Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters











Publication year range
2.
J Biol Rhythms ; 38(3): 245-258, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37226809

ABSTRACT

The origin of experimental chronobiology can be traced to observations made in the 18th and 19th centuries on the sensitive plant Mimosa, which were described in two seminal reports: Jean-Jacques d'Ortous de Mairan's "Observation Botanique" (A Botanical Observation) and Augustin Pyramus de Candolle's "Du sommeil des feuilles" (On the sleep of leaves). Both report observations of the striking daily closing and opening of Mimosa leaves in controlled environments. This review presents translations of both texts with the aim of staying as faithful as possible to the original French texts. We also present the historical context in which these texts were written and link them to subsequent experiments that aimed at testing the veracity of their central conclusions. In particular, we definitely establish that Mairan himself presented his work to the French Royal Academy of Sciences, while the published report of his observation was authored by Fontenelle, the Secretary of the Academy. In addition, we offer a translation of Mairan's own presentation, based on the hand-written minutes of the academy. Finally, we discuss the decades of work on plant rhythms that laid the foundation for modern experimental chronobiology, including translations and discussion of the insightful and prescient reports by Charles François de Cisternay Dufay, Henri Louis Duhamel du Monceau, Johann Gottfried Zinn, and Wilhelm Pfeffer, which describe their efforts to reproduce and extend Mairan's pioneering observations.


Subject(s)
Circadian Rhythm , Mimosa , Sleep , Plant Leaves
3.
Elife ; 112022 06 29.
Article in English | MEDLINE | ID: mdl-35766361

ABSTRACT

The circadian clock orchestrates daily changes in physiology and behavior to ensure internal temporal order and optimal timing across the day. In animals, a central brain clock coordinates circadian rhythms throughout the body and is characterized by a remarkable robustness that depends on synaptic connections between constituent neurons. The clock neuron network of Drosophila, which shares network motifs with clock networks in the mammalian brain yet is built of many fewer neurons, offers a powerful model for understanding the network properties of circadian timekeeping. Here, we report an assessment of synaptic connectivity within a clock network, focusing on the critical lateral neuron (LN) clock neuron classes within the Janelia hemibrain dataset. Our results reveal that previously identified anatomical and functional subclasses of LNs represent distinct connectomic types. Moreover, we identify a small number of non-clock cell subtypes representing highly synaptically coupled nodes within the clock neuron network. This suggests that neurons lacking molecular timekeeping likely play integral roles within the circadian timekeeping network. To our knowledge, this represents the first comprehensive connectomic analysis of a circadian neuronal network.


Most organisms on Earth possess an internal timekeeping system which ensures that bodily processes such as sleep, wakefulness or digestion take place at the right time. These precise daily rhythms are kept in check by a master clock in the brain. There, thousands of neurons ­ some of which carrying an internal 'molecular clock' ­ connect to each other through structures known as synapses. Exactly how the resulting network is organised to support circadian timekeeping remains unclear. To explore this question, Shafer, Gutierrez et al. focused on fruit flies, as recent efforts have systematically mapped every neuron and synaptic connection in the brain of this model organism. Analysing available data from the hemibrain connectome project at Janelia revealed that that the neurons with the most important timekeeping roles were in fact forming the fewest synapses within the network. In addition, neurons without internal molecular clocks mediated strong synaptic connections between those that did, suggesting that 'clockless' cells still play an integral role in circadian timekeeping. With this research, Shafer, Gutierrez et al. provide unexpected insights into the organisation of the master body clock. Better understanding the networks that underpin circadian rhythms will help to grasp how and why these are disrupted in obesity, depression and Alzheimer's disease.


Subject(s)
Circadian Clocks , Connectome , Drosophila Proteins , Pacemaker, Artificial , Animals , Circadian Clocks/physiology , Circadian Rhythm/physiology , Drosophila/physiology , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/physiology , Mammals/metabolism , Neurons/physiology
4.
J Biol Rhythms ; 37(4): 455-467, 2022 08.
Article in English | MEDLINE | ID: mdl-35727044

ABSTRACT

The problem of entrainment is central to circadian biology. In this regard, Drosophila has been an important model system. Owing to the simplicity of its nervous system and the availability of powerful genetic tools, the system has shed significant light on the molecular and neural underpinnings of entrainment. However, much remains to be learned regarding the molecular and physiological mechanisms underlying this important phenomenon. Under cyclic light/dark conditions, Drosophila melanogaster displays crepuscular patterns of locomotor activity with one peak anticipating dawn and the other anticipating dusk. These peaks are characterized through an estimation of their phase relative to the environmental light cycle and the extent of their anticipation of light transitions. In Drosophila chronobiology, estimations of phases are often subjective, and anticipation indices vary significantly between studies. Though there is increasing interest in building flexible analysis tools in the field, none incorporates objective measures of Drosophila activity peaks in combination with the analysis of fly activity/sleep in the same program. To this end, we have developed PHASE, a MATLAB-based program that is simple and easy to use and (i) supports the visualization and analysis of activity and sleep under entrainment, (ii) allows analysis of both activity and sleep parameters within user-defined windows within a diurnal cycle, (iii) uses a smoothing filter for the objective identification of peaks of activity (and therefore can be used to quantitatively characterize them), and (iv) offers a series of analyses for the assessment of behavioral anticipation of environmental transitions.


Subject(s)
Circadian Rhythm , Drosophila melanogaster , Animals , Circadian Rhythm/physiology , Drosophila , Drosophila melanogaster/physiology , Motor Activity/physiology , Photoperiod , Sleep
5.
Curr Biol ; 31(1): R38-R49, 2021 01 11.
Article in English | MEDLINE | ID: mdl-33434488

ABSTRACT

Sleep is critical for diverse aspects of brain function in animals ranging from invertebrates to humans. Powerful genetic tools in the fruit fly Drosophila melanogaster have identified - at an unprecedented level of detail - genes and neural circuits that regulate sleep. This research has revealed that the functions and neural principles of sleep regulation are largely conserved from flies to mammals. Further, genetic approaches to studying sleep have uncovered mechanisms underlying the integration of sleep and many different biological processes, including circadian timekeeping, metabolism, social interactions, and aging. These findings show that in flies, as in mammals, sleep is not a single state, but instead consists of multiple physiological and behavioral states that change in response to the environment, and is shaped by life history. Here, we review advances in the study of sleep in Drosophila, discuss their implications for understanding the fundamental functions of sleep that are likely to be conserved among animal species, and identify important unanswered questions in the field.


Subject(s)
Drosophila melanogaster/physiology , Sleep/physiology , Aging/physiology , Animals , Circadian Rhythm/physiology , Social Interaction
6.
J Neurosci ; 40(50): 9617-9633, 2020 12 09.
Article in English | MEDLINE | ID: mdl-33172977

ABSTRACT

Dopamine is a wake-promoting neuromodulator in mammals and fruit flies. In Drosophila melanogaster, the network of clock neurons that drives sleep/activity cycles comprises both wake-promoting and sleep-promoting cell types. The large ventrolateral neurons (l-LNvs) and small ventrolateral neurons (s-LNvs) have been identified as wake-promoting neurons within the clock neuron network. The l-LNvs are innervated by dopaminergic neurons, and earlier work proposed that dopamine signaling raises cAMP levels in the l-LNvs and thus induces excitatory electrical activity (action potential firing), which results in wakefulness and inhibits sleep. Here, we test this hypothesis by combining cAMP imaging and patch-clamp recordings in isolated brains. We find that dopamine application indeed increases cAMP levels and depolarizes the l-LNvs, but, surprisingly, it does not result in increased firing rates. Downregulation of the excitatory D1-like dopamine receptor (Dop1R1) in the l-LNvs and s-LNvs, but not of Dop1R2, abolished the depolarization of l-LNvs in response to dopamine. This indicates that dopamine signals via Dop1R1 to the l-LNvs. Downregulation of Dop1R1 or Dop1R2 in the l-LNvs and s-LNvs does not affect sleep in males. Unexpectedly, we find a moderate decrease of daytime sleep with downregulation of Dop1R1 and of nighttime sleep with downregulation of Dop1R2. Since the l-LNvs do not use Dop1R2 receptors and the s-LNvs also respond to dopamine, we conclude that the s-LNvs are responsible for the observed decrease in nighttime sleep. In summary, dopamine signaling in the wake-promoting LNvs is not required for daytime arousal, but likely promotes nighttime sleep via the s-LNvs.SIGNIFICANCE STATEMENT In insect and mammalian brains, sleep-promoting networks are intimately linked to the circadian clock, and the mechanisms underlying sleep and circadian timekeeping are evolutionarily ancient and highly conserved. Here we show that dopamine, one important sleep modulator in flies and mammals, plays surprisingly complex roles in the regulation of sleep by clock-containing neurons. Dopamine inhibits neurons in a central brain sleep center to promote sleep and excites wake-promoting circadian clock neurons. It is therefore predicted to promote wakefulness through both of these networks. Nevertheless, our results reveal that dopamine acting on wake-promoting clock neurons promotes sleep, revealing a previously unappreciated complexity in the dopaminergic control of sleep.


Subject(s)
Circadian Rhythm/physiology , Dopamine/metabolism , Neurons/metabolism , Signal Transduction/physiology , Sleep/physiology , Action Potentials/physiology , Animals , Drosophila Proteins/metabolism , Drosophila melanogaster , Female , Male , Patch-Clamp Techniques , Receptors, Dopamine/metabolism , Receptors, Dopamine D1/metabolism
7.
Curr Biol ; 30(12): 2225-2237.e5, 2020 06 22.
Article in English | MEDLINE | ID: mdl-32386535

ABSTRACT

Networks of circadian timekeeping in the brain display marked daily changes in neuronal morphology. In Drosophila melanogaster, the striking daily structural remodeling of the dorsal medial termini of the small ventral lateral neurons has long been hypothesized to mediate endogenous circadian timekeeping. To test this model, we have specifically abrogated these sites of daily neuronal remodeling through the reprogramming of neural development and assessed the effects on circadian timekeeping and clock outputs. Remarkably, the loss of these sites has no measurable effects on endogenous circadian timekeeping or on any of the major output functions of the small ventral lateral neurons. Rather, their loss reduces sites of glutamatergic sensory neurotransmission that normally encodes naturalistic time cues from the environment. These results support an alternative model: structural plasticity in critical clock neurons is the basis for proper integration of light and temperature and gates sensory inputs into circadian clock neuron networks.


Subject(s)
Circadian Clocks/physiology , Circadian Rhythm/physiology , Drosophila melanogaster/physiology , Neuronal Plasticity/physiology , Animals
8.
Elife ; 92020 04 27.
Article in English | MEDLINE | ID: mdl-32338251

ABSTRACT

A transcription factor helps young flies to sleep longer by delaying the maturation of a neural network that controls sleep.


Subject(s)
Drosophila melanogaster , Sleep , Animals
9.
Sci Rep ; 8(1): 14452, 2018 09 27.
Article in English | MEDLINE | ID: mdl-30262912

ABSTRACT

Significant recent evidence suggests that metabolism is intricately linked to the regulation and dysfunction of complex cellular and physiological responses ranging from altered metabolic programs in cancers and aging to circadian rhythms and molecular clocks. While the metabolic pathways and their fundamental control mechanisms are well established, the precise cellular mechanisms underpinning, for example, enzymatic pathway control, substrate preferences or metabolic rates, remain far less certain. Comprehensive, continuous metabolic studies on model organisms, such as the fruit fly Drosophila melanogaster, may provide a critical tool for deciphering these complex physiological responses. Here, we describe the development of a high-resolution calorimeter, which combines sensitive thermometry with optical imaging to concurrently perform measurements of the metabolic rate of ten individual flies, in real-time, with ~100 nW resolution. Using this calorimeter we have measured the mass-specific metabolic rates of flies of different genotypes, ages, and flies fed with different diets. This powerful new approach enables systematic studies of the metabolic regulation related to cellular and physiological function and disease mechanisms.


Subject(s)
Calorimetry/methods , Circadian Rhythm , Optical Imaging/methods , Thermometry/methods , Animals , Calorimetry/instrumentation , Drosophila melanogaster , Optical Imaging/instrumentation , Thermometry/instrumentation
10.
Chronobiol Int ; 35(7): 1016-1026, 2018 07.
Article in English | MEDLINE | ID: mdl-29621409

ABSTRACT

The genetic, molecular and neuronal mechanism underlying circadian activity rhythms is well characterized in the brain of Drosophila. The small ventrolateral neurons (s-LNVs) and pigment dispersing factor (PDF) expressed by them are especially important for regulating circadian locomotion. Here we describe a novel gene, Dstac, which is similar to the stac genes found in vertebrates that encode adaptor proteins, which bind and regulate L-type voltage-gated Ca2+ channels (CaChs). We show that Dstac is coexpressed with PDF by the s-LNVs and regulates circadian activity. Furthermore, the L-type CaCh, Dmca1D, appears to be expressed by the s-LNVs. Since vertebrate Stac3 regulates an L-type CaCh we hypothesize that Dstac regulates Dmca1D in s-LNVs and circadian activity.


Subject(s)
Biological Clocks/physiology , Brain/metabolism , Circadian Rhythm/physiology , Drosophila Proteins/physiology , Drosophila melanogaster/metabolism , Intracellular Signaling Peptides and Proteins/physiology , Locomotion/physiology , Animals , Biological Clocks/genetics , Circadian Rhythm/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Intracellular Signaling Peptides and Proteins/genetics , Motor Activity/physiology , Neurons/metabolism
11.
Nature ; 555(7694): 98-102, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29466329

ABSTRACT

Circadian clocks coordinate behaviour, physiology and metabolism with Earth's diurnal cycle. These clocks entrain to both light and temperature cycles, and daily environmental temperature oscillations probably contribute to human sleep patterns. However, the neural mechanisms through which circadian clocks monitor environmental temperature and modulate behaviour remain poorly understood. Here we elucidate how the circadian clock neuron network of Drosophila melanogaster processes changes in environmental temperature. In vivo calcium-imaging techniques demonstrate that the posterior dorsal neurons 1 (DN1ps), which are a discrete subset of sleep-promoting clock neurons, constantly monitor modest changes in environmental temperature. We find that these neurons are acutely inhibited by heating and excited by cooling; this is an unexpected result when considering the strong correlation between temperature and light, and the fact that light excites clock neurons. We demonstrate that the DN1ps rely on peripheral thermoreceptors located in the chordotonal organs and the aristae. We also show that the DN1ps and their thermosensory inputs are required for the normal timing of sleep in the presence of naturalistic temperature cycles. These results identify the DN1ps as a major gateway for temperature sensation into the circadian neural network, which continuously integrates temperature changes to coordinate the timing of sleep and activity.


Subject(s)
Circadian Clocks/physiology , Circadian Rhythm/physiology , Drosophila melanogaster/physiology , Neurons/physiology , Sleep/physiology , Temperature , Thermosensing/physiology , Animals , Cold Temperature , Drosophila melanogaster/anatomy & histology , Drosophila melanogaster/cytology , Female , Hot Temperature , Locomotion/physiology , Male , Nerve Net/cytology , Nerve Net/physiology , Neural Inhibition , Thermoreceptors/metabolism , Time Factors
13.
Neuron ; 95(3): 623-638.e4, 2017 Aug 02.
Article in English | MEDLINE | ID: mdl-28712652

ABSTRACT

How experiences during development cause long-lasting changes in sensory circuits and affect behavior in mature animals are poorly understood. Here we establish a novel system for mechanistic analysis of the plasticity of developing neural circuits by showing that sensory experience during development alters nociceptive behavior and circuit physiology in Drosophila larvae. Despite the convergence of nociceptive and mechanosensory inputs on common second-order neurons (SONs), developmental noxious input modifies transmission from nociceptors to their SONs, but not from mechanosensors to the same SONs, which suggests striking sensory pathway specificity. These SONs activate serotonergic neurons to inhibit nociceptor-to-SON transmission; stimulation of nociceptors during development sensitizes nociceptor presynapses to this feedback inhibition. Our results demonstrate that, unlike associative learning, which involves inputs from two sensory pathways, sensory pathway-specific plasticity in the Drosophila nociceptive circuit is in part established through feedback modulation. This study elucidates a novel mechanism that enables pathway-specific plasticity in sensory systems. VIDEO ABSTRACT.


Subject(s)
Afferent Pathways/physiology , Behavior, Animal/physiology , Nerve Net/growth & development , Neuronal Plasticity/physiology , Nociceptors/metabolism , Serotonergic Neurons/metabolism , Animals , Drosophila melanogaster
14.
Elife ; 62017 05 30.
Article in English | MEDLINE | ID: mdl-28556778

ABSTRACT

Temperature-sensing neurons in the Drosophila brain cooperate with the central circadian clock to help regulate body temperature.


Subject(s)
Drosophila Proteins , Neuropeptides , Animals , Brain , Circadian Rhythm , Drosophila , Neurons , Temperature
15.
Cell Rep ; 17(11): 2873-2881, 2016 12 13.
Article in English | MEDLINE | ID: mdl-27974202

ABSTRACT

In animals, networks of clock neurons containing molecular clocks orchestrate daily rhythms in physiology and behavior. However, how various types of clock neurons communicate and coordinate with one another to produce coherent circadian rhythms is not well understood. Here, we investigate clock neuron coupling in the brain of Drosophila and demonstrate that the fly's various groups of clock neurons display unique and complex coupling relationships to core pacemaker neurons. Furthermore, we find that coordinated free-running rhythms require molecular clock synchrony not only within the well-characterized lateral clock neuron classes but also between lateral clock neurons and dorsal clock neurons. These results uncover unexpected patterns of coupling in the clock neuron network and reveal that robust free-running behavioral rhythms require a coherence of molecular oscillations across most of the fly's clock neuron network.


Subject(s)
Brain/physiology , Circadian Rhythm/genetics , Drosophila melanogaster/genetics , Neurons/physiology , Animals , Animals, Genetically Modified , Circadian Rhythm/physiology , Drosophila melanogaster/physiology , Nerve Net/physiology
16.
Curr Opin Insect Sci ; 1: 73-80, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-25386391

ABSTRACT

Though expressed in relatively few neurons in insect nervous systems, pigment-dispersing factor (PDF) plays many roles in the control of behavior and physiology. PDF's role in circadian timekeeping is its best-understood function and the focus of this review. Here we recount the isolation and characterization of insect PDFs, review the evidence that PDF acts as a circadian clock output factor, and discuss emerging models of how PDF functions within circadian clock neuron network of Drosophila, the species in which this peptide's circadian roles are best understood.

17.
PLoS Biol ; 12(10): e1001974, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25333796

ABSTRACT

Sleep, a reversible quiescent state found in both invertebrate and vertebrate animals, disconnects animals from their environment and is highly regulated for coordination with wakeful activities, such as reproduction. The fruit fly, Drosophila melanogaster, has proven to be a valuable model for studying the regulation of sleep by circadian clock and homeostatic mechanisms. Here, we demonstrate that the sex peptide receptor (SPR) of Drosophila, known for its role in female reproduction, is also important in stabilizing sleep in both males and females. Mutants lacking either the SPR or its central ligand, myoinhibitory peptide (MIP), fall asleep normally, but have difficulty in maintaining a sleep-like state. Our analyses have mapped the SPR sleep function to pigment dispersing factor (pdf) neurons, an arousal center in the insect brain. MIP downregulates intracellular cAMP levels in pdf neurons through the SPR. MIP is released centrally before and during night-time sleep, when the sleep drive is elevated. Sleep deprivation during the night facilitates MIP secretion from specific brain neurons innervating pdf neurons. Moreover, flies lacking either SPR or MIP cannot recover sleep after the night-time sleep deprivation. These results delineate a central neuropeptide circuit that stabilizes the sleep state by feeding a slow-acting inhibitory input into the arousal system and plays an important role in sleep homeostasis.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/physiology , Peptides/metabolism , Sleep/physiology , Animals , Brain/metabolism , Cyclic AMP/metabolism , Down-Regulation , Drosophila Proteins/genetics , Female , Gene Knockdown Techniques , Homeostasis , Male , Neurons/metabolism , Peptides/genetics , Receptors, Peptide
18.
PLoS Biol ; 12(9): e1001959, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25268747

ABSTRACT

Synchronized neuronal activity is vital for complex processes like behavior. Circadian pacemaker neurons offer an unusual opportunity to study synchrony as their molecular clocks oscillate in phase over an extended timeframe (24 h). To identify where, when, and how synchronizing signals are perceived, we first studied the minimal clock neural circuit in Drosophila larvae, manipulating either the four master pacemaker neurons (LNvs) or two dorsal clock neurons (DN1s). Unexpectedly, we found that the PDF Receptor (PdfR) is required in both LNvs and DN1s to maintain synchronized LNv clocks. We also found that glutamate is a second synchronizing signal that is released from DN1s and perceived in LNvs via the metabotropic glutamate receptor (mGluRA). Because simultaneously reducing Pdfr and mGluRA expression in LNvs severely dampened Timeless clock protein oscillations, we conclude that the master pacemaker LNvs require extracellular signals to function normally. These two synchronizing signals are released at opposite times of day and drive cAMP oscillations in LNvs. Finally we found that PdfR and mGluRA also help synchronize Timeless oscillations in adult s-LNvs. We propose that differentially timed signals that drive cAMP oscillations and synchronize pacemaker neurons in circadian neural circuits will be conserved across species.


Subject(s)
Circadian Clocks/genetics , Cyclic AMP/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Neurons/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, Metabotropic Glutamate/genetics , Animals , Circadian Rhythm/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Gene Expression Regulation , Glutamic Acid/metabolism , Larva/genetics , Larva/metabolism , Neurons/cytology , Neuropeptides/metabolism , Photoperiod , Receptors, G-Protein-Coupled/metabolism , Receptors, Metabotropic Glutamate/metabolism , Signal Transduction
19.
Proc Natl Acad Sci U S A ; 109(30): 12177-82, 2012 Jul 24.
Article in English | MEDLINE | ID: mdl-22778427

ABSTRACT

The role of the central neuropeptide pigment-dispersing factor (PDF) in circadian timekeeping in Drosophila is remarkably similar to that of vasoactive intestinal peptide (VIP) in mammals. Like VIP, PDF is expressed outside the circadian network by neurons innervating the gut, but the function and mode of action of this PDF have not been characterized. Here we investigate the visceral roles of PDF by adapting cellular and physiological methods to the study of visceral responses to PDF signaling in wild-type and mutant genetic backgrounds. We find that intestinal PDF acts at a distance on the renal system, where it regulates ureter contractions. We show that PdfR, PDF's established receptor, is expressed by the muscles of the excretory system, and present evidence that PdfR-induced cAMP increases underlie the myotropic effects of PDF. These findings extend the similarities between PDF and VIP beyond their shared central role as circadian regulators, and uncover an unexpected endocrine mode of myotropic action for an intestinal neuropeptide on the renal system.


Subject(s)
Circadian Rhythm/physiology , Drosophila Proteins/metabolism , Drosophila/physiology , Neuropeptides/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/physiology , Ureter/physiology , Animals , Cyclic AMP/metabolism , DNA Primers/genetics , Immunohistochemistry , Microscopy, Electron, Transmission , Real-Time Polymerase Chain Reaction
20.
J Neurophysiol ; 108(2): 684-96, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22539819

ABSTRACT

Drosophila melanogaster is a valuable model system for the neural basis of complex behavior, but an inability to routinely interrogate physiologic connections within central neural networks of the fly brain remains a fundamental barrier to progress in the field. To address this problem, we have introduced a simple method of measuring functional connectivity based on the independent expression of the mammalian P2X2 purinoreceptor and genetically encoded Ca(2+) and cAMP sensors within separate genetically defined subsets of neurons in the adult brain. We show that such independent expression is capable of specifically rendering defined sets of neurons excitable by pulses of bath-applied ATP in a manner compatible with high-resolution Ca(2+) and cAMP imaging in putative follower neurons. Furthermore, we establish that this approach is sufficiently sensitive for the detection of excitatory and modulatory connections deep within larval and adult brains. This technically facile approach can now be used in wild-type and mutant genetic backgrounds to address functional connectivity within neuronal networks governing a wide range of complex behaviors in the fly. Furthermore, the effectiveness of this approach in the fly brain suggests that similar methods using appropriate heterologous receptors might be adopted for other widely used model systems.


Subject(s)
Brain Mapping/methods , Brain/physiology , Drosophila melanogaster/physiology , Molecular Imaging/methods , Nerve Net/physiology , Neural Pathways/physiology , Animals
SELECTION OF CITATIONS
SEARCH DETAIL