific dopamine/serotonin system interaction? The function of the serotonergic system in appetitive olfactory memory was examined. By targeting the tetanus toxin light chain (TNT) and the human inwardly rectifying potassium channel (Kir2.1) to the serotonin neurons with two different GAL4 driver combinations, the serotonergic system was inhibited. Additional use of the GAL80(ts1) system to control expression of transgenes to the adult stage of the life cycle addressed a potential developmental role of serotonin in appetitive memory. Reduction in appetitive olfactory memory performance in flies with these transgenic manipulations, without altering control behaviors, showed that the serotonergic system is also required for normal appetitive memory. Thus, serotonin appears to have a more general role in Drosophila memory, and implies an interaction with both the dopaminergic and octopaminergic systems.


Asunto(s)
Regulación de la Expresión Génica/genética , Memoria a Corto Plazo/fisiología , Vías Olfatorias/fisiología , Recompensa , Serotonina/metabolismo , Olfato/genética , Animales , Animales Modificados Genéticamente , Conducta Animal , Antígenos CD8/metabolismo , Condicionamiento Psicológico/fisiología , Drosophila , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Odorantes , Vías Olfatorias/citología , Canales de Potasio de Rectificación Interna/genética , Células Receptoras Sensoriales/fisiología , Toxina Tetánica/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Tirosina 3-Monooxigenasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA