Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
R Soc Open Sci ; 11(2): 231671, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38384778

RESUMEN

The spotted lanternfly is an emerging global invasive insect pest. Due to a lack of natural enemies where it is invasive, human intervention is required. Extensive management has been applied but the spread continues. Recently, the idea of bird-based biological controls has re-emerged and shown effective in studies. However, it is questionable, if birds are able to effectively control unfamiliar and occasionally toxic invasive pests in short timeframes. Unless, perhaps, the birds are effective social learners and toxicity of the invaders is rare. Here, we introduce a mathematical model for social learning in a great tit-like bird to investigate conditions for the emergence of a collective biological control of a pest that is occasionally toxic, like the lanternfly. We find that the social observation rate relative to the proportion of toxic lanternfly dictate when collective biological controls will emerge. We also implement the social learning model into a model of collective motion in bird-like animals, and find that it produces results consistent with the mathematical model. Our work suggests that social birds may be useful in managing the spotted lanternfly, and that removing the toxicity-inducing preferred host of the lanternfly should be a priority to facilitate this.

2.
Sci Rep ; 13(1): 22600, 2023 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-38114694

RESUMEN

Many models developed to forecast and attempt to understand the COVID-19 pandemic are highly complex, and few take collective behavior into account. As the pandemic progressed individual recurrent infection was observed and simpler susceptible-infected type models were introduced. However, these do not include mechanisms to model collective behavior. Here, we introduce an extension of the SIS model that accounts for collective behavior and show that it has four equilibria. Two of the equilibria are the standard SIS model equilibria, a third is always unstable, and a fourth where collective behavior and infection prevalence interact to produce either node-like or oscillatory dynamics. We then parameterized the model using estimates of the transmission and recovery rates for COVID-19 and present phase diagrams for fixed recovery rate and free transmission rate, and both rates fixed. We observe that regions of oscillatory dynamics exist in both cases and that the collective behavior parameter regulates their extent. Finally, we show that the system exhibits hysteresis when the collective behavior parameter varies over time. This model provides a minimal framework for explaining oscillatory phenomena such as recurring waves of infection and hysteresis effects observed in COVID-19, and other SIS-type epidemics, in terms of collective behavior.


Asunto(s)
COVID-19 , Enfermedades Transmisibles , Humanos , Enfermedades Transmisibles/epidemiología , Pandemias , COVID-19/epidemiología , Susceptibilidad a Enfermedades/epidemiología , Predicción
3.
bioRxiv ; 2023 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-37986870

RESUMEN

In Drosophila melanogaster, the P1 (pC1) cluster of male-specific neurons both integrates sensory cues and drives or modulates behavioral programs such as courtship, in addition to contributing to a social arousal state. The behavioral function of these neurons is linked to the genes they express, which underpin their capacity for synaptic signaling, neuromodulation, and physiology. Yet, P1 (pC1) neurons have not been fully characterized at the transcriptome level. Moreover, it is unknown how the molecular landscape of P1 (pC1) neurons acutely changes after flies engage in social behaviors, where baseline P1 (pC1) neural activity is expected to increase. To address these two gaps, we use single cell-type RNA sequencing to profile and compare the transcriptomes of P1 (pC1) neurons harvested from socially paired versus solitary male flies. Compared to control transcriptome datasets, we find that P1 (pC1) neurons are enriched in 2,665 genes, including those encoding receptors, neuropeptides, and cell-adhesion molecules (dprs/DIPs). Furthermore, courtship is characterized by changes in ~300 genes, including those previously implicated in regulating behavior (e.g. DopEcR, Octß3R, Fife, kairos, rad). Finally, we identify a suite of genes that link conspecific courtship with the innate immune system. Together, these data serve as a molecular map for future studies of an important set of higher-order and sexually-dimorphic neurons.

