Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Cell ; 178(4): 901-918.e16, 2019 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-31398343

RESUMEN

Physiology and metabolism are often sexually dimorphic, but the underlying mechanisms remain incompletely understood. Here, we use the intestine of Drosophila melanogaster to investigate how gut-derived signals contribute to sex differences in whole-body physiology. We find that carbohydrate handling is male-biased in a specific portion of the intestine. In contrast to known sexual dimorphisms in invertebrates, the sex differences in intestinal carbohydrate metabolism are extrinsically controlled by the adjacent male gonad, which activates JAK-STAT signaling in enterocytes within this intestinal portion. Sex reversal experiments establish roles for this male-biased intestinal metabolic state in controlling food intake and sperm production through gut-derived citrate. Our work uncovers a male gonad-gut axis coupling diet and sperm production, revealing that metabolic communication across organs is physiologically important. The instructive role of citrate in inter-organ communication might be significant in more biological contexts than previously recognized.


Asunto(s)
Metabolismo de los Hidratos de Carbono/fisiología , Drosophila melanogaster/metabolismo , Ingestión de Alimentos/fisiología , Mucosa Intestinal/metabolismo , Caracteres Sexuales , Maduración del Esperma/fisiología , Animales , Ácido Cítrico/metabolismo , Proteínas de Drosophila/metabolismo , Femenino , Expresión Génica , Quinasas Janus/metabolismo , Masculino , RNA-Seq , Factores de Transcripción STAT/metabolismo , Transducción de Señal , Azúcares/metabolismo , Testículo/metabolismo
2.
Nature ; 587(7834): 455-459, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33116314

RESUMEN

Reproduction induces increased food intake across females of many animal species1-4, providing a physiologically relevant paradigm for the exploration of appetite regulation. Here, by examining the diversity of enteric neurons in Drosophila melanogaster, we identify a key role for gut-innervating neurons with sex- and reproductive state-specific activity in sustaining the increased food intake of mothers during reproduction. Steroid and enteroendocrine hormones functionally remodel these neurons, which leads to the release of their neuropeptide onto the muscles of the crop-a stomach-like organ-after mating. Neuropeptide release changes the dynamics of crop enlargement, resulting in increased food intake, and preventing the post-mating remodelling of enteric neurons reduces both reproductive hyperphagia and reproductive fitness. The plasticity of enteric neurons is therefore key to reproductive success. Our findings provide a mechanism to attain the positive energy balance that sustains gestation, dysregulation of which could contribute to infertility or weight gain.


Asunto(s)
Drosophila melanogaster/citología , Drosophila melanogaster/fisiología , Ingestión de Alimentos/fisiología , Ingestión de Energía/fisiología , Madres , Neuronas/metabolismo , Reproducción/fisiología , Estructuras Animales/citología , Estructuras Animales/inervación , Estructuras Animales/metabolismo , Animales , Regulación del Apetito/fisiología , Femenino , Hiperfagia/metabolismo , Masculino , Neuropéptidos/metabolismo
3.
Learn Mem ; 31(5)2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38862167

RESUMEN

Providing metabolic support to neurons is now recognized as a major function of glial cells that is conserved from invertebrates to vertebrates. However, research in this field has focused for more than two decades on the relevance of lactate and glial glycolysis for neuronal energy metabolism, while overlooking many other facets of glial metabolism and their impact on neuronal physiology, circuit activity, and behavior. Here, we review recent work that has unveiled new features of glial metabolism, especially in Drosophila, in the modulation of behavioral traits involving the mushroom bodies (MBs). These recent findings reveal that spatially and biochemically distinct modes of glucose-derived neuronal fueling are implemented within the MB in a memory type-specific manner. In addition, cortex glia are endowed with several antioxidant functions, whereas astrocytes can serve as pro-oxidant agents that are beneficial to redox signaling underlying long-term memory. Finally, glial fatty acid oxidation seems to play a dual fail-safe role: first, as a mode of energy production upon glucose shortage, and, second, as a factor underlying the clearance of excessive oxidative load during sleep. Altogether, these integrated studies performed in Drosophila indicate that glial metabolism has a deterministic role on behavior.


Asunto(s)
Conducta Animal , Cuerpos Pedunculados , Neuroglía , Animales , Cuerpos Pedunculados/metabolismo , Cuerpos Pedunculados/fisiología , Neuroglía/metabolismo , Neuroglía/fisiología , Conducta Animal/fisiología , Drosophila , Metabolismo Energético/fisiología
4.
J Neurosci ; 40(21): 4219-4229, 2020 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-32303647

RESUMEN

In Drosophila, the mushroom bodies (MB) constitute the central brain structure for olfactory associative memory. As in mammals, the cAMP/PKA pathway plays a key role in memory formation. In the MB, Rutabaga (Rut) adenylate cyclase acts as a coincidence detector during associative conditioning to integrate calcium influx resulting from acetylcholine stimulation and G-protein activation resulting from dopaminergic stimulation. Amnesiac encodes a secreted neuropeptide required in the MB for two phases of aversive olfactory memory. Previous sequence analysis has revealed strong homology with the mammalian pituitary adenylate cyclase-activating peptide (PACAP). Here, we examined whether amnesiac is involved in cAMP/PKA dynamics in response to dopamine and acetylcholine co-stimulation in living flies. Experiments were conducted with both sexes, or with either sex. Our data show that amnesiac is necessary for the PKA activation process that results from coincidence detection in the MB. Since PACAP peptide is cleaved by the human membrane neprilysin hNEP, we searched for an interaction between Amnesiac and Neprilysin 1 (Nep1), a fly neprilysin involved in memory. We show that when Nep1 expression is acutely knocked down in adult MB, memory deficits displayed by amn hypomorphic mutants are rescued. Consistently, Nep1 inhibition also restores normal PKA activation in amn mutant flies. Taken together, the results suggest that Nep1 targets Amnesiac degradation to terminate its signaling function. Our work thus highlights a key role for Amnesiac in establishing within the MB the PKA dynamics that sustain middle-term memory (MTM) formation, a function modulated by Nep1.SIGNIFICANCE STATEMENT The Drosophila amnesiac gene encodes a secreted neuropeptide whose expression is required for specific memory phases in the mushroom bodies (MB), the olfactory memory center. Here, we show that Amnesiac is required for PKA activation resulting from coincidence detection, a mechanism by which the MB integrate two spatially distinct stimuli to encode associative memory. Furthermore, our results uncover a functional relationship between Amnesiac and Neprilysin 1 (Nep1), a membrane peptidase involved in memory and expressed in the MB. These results suggest that Nep1 modulates Amnesiac levels. We propose that on conditioning, Amnesiac release from the MB allows, via an autocrine process, the sustaining of PKA activation-mediating memory, which subsequently is inactivated by Nep1 degradation.


Asunto(s)
Reacción de Prevención/fisiología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Proteínas de Drosophila/genética , Memoria/fisiología , Cuerpos Pedunculados/metabolismo , Neprilisina/metabolismo , Neuropéptidos/genética , Animales , Animales Modificados Genéticamente , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Neuropéptidos/metabolismo , Olfato/fisiología
6.
Nature ; 488(7412): 512-6, 2012 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-22810589

RESUMEN

Animals approach stimuli that predict a pleasant outcome. After the paired presentation of an odour and a reward, Drosophila melanogaster can develop a conditioned approach towards that odour. Despite recent advances in understanding the neural circuits for associative memory and appetitive motivation, the cellular mechanisms for reward processing in the fly brain are unknown. Here we show that a group of dopamine neurons in the protocerebral anterior medial (PAM) cluster signals sugar reward by transient activation and inactivation of target neurons in intact behaving flies. These dopamine neurons are selectively required for the reinforcing property of, but not a reflexive response to, the sugar stimulus. In vivo calcium imaging revealed that these neurons are activated by sugar ingestion and the activation is increased on starvation. The output sites of the PAM neurons are mainly localized to the medial lobes of the mushroom bodies (MBs), where appetitive olfactory associative memory is formed. We therefore propose that the PAM cluster neurons endow a positive predictive value to the odour in the MBs. Dopamine in insects is known to mediate aversive reinforcement signals. Our results highlight the cellular specificity underlying the various roles of dopamine and the importance of spatially segregated local circuits within the MBs.


Asunto(s)
Neuronas Dopaminérgicas/fisiología , Drosophila melanogaster/citología , Drosophila melanogaster/fisiología , Memoria/fisiología , Odorantes/análisis , Recompensa , Animales , Conducta Apetitiva/fisiología , Señalización del Calcio , Dendritas/fisiología , Dopamina/metabolismo , Neuronas Dopaminérgicas/citología , Cuerpos Pedunculados/citología , Cuerpos Pedunculados/metabolismo , Olfato/genética , Olfato/fisiología
7.
Proc Natl Acad Sci U S A ; 112(2): 578-83, 2015 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-25548178

RESUMEN

Drosophila melanogaster can acquire a stable appetitive olfactory memory when the presentation of a sugar reward and an odor are paired. However, the neuronal mechanisms by which a single training induces long-term memory are poorly understood. Here we show that two distinct subsets of dopamine neurons in the fly brain signal reward for short-term (STM) and long-term memories (LTM). One subset induces memory that decays within several hours, whereas the other induces memory that gradually develops after training. They convey reward signals to spatially segregated synaptic domains of the mushroom body (MB), a potential site for convergence. Furthermore, we identified a single type of dopamine neuron that conveys the reward signal to restricted subdomains of the mushroom body lobes and induces long-term memory. Constant appetitive memory retention after a single training session thus comprises two memory components triggered by distinct dopamine neurons.


Asunto(s)
Neuronas Dopaminérgicas/fisiología , Drosophila melanogaster/fisiología , Animales , Animales Modificados Genéticamente , Conducta Apetitiva/fisiología , Carbohidratos , Drosophila melanogaster/anatomía & histología , Drosophila melanogaster/genética , Femenino , Aprendizaje/fisiología , Memoria a Largo Plazo/fisiología , Memoria a Corto Plazo/fisiología , Cuerpos Pedunculados/fisiología , Odorantes , Recompensa , Olfato/fisiología , Gusto/fisiología
8.
bioRxiv ; 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38948698

RESUMEN

Relevance-based selectivity and high energy cost are two distinct features of long-term memory (LTM) formation that warrant its default inhibition. Spaced repetition of learning is a highly conserved cognitive mechanism that can lift this inhibition. Here, we questioned how the spacing effect integrates experience selection and energy efficiency at the cellular and molecular levels. We showed in Drosophila that spaced training triggers LTM formation by extending over several hours an increased mitochondrial metabolic activity in neurons of the associative memory center, the mushroom bodies (MBs). We found that this effect is mediated by PKCδ, a member of the so-called 'novel PKC' family of enzymes, which uncovers the critical function of PKCδ in neurons as a regulator of mitochondrial metabolism for LTM. Additionally, PKCδ activation and translocation to mitochondria result from LTM-specific dopamine signaling on MB neurons. By bridging experience-dependent neuronal circuit activity with metabolic modulation of memory-encoding neurons, PKCδ signaling binds the cognitive and metabolic constraints underlying LTM formation into a unified gating mechanism.

9.
Curr Biol ; 34(9): 1904-1917.e6, 2024 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-38642548

RESUMEN

Neurons have differential and fluctuating energy needs across distinct cellular compartments, shaped by brain electrochemical activity associated with cognition. In vitro studies show that mitochondria transport from soma to axons is key to maintaining neuronal energy homeostasis. Nevertheless, whether the spatial distribution of neuronal mitochondria is dynamically adjusted in vivo in an experience-dependent manner remains unknown. In Drosophila, associative long-term memory (LTM) formation is initiated by an early and persistent upregulation of mitochondrial pyruvate flux in the axonal compartment of neurons in the mushroom body (MB). Through behavior experiments, super-resolution analysis of mitochondria morphology in the neuronal soma and in vivo mitochondrial fluorescence recovery after photobleaching (FRAP) measurements in the axons, we show that LTM induction, contrary to shorter-lived memories, is sustained by the departure of some mitochondria from MB neuronal soma and increased mitochondrial dynamics in the axonal compartment. Accordingly, impairing mitochondrial dynamics abolished the increased pyruvate consumption, specifically after spaced training and in the MB axonal compartment, thereby preventing LTM formation. Our results thus promote reorganization of the mitochondrial network in neurons as an integral step in elaborating high-order cognitive processes.


Asunto(s)
Memoria a Largo Plazo , Dinámicas Mitocondriales , Cuerpos Pedunculados , Animales , Axones/metabolismo , Axones/fisiología , Drosophila melanogaster/fisiología , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Memoria a Largo Plazo/fisiología , Mitocondrias/metabolismo , Mitocondrias/fisiología , Dinámicas Mitocondriales/fisiología , Cuerpos Pedunculados/fisiología , Cuerpos Pedunculados/metabolismo , Neuronas/metabolismo , Neuronas/fisiología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas de Unión al GTP rho/genética , Proteínas de Unión al GTP rho/metabolismo
10.
Nat Metab ; 5(11): 2002-2019, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37932430

RESUMEN

Glucose is the primary source of energy for the brain; however, it remains controversial whether, upon neuronal activation, glucose is primarily used by neurons for ATP production or if it is partially oxidized in astrocytes, as proposed by the astrocyte-neuron lactate shuttle model for glutamatergic neurons. Thus, an in vivo picture of glucose metabolism during cognitive processes is missing. Here, we uncover in Drosophila melanogaster a glia-to-neuron alanine transfer involving alanine aminotransferase that sustains memory formation. Following associative conditioning, glycolysis in glial cells produces alanine, which is back-converted into pyruvate in cholinergic neurons of the olfactory memory center to uphold their increased mitochondrial needs. Alanine, as a mediator of glia-neuron coupling, could be an alternative to lactate in cholinergic systems. In parallel, a dedicated glial glucose transporter imports glucose specifically for long-term memory, by directly transferring it to neurons for use by the pentose phosphate pathway. Our results demonstrate in vivo the compartmentalization of glucose metabolism between neurons and glial cells during memory formation.


Asunto(s)
Alanina , Drosophila , Animales , Drosophila/metabolismo , Alanina/metabolismo , Drosophila melanogaster , Neuroglía/metabolismo , Glucólisis , Neuronas/metabolismo , Glucosa/metabolismo , Mitocondrias/metabolismo , Ácido Láctico/metabolismo
11.
bioRxiv ; 2023 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-37333418

RESUMEN

During neuronal circuit formation, local control of axonal organelles ensures proper synaptic connectivity. Whether this process is genetically encoded is unclear and if so, its developmental regulatory mechanisms remain to be identified. We hypothesized that developmental transcription factors regulate critical parameters of organelle homeostasis that contribute to circuit wiring. We combined cell type-specific transcriptomics with a genetic screen to discover such factors. We identified Telomeric Zinc finger-Associated Protein (TZAP) as a temporal developmental regulator of neuronal mitochondrial homeostasis genes, including Pink1 . In Drosophila , loss of dTzap function during visual circuit development leads to loss of activity-dependent synaptic connectivity, that can be rescued by Pink1 expression. At the cellular level, loss of dTzap/TZAP leads to defects in mitochondrial morphology, attenuated calcium uptake and reduced synaptic vesicle release in fly and mammalian neurons. Our findings highlight developmental transcriptional regulation of mitochondrial homeostasis as a key factor in activity-dependent synaptic connectivity.

12.
Life Sci Alliance ; 6(4)2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36720500

RESUMEN

FTSJ1 is a conserved human 2'-O-methyltransferase (Nm-MTase) that modifies several tRNAs at position 32 and the wobble position 34 in the anticodon loop. Its loss of function has been linked to X-linked intellectual disability (XLID), and more recently to cancers. However, the molecular mechanisms underlying these pathologies are currently unclear. Here, we report a novel FTSJ1 pathogenic variant from an X-linked intellectual disability patient. Using blood cells derived from this patient and other affected individuals carrying FTSJ1 mutations, we performed an unbiased and comprehensive RiboMethSeq analysis to map the ribose methylation on all human tRNAs and identify novel targets. In addition, we performed a transcriptome analysis in these cells and found that several genes previously associated with intellectual disability and cancers were deregulated. We also found changes in the miRNA population that suggest potential cross-regulation of some miRNAs with these key mRNA targets. Finally, we show that differentiation of FTSJ1-depleted human neural progenitor cells into neurons displays long and thin spine neurites compared with control cells. These defects are also observed in Drosophila and are associated with long-term memory deficits. Altogether, our study adds insight into FTSJ1 pathologies in humans and flies by the identification of novel FTSJ1 targets and the defect in neuron morphology.


Asunto(s)
Discapacidad Intelectual , Ribosa , Humanos , Metilación , Discapacidad Intelectual/genética , Metiltransferasas/genética , ARN de Transferencia/genética , ARN de Transferencia/metabolismo , Neuronas/metabolismo , Proteínas Nucleares/genética
13.
Nat Metab ; 4(2): 213-224, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35177854

RESUMEN

During starvation, mammalian brains can adapt their metabolism, switching from glucose to alternative peripheral fuel sources. In the Drosophila starved brain, memory formation is subject to adaptative plasticity, but whether this adaptive plasticity relies on metabolic adaptation remains unclear. Here we show that during starvation, neurons of the fly olfactory memory centre import and use ketone bodies (KBs) as an energy substrate to sustain aversive memory formation. We identify local providers within the brain, the cortex glia, that use their own lipid store to synthesize KBs before exporting them to neurons via monocarboxylate transporters. Finally, we show that the master energy sensor AMP-activated protein kinase regulates both lipid mobilization and KB export in cortex glia. Our data provide a general schema of the metabolic interactions within the brain to support memory when glucose is scarce.


Asunto(s)
Cuerpos Cetónicos , Inanición , Animales , Drosophila/metabolismo , Glucosa/metabolismo , Cuerpos Cetónicos/metabolismo , Mamíferos/metabolismo , Neuroglía/metabolismo , Neuronas/metabolismo , Inanición/metabolismo
14.
Cell Rep ; 36(8): 109620, 2021 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-34433052

RESUMEN

Brain function relies almost solely on glucose as an energy substrate. The main model of brain metabolism proposes that glucose is taken up and converted into lactate by astrocytes to fuel the energy-demanding neuronal activity underlying plasticity and memory. Whether direct neuronal glucose uptake is required for memory formation remains elusive. We uncover, in Drosophila, a mechanism of glucose shuttling to neurons from cortex glia, an exclusively perisomatic glial subtype, upon formation of olfactory long-term memory (LTM). In vivo imaging reveals that, downstream of cholinergic activation of cortex glia, autocrine insulin signaling increases glucose concentration in glia. Glucose is then transferred from glia to the neuronal somata in the olfactory memory center to fuel the pentose phosphate pathway and allow LTM formation. In contrast, our results indicate that the increase in neuronal glucose metabolism, although crucial for LTM formation, is not routed to glycolysis.


Asunto(s)
Memoria a Largo Plazo/fisiología , Neuroglía/metabolismo , Neuronas/metabolismo , Vía de Pentosa Fosfato/fisiología , Animales , Astrocitos/metabolismo , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , Glucosa/metabolismo
15.
Neuron ; 104(3): 432-435, 2019 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-31697917

RESUMEN

Perseverance in foraging is a high-risk/high-gain strategy. In this issue of Neuron, Sayin et al. (2019) decipher the neuronal circuit that arbitrates this choice in Drosophila. The fly's remarkable tenacity illuminates the interaction between working memory and decision making.


Asunto(s)
Proteínas de Drosophila , Drosophila , Animales , Atención , Memoria a Corto Plazo , Neuronas
16.
Curr Biol ; 28(11): 1783-1793.e4, 2018 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-29779874

RESUMEN

Memory consolidation is a crucial step for long-term memory (LTM) storage. However, we still lack a clear picture of how memory consolidation is regulated at the neuronal circuit level. Here, we took advantage of the well-described anatomy of the Drosophila olfactory memory center, the mushroom body (MB), to address this question in the context of appetitive LTM. The MB lobes, which are made by the fascicled axons of the MB intrinsic neurons, are organized into discrete anatomical modules, each covered by the terminals of a defined type of dopaminergic neuron (DAN) and the dendrites of a corresponding type of MB output neuron (MBON). We previously revealed the essential role of one DAN, the MP1 neuron, in the formation of appetitive LTM. The MP1 neuron is anatomically matched to the GABAergic MBON MVP2, which has been attributed feedforward inhibitory functions recently. Here, we used behavior experiments and in vivo imaging to challenge the existence of MP1-MVP2 synapses and investigate their role in appetitive LTM consolidation. We show that MP1 and MVP2 neurons form an anatomically and functionally recurrent circuit, which features a feedback inhibition that regulates consolidation of appetitive memory. This circuit involves two opposite type 1 and type 2 dopamine receptors in MVP2 neurons and the metabotropic GABAB-R1 receptor in MP1 neurons. We propose that this dual-receptor feedback supports a bidirectional self-regulation of MP1 input to the MB. This mechanism displays striking similarities with the mammalian reward system, in which modulation of the dopaminergic signal is primarily assigned to inhibitory neurons.


Asunto(s)
Dopamina/metabolismo , Neuronas Dopaminérgicas/metabolismo , Drosophila/fisiología , Neuronas GABAérgicas/fisiología , Memoria a Largo Plazo/fisiología , Cuerpos Pedunculados/fisiología , Percepción Olfatoria/fisiología , Animales , Neuronas GABAérgicas/efectos de los fármacos , Memoria a Largo Plazo/efectos de los fármacos , Cuerpos Pedunculados/efectos de los fármacos , Odorantes
17.
Neuron ; 98(2): 350-365.e5, 2018 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-29673482

RESUMEN

A key function of the brain is to filter essential information and store it in the form of stable, long-term memory (LTM). We demonstrate here that the Dunce (Dnc) phosphodiesterase, an important enzyme that degrades cAMP, acts as a molecular switch that controls LTM formation in Drosophila. We show that, during LTM formation, Dnc is inhibited in the SPN, a pair of newly characterized serotonergic neurons, which stimulates the cAMP/PKA pathway. As a consequence, the SPN activates downstream dopaminergic neurons, opening the gate for LTM formation in the olfactory memory center, the mushroom body. Strikingly, transient inhibition of Dnc in the SPN by RNAi was sufficient to induce LTM formation with a training protocol that normally generates only short-lived memory. Thus, Dnc activity in the SPN acts as a memory checkpoint to guarantee that only the most relevant learned experiences are consolidated into stable memory.