4.
Hum Mol Genet ; 33(1): 12-32, 2023 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-37712894

RESUMEN

Genes mutated in monogenic neurodevelopmental disorders are broadly expressed. This observation supports the concept that monogenic neurodevelopmental disorders are systemic diseases that profoundly impact neurodevelopment. We tested the systemic disease model focusing on Rett syndrome, which is caused by mutations in MECP2. Transcriptomes and proteomes of organs and brain regions from Mecp2-null mice as well as diverse MECP2-null male and female human cells were assessed. Widespread changes in the steady-state transcriptome and proteome were identified in brain regions and organs of presymptomatic Mecp2-null male mice as well as mutant human cell lines. The extent of these transcriptome and proteome modifications was similar in cortex, liver, kidney, and skeletal muscle and more pronounced than in the hippocampus and striatum. In particular, Mecp2- and MECP2-sensitive proteomes were enriched in synaptic and metabolic annotated gene products, the latter encompassing lipid metabolism and mitochondrial pathways. MECP2 mutations altered pyruvate-dependent mitochondrial respiration while maintaining the capacity to use glutamine as a mitochondrial carbon source. We conclude that mutations in Mecp2/MECP2 perturb lipid and mitochondrial metabolism systemically limiting cellular flexibility to utilize mitochondrial fuels.


Asunto(s)
Proteoma , Síndrome de Rett , Animales , Femenino , Humanos , Masculino , Ratones , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Proteína 2 de Unión a Metil-CpG/genética , Proteína 2 de Unión a Metil-CpG/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Proteoma/genética , Proteoma/metabolismo , Síndrome de Rett/genética , Síndrome de Rett/metabolismo
5.
Elife ; 122023 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-37171075

RESUMEN

Mitochondria influence cellular function through both cell-autonomous and non-cell autonomous mechanisms, such as production of paracrine and endocrine factors. Here, we demonstrate that mitochondrial regulation of the secretome is more extensive than previously appreciated, as both genetic and pharmacological disruption of the electron transport chain caused upregulation of the Alzheimer's disease risk factor apolipoprotein E (APOE) and other secretome components. Indirect disruption of the electron transport chain by gene editing of SLC25A mitochondrial membrane transporters as well as direct genetic and pharmacological disruption of either complexes I, III, or the copper-containing complex IV of the electron transport chain elicited upregulation of APOE transcript, protein, and secretion, up to 49-fold. These APOE phenotypes were robustly expressed in diverse cell types and iPSC-derived human astrocytes as part of an inflammatory gene expression program. Moreover, age- and genotype-dependent decline in brain levels of respiratory complex I preceded an increase in APOE in the 5xFAD mouse model. We propose that mitochondria act as novel upstream regulators of APOE-dependent cellular processes in health and disease.


Asunto(s)
Apolipoproteína E4 , Mitocondrias , Animales , Humanos , Ratones , Apolipoproteína E4/genética , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Astrocitos/metabolismo , Genotipo , Mitocondrias/metabolismo , Mitocondrias/patología
6.
bioRxiv ; 2023 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-37066332

RESUMEN

Genes mutated in monogenic neurodevelopmental disorders are broadly expressed. This observation supports the concept that monogenic neurodevelopmental disorders are systemic diseases that profoundly impact neurodevelopment. We tested the systemic disease model focusing on Rett syndrome, which is caused by mutations in MECP2. Transcriptomes and proteomes of organs and brain regions from Mecp2-null mice as well as diverse MECP2-null male and female human cells were assessed. Widespread changes in the steady-state transcriptome and proteome were identified in brain regions and organs of presymptomatic Mecp2-null male mice as well as mutant human cell lines. The extent of these transcriptome and proteome modifications was similar in cortex, liver, kidney, and skeletal muscle and more pronounced than in the hippocampus and striatum. In particular, Mecp2- and MECP2-sensitive proteomes were enriched in synaptic and metabolic annotated gene products, the latter encompassing lipid metabolism and mitochondrial pathways. MECP2 mutations altered pyruvate-dependent mitochondrial respiration while maintaining the capacity to use glutamine as a mitochondrial carbon source. We conclude that mutations in Mecp2/MECP2 perturb lipid and mitochondrial metabolism systemically limiting cellular flexibility to utilize mitochondrial fuels.

7.
eNeuro ; 8(4)2021.
Artículo en Inglés | MEDLINE | ID: mdl-34312306

RESUMEN

Mitochondrial composition varies by organ and their constituent cell types. This mitochondrial diversity likely determines variations in mitochondrial function. However, the heterogeneity of mitochondria in the brain remains underexplored despite the large diversity of cell types in neuronal tissue. Here, we used molecular systems biology tools to address whether mitochondrial composition varies by brain region and neuronal cell type in mice. We reasoned that proteomics and transcriptomics of microdissected brain regions combined with analysis of single-cell mRNA sequencing (scRNAseq) could reveal the extent of mitochondrial compositional diversity. We selected nuclear encoded gene products forming complexes of fixed stoichiometry, such as the respiratory chain complexes and the mitochondrial ribosome, as well as molecules likely to perform their function as monomers, such as the family of SLC25 transporters. We found that the proteome encompassing these nuclear-encoded mitochondrial genes and obtained from microdissected brain tissue segregated the hippocampus, striatum, and cortex from each other. Nuclear-encoded mitochondrial transcripts could only segregate cell types and brain regions when the analysis was performed at the single-cell level. In fact, single-cell mitochondrial transcriptomes were able to distinguish glutamatergic and distinct types of GABAergic neurons from one another. Within these cell categories, unique SLC25A transporters were able to identify distinct cell subpopulations. Our results demonstrate heterogeneous mitochondrial composition across brain regions and cell types. We postulate that mitochondrial heterogeneity influences regional and cell type-specific mechanisms in health and disease.


Asunto(s)
Genes Mitocondriales , Neuronas , Animales , Núcleo Celular , Hipocampo , Ratones , Mitocondrias/genética , Neuronas/metabolismo
8.
Front Integr Neurosci ; 13: 30, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31379529

RESUMEN

Neurodevelopmental disorders represent a challenging biological and medical problem due to their genetic and phenotypic complexity. In many cases, we lack the comprehensive understanding of disease mechanisms necessary for targeted therapeutic development. One key component that could improve both mechanistic understanding and clinical trial design is reliable molecular biomarkers. Presently, no objective biological markers exist to evaluate most neurodevelopmental disorders. Here, we discuss how systems biology and "omic" approaches can address the mechanistic and biomarker limitations in these afflictions. We present heuristic principles for testing the potential of systems biology to identify mechanisms and biomarkers of disease in the example of Rett syndrome, a neurodevelopmental disorder caused by a well-defined monogenic defect in methyl-CpG-binding protein 2 (MECP2). We propose that such an approach can not only aid in monitoring clinical disease severity but also provide a measure of target engagement in clinical trials. By deepening our understanding of the "big picture" of systems biology, this approach could even help generate hypotheses for drug development programs, hopefully resulting in new treatments for these devastating conditions.

9.
Behav Brain Funct ; 15(1): 7, 2019 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-30992041

RESUMEN

Our understanding of the networks of genes and protein functions involved in Alcohol Use Disorder (AUD) remains incomplete, as do the mechanisms by which these networks lead to AUD phenotypes. The fruit fly (Drosophila melanogaster) is an efficient model for functional and mechanistic characterization of the genes involved in alcohol behavior. The fly offers many advantages as a model organism for investigating the molecular and cellular mechanisms of alcohol-related behaviors, and for understanding the underlying neural circuitry driving behaviors, such as locomotor stimulation, sedation, tolerance, and appetitive (reward) learning and memory. Fly researchers are able to use an extensive variety of tools for functional characterization of gene products. To understand how the fly can guide our understanding of AUD in the era of Big Data we will explore these tools, and review some of the gene networks identified in the fly through their use, including chromatin-remodeling, glial, cellular stress, and innate immunity genes. These networks hold great potential as translational drug targets, making it prudent to conduct further research into how these gene mechanisms are involved in alcohol behavior.


Asunto(s)
Alcoholismo/genética , Alcoholismo/metabolismo , Conducta Animal/efectos de los fármacos , Animales , Conducta Animal/fisiología , Macrodatos , Modelos Animales de Enfermedad , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Etanol/metabolismo , Aprendizaje/efectos de los fármacos , Memoria/efectos de los fármacos , Recompensa
10.
J Neurosci ; 39(18): 3561-3581, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30833507

RESUMEN

Neurodevelopmental disorders offer insight into synaptic mechanisms. To unbiasedly uncover these mechanisms, we studied the 22q11.2 syndrome, a recurrent copy number variant, which is the highest schizophrenia genetic risk factor. We quantified the proteomes of 22q11.2 mutant human fibroblasts from both sexes and mouse brains carrying a 22q11.2-like defect, Df(16)A+/- Molecular ontologies defined mitochondrial compartments and pathways as some of top ranked categories. In particular, we identified perturbations in the SLC25A1-SLC25A4 mitochondrial transporter interactome as associated with the 22q11.2 genetic defect. Expression of SLC25A1-SLC25A4 interactome components was affected in neuronal cells from schizophrenia patients. Furthermore, hemideficiency of the Drosophila SLC25A1 or SLC25A4 orthologues, dSLC25A1-sea and dSLC25A4-sesB, affected synapse morphology, neurotransmission, plasticity, and sleep patterns. Our findings indicate that synapses are sensitive to partial loss of function of mitochondrial solute transporters. We propose that mitoproteomes regulate synapse development and function in normal and pathological conditions in a cell-specific manner.SIGNIFICANCE STATEMENT We address the central question of how to comprehensively define molecular mechanisms of the most prevalent and penetrant microdeletion associated with neurodevelopmental disorders, the 22q11.2 microdeletion syndrome. This complex mutation reduces gene dosage of ∼63 genes in humans. We describe a disruption of the mitoproteome in 22q11.2 patients and brains of a 22q11.2 mouse model. In particular, we identify a network of inner mitochondrial membrane transporters as a hub required for synapse function. Our findings suggest that mitochondrial composition and function modulate the risk of neurodevelopmental disorders, such as schizophrenia.


Asunto(s)
Síndrome de Deleción 22q11/metabolismo , Encéfalo/metabolismo , Mitocondrias/metabolismo , Neuronas/metabolismo , Sinapsis/metabolismo , Translocador 1 del Nucleótido Adenina/metabolismo , Animales , Conducta Animal , Línea Celular , Deleción Cromosómica , Cromosomas Humanos Par 22/metabolismo , Drosophila , Femenino , Fibroblastos/metabolismo , Humanos , Masculino , Proteínas Mitocondriales/metabolismo , Transportadores de Anión Orgánico/metabolismo , Proteoma , Esquizofrenia/metabolismo
11.
Cell Rep ; 15(7): 1580-1596, 2016 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-27160913

RESUMEN

Learning and memory formation in Drosophila rely on a network of neurons in the mushroom bodies (MBs). Whereas numerous studies have delineated roles for individual cell types within this network in aspects of learning or memory, whether or not these cells can also be distinguished by the genes they express remains unresolved. In addition, the changes in gene expression that accompany long-term memory formation within the MBs have not yet been studied by neuron type. Here, we address both issues by performing RNA sequencing on single cell types (harvested via patch pipets) within the MB. We discover that the expression of genes that encode cell surface receptors is sufficient to identify cell types and that a subset of these genes, required for sensory transduction in peripheral sensory neurons, is not only expressed within individual neurons of the MB in the central brain, but is also critical for memory formation.


Asunto(s)
Drosophila melanogaster/citología , Drosophila melanogaster/genética , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica , Memoria , Cuerpos Pedunculados/citología , Cuerpos Pedunculados/metabolismo , Animales , Ontología de Genes , Genes de Insecto , Neuronas/citología , Neuronas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Reproducibilidad de los Resultados , Análisis de Secuencia de ARN
12.
J Biol Chem ; 287(39): 32406-14, 2012 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-22829591

RESUMEN

Sleep length and metabolic dysfunction are correlated, but the causal relationship between these processes is unclear. Octopamine promotes wakefulness in the fly by acting through the insulin-producing cells (IPCs) in the fly brain. To determine if insulin signaling mediates the effects of octopamine on sleep:wake behavior, we assayed flies in which insulin signaling activity was genetically altered. We found that increasing insulin signaling does not promote wake, nor does insulin appear to mediate the wake-promoting effects of octopamine. Octopamine also affects metabolism in invertebrate species, including, as we show here, Drosophila melanogaster. Triglycerides are decreased in mutants with compromised octopamine signaling and elevated in flies with increased activity of octopaminergic neurons. Interestingly, this effect is mediated at least partially by insulin, suggesting that effects of octopamine on metabolism are independent of its effects on sleep. We further investigated the relative contribution of metabolic and sleep phenotypes to the starvation response of flies with altered octopamine signaling. Hyperactivity (indicative of foraging) induced by starvation was elevated in octopamine receptor mutants, despite their high propensity for sleep, indicating that their metabolic state dictates their behavioral response under these conditions. Moreover, flies with increased octopamine signaling do not suppress sleep in response to starvation, even though they are normally hyper-aroused, most likely because of their high triglyceride levels. Together, these data suggest that observed correlations between sleep and metabolic phenotypes can result from shared molecular pathways rather than causality, and environmental conditions can lead to the dominance of one phenotype over the other.


Asunto(s)
Octopamina/metabolismo , Transducción de Señal/fisiología , Sueño/fisiología , Animales , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Insulina/genética , Insulina/metabolismo , Mutación , Octopamina/genética , Receptores de Amina Biogénica/genética , Receptores de Amina Biogénica/metabolismo , Triglicéridos/genética , Triglicéridos/metabolismo
13.
PLoS One ; 6(5): e19921, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21625640

RESUMEN

A proper balance of lipid breakdown and synthesis is essential for achieving energy homeostasis as alterations in either of these processes can lead to pathological states such as obesity. The regulation of lipid metabolism is quite complex with multiple signals integrated to control overall triglyceride levels in metabolic tissues. Based upon studies demonstrating effects of the circadian clock on metabolism, we sought to determine if the central clock cells in the Drosophila brain contribute to lipid levels in the fat body, the main nutrient storage organ of the fly. Here, we show that altering the function of the Drosophila central clock neurons leads to an increase in fat body triglycerides. We also show that although triglyceride levels are not affected by age, they are increased by expression of the amyloid-beta protein in central clock neurons. The effect on lipid storage seems to be independent of circadian clock output as changes in triglycerides are not always observed in genetic manipulations that result in altered locomotor rhythms. These data demonstrate that the activity of the central clock neurons is necessary for proper lipid storage.


Asunto(s)
Relojes Biológicos/fisiología , Proteínas CLOCK/metabolismo , Ritmo Circadiano/fisiología , Proteínas de Drosophila/metabolismo , Drosophila/fisiología , Neuronas/metabolismo , Triglicéridos/metabolismo , Animales , Femenino , Neuronas/citología
14.
Genes Dev ; 24(12): 1220-35, 2010 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-20551171

RESUMEN

Almost 20 years ago, the gene underlying fatal familial insomnia was discovered, and first suggested the concept that a single gene can regulate sleep. In the two decades since, there have been many advances in the field of behavioral genetics, but it is only in the past 10 years that the genetic analysis of sleep has emerged as an important discipline. Major findings include the discovery of a single gene underlying the sleep disorder narcolepsy, and identification of loci that make quantitative contributions to sleep characteristics. The sleep field has also expanded its focus from mammalian model organisms to Drosophila, zebrafish, and worms, which is allowing the application of novel genetic approaches. Researchers have undertaken large-scale screens to identify new genes that regulate sleep, and are also probing questions of sleep circuitry and sleep function on a molecular level. As genetic tools continue to be refined in each model organism, the genes that support a specific function in sleep will become more apparent. Thus, while our understanding of sleep still remains rudimentary, rapid progress is expected from these recently initiated studies.


Asunto(s)
Sueño/genética , Animales , Ritmo Circadiano/fisiología , Homeostasis/genética , Humanos , Patrón de Herencia/genética , Neuroquímica , Sueño/fisiología
15.
Neuron ; 65(5): 670-81, 2010 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-20223202

RESUMEN

An understanding of sleep requires the identification of distinct cellular circuits that mediate the action of specific sleep:wake-regulating molecules, but such analysis has been very limited. We identify here a circuit that underlies the wake-promoting effects of octopamine in Drosophila. Using MARCM, we identified the ASM cells in the medial protocerebrum as the wake-promoting octopaminergic cells. We then blocked octopamine signaling in random areas of the fly brain and mapped the postsynaptic effect to insulin-secreting neurons of the pars intercerebralis (PI). These PI neurons show altered potassium channel function as well as an increase in cAMP in response to octopamine, and genetic manipulation of their electrical excitability alters sleep:wake behavior. Effects of octopamine on sleep:wake are mediated by the cAMP-dependent isoform of the OAMB receptor. These studies define the cellular and molecular basis of octopamine action and suggest that the PI is a sleep:wake-regulating neuroendocrine structure like the mammalian hypothalamus.


Asunto(s)
Agonistas alfa-Adrenérgicos/farmacología , Red Nerviosa/efectos de los fármacos , Octopamina/farmacología , Sueño/efectos de los fármacos , Vigilia/efectos de los fármacos , Animales , Animales Modificados Genéticamente , Conducta Animal , Ritmo Circadiano/efectos de los fármacos , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Drosophila , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Estimulación Eléctrica/métodos , Proteínas Fluorescentes Verdes/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/genética , Red Nerviosa/fisiología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Técnicas de Placa-Clamp , Bloqueadores de los Canales de Potasio/farmacología , Receptores de Neurotransmisores/genética , Receptores de Neurotransmisores/metabolismo , Sueño/fisiología , Tetraetilamonio/farmacología , Vigilia/fisiología
16.
J Neurosci ; 29(35): 11029-37, 2009 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-19726661

RESUMEN

Caffeine is one of the most widely consumed stimulants in the world and has been proposed to promote wakefulness by antagonizing function of the adenosine A2A receptor. Here, we show that chronic administration of caffeine reduces and fragments sleep in Drosophila and also lengthens circadian period. To identify the mechanisms underlying these effects of caffeine, we first generated mutants of the only known adenosine receptor in flies (dAdoR), which by sequence is most similar to the mammalian A2A receptor. Mutants lacking dAdoR have normal amounts of baseline sleep, as well as normal homeostatic responses to sleep deprivation. Surprisingly, these mutants respond normally to caffeine. On the other hand, the effects of caffeine on sleep and circadian rhythms are mimicked by a potent phosphodiesterase inhibitor, IBMX (3-isobutyl-1-methylxanthine). Using in vivo fluorescence resonance energy transfer imaging, we find that caffeine induces widespread increase in cAMP levels throughout the brain. Finally, the effects of caffeine on sleep are blocked in flies that have reduced neuronal PKA activity. We suggest that chronic administration of caffeine promotes wakefulness in Drosophila, at least in part, by inhibiting cAMP phosphodiesterase activity.


Asunto(s)
Cafeína/farmacología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Receptores Purinérgicos P1/fisiología , Sueño/efectos de los fármacos , Sueño/fisiología , Animales , Línea Celular , Drosophila , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Femenino
17.
J Neurosci ; 28(38): 9377-85, 2008 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-18799671

RESUMEN

Sleep is a fundamental process, but its regulation and function are still not well understood. The Drosophila model for sleep provides a powerful system to address the genetic and molecular mechanisms underlying sleep and wakefulness. Here we show that a Drosophila biogenic amine, octopamine, is a potent wake-promoting signal. Mutations in the octopamine biosynthesis pathway produced a phenotype of increased sleep, which was restored to wild-type levels by pharmacological treatment with octopamine. Moreover, electrical silencing of octopamine-producing cells decreased wakefulness, whereas excitation of these neurons promoted wakefulness. Because protein kinase A (PKA) is a putative target of octopamine signaling and is also implicated in Drosophila sleep, we investigated its role in the effects of octopamine on sleep. We found that decreased PKA activity in neurons rendered flies insensitive to the wake-promoting effects of octopamine. However, this effect of PKA was not exerted in the mushroom bodies, a site previously associated with PKA action on sleep. These studies identify a novel pathway that regulates sleep in Drosophila.


Asunto(s)
Encéfalo/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Drosophila/metabolismo , Octopamina/biosíntesis , Sueño/genética , Administración Oral , Animales , Encéfalo/citología , Drosophila/genética , Proteínas de Drosophila/metabolismo , Femenino , Proteínas Fluorescentes Verdes , Masculino , Actividad Motora/efectos de los fármacos , Actividad Motora/genética , Cuerpos Pedunculados/citología , Cuerpos Pedunculados/metabolismo , Mutación/genética , Neuronas/metabolismo , Norepinefrina/metabolismo , Octopamina/genética , Octopamina/farmacología , Sueño/efectos de los fármacos , Tiramina/metabolismo , Tirosina Descarboxilasa/metabolismo , Vigilia/efectos de los fármacos , Vigilia/genética
18.
J Neurosci ; 26(36): 9250-63, 2006 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-16957081

RESUMEN

The death of motor neurons in amyotrophic lateral sclerosis (ALS) is thought to result from the interaction of a variety of factors including excitotoxicity, accumulation of toxic proteins, and abnormal axonal transport. Previously, we found that the susceptibility of motor neurons to excitotoxic insults can be limited by inhibiting signals evoked by brain-derived neurotrophic factor (BDNF) activation of the receptor tyrosine kinase B (TrkB). Here we show that this can be achieved by direct kinase inhibition or by blockade of a transactivation pathway that uses adenosine A2a receptors and src-family kinases (SFKs). Downstream signaling cascades (such as mitogen-activated protein kinase and phosphatidylinositol-3 kinase) are inhibited by these blockers. In addition to protecting motor neurons from excitotoxic insult, these agents also prevent toxicity that follows from the expression of mutant proteins (G85R superoxide dismutase 1; G59S p150(glued)) that cause familial motor neuron disease. TrkB, adenosine A2a receptors, and SFKs associate into complexes in lipid raft and nonlipid raft membranes and the signaling from lipids rafts may be particularly important because their disruption by cholesterol depletion blocks the ability of BDNF to render motor neurons vulnerable to insult. The neuroprotective versatility of Trk antagonism suggests that it may have broad utility in the treatment of ALS patients.


Asunto(s)
Antagonistas del Receptor de Adenosina A2 , Esclerosis Amiotrófica Lateral/metabolismo , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Neuronas Motoras/metabolismo , Neuronas Motoras/patología , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Esclerosis Amiotrófica Lateral/inducido químicamente , Esclerosis Amiotrófica Lateral/patología , Esclerosis Amiotrófica Lateral/prevención & control , Animales , Células Cultivadas , Ácido Kaínico , Neuronas Motoras/efectos de los fármacos , Fármacos Neuroprotectores/administración & dosificación , Ratas , Ratas Sprague-Dawley , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptor de Adenosina A2A/metabolismo
19.
Nature ; 441(7094): 757-60, 2006 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-16760980

RESUMEN

Sleep is one of the few major whole-organ phenomena for which no function and no underlying mechanism have been conclusively demonstrated. Sleep could result from global changes in the brain during wakefulness or it could be regulated by specific loci that recruit the rest of the brain into the electrical and metabolic states characteristic of sleep. Here we address this issue by exploiting the genetic tractability of the fruitfly, Drosophila melanogaster, which exhibits the hallmarks of vertebrate sleep. We show that large changes in sleep are achieved by spatial and temporal enhancement of cyclic-AMP-dependent protein kinase (PKA) activity specifically in the adult mushroom bodies of Drosophila. Other manipulations of the mushroom bodies, such as electrical silencing, increasing excitation or ablation, also alter sleep. These results link sleep regulation to an anatomical locus known to be involved in learning and memory.


Asunto(s)
Envejecimiento/fisiología , Drosophila melanogaster/fisiología , Cuerpos Pedunculados/fisiología , Sueño/fisiología , Animales , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/biosíntesis , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Drosophila melanogaster/efectos de los fármacos , Drosophila melanogaster/enzimología , Drosophila melanogaster/genética , Femenino , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Homeostasis , Aprendizaje/fisiología , Mifepristona/farmacología , Modelos Animales , Cuerpos Pedunculados/efectos de los fármacos , Sueño/efectos de los fármacos , Sueño/genética
20.
J Comp Neurol ; 494(5): 845-61, 2006 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-16374809

RESUMEN

Emotions, stress, hunger, and circadian rhythms all promote wakefulness and behavioral arousal. Little is known about the pathways mediating these influences, but the orexin-producing neurons of the hypothalamus may play an essential role. These cells heavily innervate many wake-promoting brain regions, and mice lacking the orexin neurons have narcolepsy and fail to rouse in response to hunger (Yamanaka et al. [2003] Neuron 38:701-713). To identify the afferents to the orexin neurons, we first injected a retrograde tracer into the orexin neuron field of rats. Retrogradely labeled neurons were abundant in the allocortex, claustrum, lateral septum, bed nucleus of the stria terminalis, and in many hypothalamic regions including the preoptic area, dorsomedial nucleus, lateral hypothalamus, and posterior hypothalamus. Retrograde labeling in the brainstem was generally more modest, but labeling was strong in the periaqueductal gray matter, dorsal raphe nucleus, and lateral parabrachial nucleus. Injection of an anterograde tracer confirmed that most of these regions directly innervate the orexin neurons, with some of the heaviest input coming from the lateral septum, preoptic area, and posterior hypothalamus. In addition, hypothalamic regions preferentially innervate orexin neurons in the medial and perifornical parts of the field, but most projections from the brainstem target the lateral part of the field. Inputs from the suprachiasmatic nucleus are mainly relayed via the subparaventricular zone and dorsomedial nucleus. These observations suggest that the orexin neurons may integrate a variety of interoceptive and homeostatic signals to increase behavioral arousal in response to hunger, stress, circadian signals, and autonomic challenges.


Asunto(s)
Vías Aferentes/citología , Encéfalo/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neuropéptidos/metabolismo , Vigilia/fisiología , Vías Aferentes/metabolismo , Animales , Encéfalo/citología , Tronco Encefálico/citología , Tronco Encefálico/metabolismo , Hipocampo/citología , Hipocampo/metabolismo , Hipotálamo/citología , Hipotálamo/metabolismo , Masculino , Orexinas , Ratas , Ratas Sprague-Dawley , Telencéfalo/citología , Telencéfalo/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...