Asunto(s)
Proteínas de Drosophila/metabolismo , Locomoción/fisiología , Memoria a Largo Plazo/fisiología , Neuronas Serotoninérgicas/metabolismo , Animales , Animales Modificados Genéticamente , Proteínas de Drosophila/análisis , Drosophila melanogaster , Femenino , Neuronas Serotoninérgicas/química
18.
Neuron ; 100(3): 651-668.e8, 2018 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-30244885

RESUMEN

The behavioral response to a sensory stimulus may depend on both learned and innate neuronal representations. How these circuits interact to produce appropriate behavior is unknown. In Drosophila, the lateral horn (LH) and mushroom body (MB) are thought to mediate innate and learned olfactory behavior, respectively, although LH function has not been tested directly. Here we identify two LH cell types (PD2a1 and PD2b1) that receive input from an MB output neuron required for recall of aversive olfactory memories. These neurons are required for aversive memory retrieval and modulated by training. Connectomics data demonstrate that PD2a1 and PD2b1 neurons also receive direct input from food odor-encoding neurons. Consistent with this, PD2a1 and PD2b1 are also necessary for unlearned attraction to some odors, indicating that these neurons have a dual behavioral role. This provides a circuit mechanism by which learned and innate olfactory information can interact in identified neurons to produce appropriate behavior. VIDEO ABSTRACT.


Asunto(s)
Memoria/fisiología , Recuerdo Mental/fisiología , Cuerpos Pedunculados/fisiología , Red Nerviosa/fisiología , Odorantes , Olfato/fisiología , Animales , Animales Modificados Genéticamente , Conectoma/métodos , Drosophila , Cuerpos Pedunculados/química , Red Nerviosa/química
19.
Nat Commun ; 8: 15510, 2017 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-28580949

RESUMEN

Efficient energy use has constrained the evolution of nervous systems. However, it is unresolved whether energy metabolism may resultantly regulate major brain functions. Our observation that Drosophila flies double their sucrose intake at an early stage of long-term memory formation initiated the investigation of how energy metabolism intervenes in this process. Cellular-resolution imaging of energy metabolism reveals a concurrent elevation of energy consumption in neurons of the mushroom body, the fly's major memory centre. Strikingly, upregulation of mushroom body energy flux is both necessary and sufficient to drive long-term memory formation. This effect is triggered by a specific pair of dopaminergic neurons afferent to the mushroom bodies, via the D5-like DAMB dopamine receptor. Hence, dopamine signalling mediates an energy switch in the mushroom body that controls long-term memory encoding. Our data thus point to an instructional role for energy flux in the execution of demanding higher brain functions.


Asunto(s)
Drosophila melanogaster/metabolismo , Memoria a Largo Plazo/fisiología , Memoria/fisiología , Cuerpos Pedunculados/metabolismo , Animales , Animales Modificados Genéticamente , Conducta Animal/fisiología , Encéfalo/fisiología , Condicionamiento Clásico/fisiología , Dopamina/metabolismo , Neuronas Dopaminérgicas/metabolismo , Proteínas de Drosophila/metabolismo , Metabolismo Energético , Femenino , Genotipo , Masculino , Neuronas/metabolismo , Receptores Dopaminérgicos/metabolismo , Olfato/fisiología , Sacarosa/química , Activación Transcripcional , Regulación hacia Arriba
20.
Cell Rep ; 11(8): 1280-92, 2015 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-25981036

RESUMEN

Understanding how the various memory components are encoded and how they interact to guide behavior requires knowledge of the underlying neural circuits. Currently, aversive olfactory memory in Drosophila is behaviorally subdivided into four discrete phases. Among these, short- and long-term memories rely, respectively, on the γ and α/ß Kenyon cells (KCs), two distinct subsets of the ∼2,000 neurons in the mushroom body (MB). Whereas V2 efferent neurons retrieve memory from α/ß KCs, the neurons that retrieve short-term memory are unknown. We identified a specific pair of MB efferent neurons, named M6, that retrieve memory from γ KCs. Moreover, our network analysis revealed that six discrete memory phases actually exist, three of which have been conflated in the past. At each time point, two distinct memory components separately recruit either V2 or M6 output pathways. Memory retrieval thus features a dramatic convergence from KCs to MB efferent neurons.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Memoria/fisiología , Cuerpos Pedunculados/metabolismo , Neuronas/metabolismo , Animales , Animales Modificados Genéticamente , Femenino
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA