Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
1.
Cell ; 162(4): 836-48, 2015 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-26276633

RESUMEN

Circadian clocks regulate membrane excitability in master pacemaker neurons to control daily rhythms of sleep and wake. Here, we find that two distinctly timed electrical drives collaborate to impose rhythmicity on Drosophila clock neurons. In the morning, a voltage-independent sodium conductance via the NA/NALCN ion channel depolarizes these neurons. This current is driven by the rhythmic expression of NCA localization factor-1, linking the molecular clock to ion channel function. In the evening, basal potassium currents peak to silence clock neurons. Remarkably, daily antiphase cycles of sodium and potassium currents also drive mouse clock neuron rhythms. Thus, we reveal an evolutionarily ancient strategy for the neural mechanisms that govern daily sleep and wake.


Asunto(s)
Relojes Circadianos , Ritmo Circadiano , Drosophila/fisiología , Animales , Relojes Biológicos , Membrana Celular/metabolismo , Drosophila/citología , Proteínas de Drosophila/metabolismo , Técnicas de Silenciamiento del Gen , Canales Iónicos/genética , Canales Iónicos/metabolismo , Proteínas de la Membrana , Ratones , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Técnicas de Placa-Clamp , Potasio/metabolismo , Sodio/metabolismo
2.
PLoS Biol ; 20(1): e3001456, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35081110

RESUMEN

In traumatic brain injury (TBI), the initial injury phase is followed by a secondary phase that contributes to neurodegeneration, yet the mechanisms leading to neuropathology in vivo remain to be elucidated. To address this question, we developed a Drosophila head-specific model for TBI termed Drosophila Closed Head Injury (dCHI), where well-controlled, nonpenetrating strikes are delivered to the head of unanesthetized flies. This assay recapitulates many TBI phenotypes, including increased mortality, impaired motor control, fragmented sleep, and increased neuronal cell death. TBI results in significant changes in the transcriptome, including up-regulation of genes encoding antimicrobial peptides (AMPs). To test the in vivo functional role of these changes, we examined TBI-dependent behavior and lethality in mutants of the master immune regulator NF-κB, important for AMP induction, and found that while sleep and motor function effects were reduced, lethality effects were enhanced. Similarly, loss of most AMP classes also renders flies susceptible to lethal TBI effects. These studies validate a new Drosophila TBI model and identify immune pathways as in vivo mediators of TBI effects.


Asunto(s)
Lesiones Traumáticas del Encéfalo/patología , Drosophila melanogaster , Neuroglía/inmunología , Animales , Péptidos Antimicrobianos/genética , Péptidos Antimicrobianos/metabolismo , Lesiones Traumáticas del Encéfalo/inmunología , Lesiones Traumáticas del Encéfalo/mortalidad , Modelos Animales de Enfermedad , Inmunidad Innata , Locomoción , Masculino , Mutación , FN-kappa B/genética , FN-kappa B/metabolismo , Trastornos del Sueño-Vigilia , Transcriptoma
3.
PLoS Comput Biol ; 20(4): e1012029, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38648221

RESUMEN

The circadian clock is an evolutionarily-conserved molecular oscillator that enables species to anticipate rhythmic changes in their environment. At a molecular level, the core clock genes induce circadian oscillations in thousands of genes in a tissue-specific manner, orchestrating myriad biological processes. While previous studies have investigated how the core clock circuit responds to environmental perturbations such as temperature, the downstream effects of such perturbations on circadian regulation remain poorly understood. By analyzing bulk-RNA sequencing of Drosophila fat bodies harvested from flies subjected to different environmental conditions, we demonstrate a highly condition-specific circadian transcriptome: genes are cycling in a temperature-specific manner, and the distributions of their phases also differ between the two conditions. Further employing a reference-based gene regulatory network (Reactome), we find evidence of increased gene-gene coordination at low temperatures and synchronization of rhythmic genes that are network neighbors. We report that the phase differences between cycling genes increase as a function of geodesic distance in the low temperature condition, suggesting increased coordination of cycling on the gene regulatory network. Our results suggest a potential mechanism whereby the circadian clock mediates the fly's response to seasonal changes in temperature.


Asunto(s)
Relojes Circadianos , Ritmo Circadiano , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Temperatura , Animales , Ritmo Circadiano/genética , Ritmo Circadiano/fisiología , Redes Reguladoras de Genes/genética , Relojes Circadianos/genética , Relojes Circadianos/fisiología , Regulación de la Expresión Génica/genética , Drosophila melanogaster/genética , Drosophila melanogaster/fisiología , Drosophila/genética , Drosophila/fisiología , Transcriptoma/genética , Biología Computacional , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Transcripción Genética/genética
4.
Hum Mol Genet ; 31(7): 1141-1150, 2022 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-34750631

RESUMEN

Disrupted circadian rhythms are a prominent feature of multiple neurodegenerative diseases. Yet mechanisms linking Tau to rhythmic behavior remain unclear. Here, we find that expression of a phosphomimetic human Tau mutant (TauE14) in Drosophila circadian pacemaker neurons disrupts free-running rhythmicity. While cell number and oscillations of the core clock protein PERIOD are unaffected in the small LNv (sLNv) neurons important for free running rhythms, we observe a near complete loss of the major LNv neuropeptide pigment dispersing factor (PDF) in the dorsal axonal projections of the sLNvs. This was accompanied by a ~50% reduction in the area of the dorsal terminals and a modest decrease in cell body PDF levels. Expression of wild-type Tau also reduced axonal PDF levels but to a lesser extent than TauE14. TauE14 also induces a complete loss of mitochondria from these sLNv projections. However, mitochondria were increased in sLNv cell bodies in TauE14 flies. These results suggest that TauE14 disrupts axonal transport of neuropeptides and mitochondria in circadian pacemaker neurons, providing a mechanism by which Tau can disrupt circadian behavior prior to cell loss.


Asunto(s)
Relojes Circadianos , Proteínas de Drosophila , Neuropéptidos , Animales , Relojes Circadianos/genética , Ritmo Circadiano/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Mitocondrias/genética , Mitocondrias/metabolismo , Neuronas/metabolismo , Neuropéptidos/metabolismo
5.
Proc Natl Acad Sci U S A ; 118(47)2021 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-34799448

RESUMEN

Circadian transcriptional timekeepers in pacemaker neurons drive profound daily rhythms in sleep and wake. Here we reveal a molecular pathway that links core transcriptional oscillators to neuronal and behavioral rhythms. Using two independent genetic screens, we identified mutants of Transport and Golgi organization 10 (Tango10) with poor behavioral rhythmicity. Tango10 expression in pacemaker neurons expressing the neuropeptide PIGMENT-DISPERSING FACTOR (PDF) is required for robust rhythms. Loss of Tango10 results in elevated PDF accumulation in nerve terminals even in mutants lacking a functional core clock. TANGO10 protein itself is rhythmically expressed in PDF terminals. Mass spectrometry of TANGO10 complexes reveals interactions with the E3 ubiquitin ligase CULLIN 3 (CUL3). CUL3 depletion phenocopies Tango10 mutant effects on PDF even in the absence of the core clock gene timeless Patch clamp electrophysiology in Tango10 mutant neurons demonstrates elevated spontaneous firing potentially due to reduced voltage-gated Shaker-like potassium currents. We propose that Tango10/Cul3 transduces molecular oscillations from the core clock to neuropeptide release important for behavioral rhythms.


Asunto(s)
Relojes Circadianos/fisiología , Proteínas de Drosophila/metabolismo , Neuropéptidos/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Translocador Nuclear del Receptor de Aril Hidrocarburo/genética , Translocador Nuclear del Receptor de Aril Hidrocarburo/metabolismo , Proteínas Cullin/genética , Proteínas Cullin/metabolismo , Drosophila , Proteínas de Drosophila/genética , Neuronas/metabolismo , Neuropéptidos/genética , Proteómica , Sueño
6.
PLoS Genet ; 15(10): e1008356, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31593562

RESUMEN

Disrupted circadian rhythms is a prominent and early feature of neurodegenerative diseases including Huntington's disease (HD). In HD patients and animal models, striatal and hypothalamic neurons expressing molecular circadian clocks are targets of mutant Huntingtin (mHtt) pathogenicity. Yet how mHtt disrupts circadian rhythms remains unclear. In a genetic screen for modifiers of mHtt effects on circadian behavior in Drosophila, we discovered a role for the neurodegenerative disease gene Ataxin2 (Atx2). Genetic manipulations of Atx2 modify the impact of mHtt on circadian behavior as well as mHtt aggregation and demonstrate a role for Atx2 in promoting mHtt aggregation as well as mHtt-mediated neuronal dysfunction. RNAi knockdown of the Fragile X mental retardation gene, dfmr1, an Atx2 partner, also partially suppresses mHtt effects and Atx2 effects depend on dfmr1. Atx2 knockdown reduces the cAMP response binding protein A (CrebA) transcript at dawn. CrebA transcript level shows a prominent diurnal regulation in clock neurons. Loss of CrebA also partially suppresses mHtt effects on behavior and cell loss and restoration of CrebA can suppress Atx2 effects. Our results indicate a prominent role of Atx2 in mediating mHtt pathology, specifically via its regulation of CrebA, defining a novel molecular pathway in HD pathogenesis.


Asunto(s)
Ataxina-2/genética , Relojes Circadianos/genética , Proteína de Unión al Elemento de Respuesta al AMP Cíclico/genética , Proteínas de Drosophila/genética , Proteína Huntingtina/genética , Enfermedad de Huntington/genética , Animales , Ritmo Circadiano/genética , Modelos Animales de Enfermedad , Drosophila melanogaster/genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Humanos , Enfermedad de Huntington/patología , Proteínas Mutantes/genética , Neuronas/metabolismo , Transducción de Señal/genética
7.
Proc Natl Acad Sci U S A ; 115(39): E9247-E9256, 2018 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-30201705

RESUMEN

Circadian clocks play a key role in regulating a vast array of biological processes, with significant implications for human health. Accurate assessment of physiological time using transcriptional biomarkers found in human blood can significantly improve diagnosis of circadian disorders and optimize the delivery time of therapeutic treatments. To be useful, such a test must be accurate, minimally burdensome to the patient, and readily generalizable to new data. A major obstacle in development of gene expression biomarker tests is the diversity of measurement platforms and the inherent variability of the data, often resulting in predictors that perform well in the original datasets but cannot be universally applied to new samples collected in other settings. Here, we introduce TimeSignature, an algorithm that robustly infers circadian time from gene expression. We demonstrate its application in data from three independent studies using distinct microarrays and further validate it against a new set of samples profiled by RNA-sequencing. Our results show that TimeSignature is more accurate and efficient than competing methods, estimating circadian time to within 2 h for the majority of samples. Importantly, we demonstrate that once trained on data from a single study, the resulting predictor can be universally applied to yield highly accurate results in new data from other studies independent of differences in study population, patient protocol, or assay platform without renormalizing the data or retraining. This feature is unique among expression-based predictors and addresses a major challenge in the development of generalizable, clinically useful tests.


Asunto(s)
Relojes Circadianos/genética , Perfilación de la Expresión Génica/métodos , Aprendizaje Automático , Biomarcadores/sangre , Ritmo Circadiano/genética , Expresión Génica , Genes/genética , Humanos , Modelos Estadísticos , Reproducibilidad de los Resultados , Sueño , Transcriptoma
8.
Proc Natl Acad Sci U S A ; 114(6): 1407-1412, 2017 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-28115724

RESUMEN

Laboratory studies have demonstrated that circadian clocks align physiology and behavior to 24-h environmental cycles. Examination of athletic performance has been used to discern the functions of these clocks in humans outside of controlled settings. Here, we examined the effects of jet lag, that is, travel that shifts the alignment of 24-h environmental cycles relative to the endogenous circadian clock, on specific performance metrics in Major League Baseball. Accounting for potential differences in home and away performance, travel direction, and team confounding variables, we observed that jet-lag effects were largely evident after eastward travel with very limited effects after westward travel, consistent with the >24-h period length of the human circadian clock. Surprisingly, we found that jet lag impaired major parameters of home-team offensive performance, for example, slugging percentage, but did not similarly affect away-team offensive performance. On the other hand, jet lag impacted both home and away defensive performance. Remarkably, the vast majority of these effects for both home and away teams could be explained by a single measure, home runs allowed. Rather than uniform effects, these results reveal surprisingly specific effects of circadian misalignment on athletic performance under natural conditions.


Asunto(s)
Rendimiento Atlético/fisiología , Béisbol/fisiología , Ritmo Circadiano/fisiología , Síndrome Jet Lag/fisiopatología , Béisbol/estadística & datos numéricos , Relojes Circadianos/fisiología , Humanos , Modelos Lineales , Análisis Multivariante , Factores de Tiempo , Viaje
9.
N Engl J Med ; 384(20): e76, 2021 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-34010544
11.
PLoS Biol ; 12(3): e1001810, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24643294

RESUMEN

Molecular circadian clocks are interconnected via neural networks. In Drosophila, PIGMENT-DISPERSING FACTOR (PDF) acts as a master network regulator with dual functions in synchronizing molecular oscillations between disparate PDF(+) and PDF(-) circadian pacemaker neurons and controlling pacemaker neuron output. Yet the mechanisms by which PDF functions are not clear. We demonstrate that genetic inhibition of protein kinase A (PKA) in PDF(-) clock neurons can phenocopy PDF mutants while activated PKA can partially rescue PDF receptor mutants. PKA subunit transcripts are also under clock control in non-PDF DN1p neurons. To address the core clock target of PDF, we rescued per in PDF neurons of arrhythmic per°¹ mutants. PDF neuron rescue induced high amplitude rhythms in the clock component TIMELESS (TIM) in per-less DN1p neurons. Complete loss of PDF or PKA inhibition also results in reduced TIM levels in non-PDF neurons of per°¹ flies. To address how PDF impacts pacemaker neuron output, we focally applied PDF to DN1p neurons and found that it acutely depolarizes and increases firing rates of DN1p neurons. Surprisingly, these effects are reduced in the presence of an adenylate cyclase inhibitor, yet persist in the presence of PKA inhibition. We have provided evidence for a signaling mechanism (PKA) and a molecular target (TIM) by which PDF resets and synchronizes clocks and demonstrates an acute direct excitatory effect of PDF on target neurons to control neuronal output. The identification of TIM as a target of PDF signaling suggests it is a multimodal integrator of cell autonomous clock, environmental light, and neural network signaling. Moreover, these data reveal a bifurcation of PKA-dependent clock effects and PKA-independent output effects. Taken together, our results provide a molecular and cellular basis for the dual functions of PDF in clock resetting and pacemaker output.


Asunto(s)
Ritmo Circadiano/genética , Proteínas de Drosophila/fisiología , Drosophila/metabolismo , Neuronas/metabolismo , Neuropéptidos/fisiología , Animales , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Drosophila/genética , Drosophila/fisiología , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Red Nerviosa , Neuronas/fisiología , Neuropéptidos/genética , Neuropéptidos/metabolismo , Transducción de Señal
12.
Nature ; 470(7334): 399-403, 2011 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-21331043

RESUMEN

Daily oscillations of gene expression underlie circadian behaviours in multicellular organisms. While attention has been focused on transcriptional and post-translational mechanisms, other post-transcriptional modes have been less clearly delineated. Here we report mutants of a novel Drosophila gene twenty-four (tyf) that show weak behavioural rhythms. Weak rhythms are accompanied by marked reductions in the levels of the clock protein Period (PER) as well as more modest effects on Timeless (TIM). Nonetheless, PER induction in pacemaker neurons can rescue tyf mutant rhythms. TYF associates with a 5'-cap-binding complex, poly(A)-binding protein (PABP), as well as per and tim transcripts. Furthermore, TYF activates reporter expression when tethered to reporter messenger RNA even in vitro. Taken together, these data indicate that TYF potently activates PER translation in pacemaker neurons to sustain robust rhythms, revealing a new and important role for translational control in the Drosophila circadian clock.


Asunto(s)
Relojes Circadianos/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/fisiología , Genes de Insecto/genética , Proteínas Circadianas Period/biosíntesis , Biosíntesis de Proteínas/fisiología , Animales , Relojes Circadianos/fisiología , Ritmo Circadiano/genética , Ritmo Circadiano/fisiología , Proteínas de Drosophila/biosíntesis , Proteínas de Drosophila/genética , Genes Reporteros/genética , Mutación/genética , Neuronas/metabolismo , Neuronas/fisiología , Proteínas Circadianas Period/genética , Proteínas Circadianas Period/metabolismo , Proteínas de Unión a Poli(A)/metabolismo , Unión Proteica , Biosíntesis de Proteínas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Regulación hacia Arriba
14.
PLoS Comput Biol ; 11(3): e1004094, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25793520

RESUMEN

Robust methods for identifying patterns of expression in genome-wide data are important for generating hypotheses regarding gene function. To this end, several analytic methods have been developed for detecting periodic patterns. We improve one such method, JTK_CYCLE, by explicitly calculating the null distribution such that it accounts for multiple hypothesis testing and by including non-sinusoidal reference waveforms. We term this method empirical JTK_CYCLE with asymmetry search, and we compare its performance to JTK_CYCLE with Bonferroni and Benjamini-Hochberg multiple hypothesis testing correction, as well as to five other methods: cyclohedron test, address reduction, stable persistence, ANOVA, and F24. We find that ANOVA, F24, and JTK_CYCLE consistently outperform the other three methods when data are limited and noisy; empirical JTK_CYCLE with asymmetry search gives the greatest sensitivity while controlling for the false discovery rate. Our analysis also provides insight into experimental design and we find that, for a fixed number of samples, better sensitivity and specificity are achieved with higher numbers of replicates than with higher sampling density. Application of the methods to detecting circadian rhythms in a metadataset of microarrays that quantify time-dependent gene expression in whole heads of Drosophila melanogaster reveals annotations that are enriched among genes with highly asymmetric waveforms. These include a wide range of oxidation reduction and metabolic genes, as well as genes with transcripts that have multiple splice forms.


Asunto(s)
Ritmo Circadiano/genética , Perfilación de la Expresión Génica/métodos , Genómica/métodos , Modelos Genéticos , Modelos Estadísticos , Animales , Simulación por Computador , Drosophila melanogaster/genética , Genoma de los Insectos/genética
15.
PLoS Genet ; 8(10): e1003003, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23055946

RESUMEN

Sleep is homeostatically regulated, such that sleep drive reflects the duration of prior wakefulness. However, despite the discovery of genes important for sleep, a coherent molecular model for sleep homeostasis has yet to emerge. To better understand the function and regulation of sleep, we employed a reverse-genetics approach in Drosophila. An insertion in the BTB domain protein CG32810/insomniac (inc) exhibited one of the strongest baseline sleep phenotypes thus far observed, a ~10 h sleep reduction. Importantly, this is coupled to a reduced homeostatic response to sleep deprivation, consistent with a disrupted sleep homeostat. Knockdown of the INC-interacting protein, the E3 ubiquitin ligase Cul3, results in reduced sleep duration, consolidation, and homeostasis, suggesting an important role for protein turnover in mediating INC effects. Interestingly, inc and Cul3 expression in post-mitotic neurons during development contributes to their adult sleep functions. Similar to flies with increased dopaminergic signaling, loss of inc and Cul3 result in hyper-arousability to a mechanical stimulus in adult flies. Furthermore, the inc sleep duration phenotype can be rescued by pharmacological inhibition of tyrosine hydroxylase, the rate-limiting enzyme for dopamine biosynthesis. Taken together, these results establish inc and Cul3 as important new players in setting the sleep homeostat and a dopaminergic arousal pathway in Drosophila.


Asunto(s)
Nivel de Alerta/fisiología , Proteínas Cullin/genética , Proteínas de Drosophila/genética , Drosophila/genética , Homeostasis/genética , Transducción de Señal , Sueño/genética , Animales , Conducta Animal , Neuronas Colinérgicas/metabolismo , Proteínas Cullin/metabolismo , Dopamina/metabolismo , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , Regulación del Desarrollo de la Expresión Génica , Masculino , Mutación , Fenotipo , Unión Proteica , Interferencia de ARN
17.
Annu Rev Physiol ; 72: 605-24, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20148690

RESUMEN

Circadian clocks organize behavior and physiology to adapt to daily environmental cycles. Genetic approaches in the fruit fly, Drosophila melanogaster, have revealed widely conserved molecular gears of these 24-h timers. Yet much less is known about how these cell-autonomous clocks confer temporal information to modulate cellular functions. Here we discuss our current knowledge of circadian clock function in Drosophila, providing an overview of the molecular underpinnings of circadian clocks. We then describe the neural network important for circadian rhythms of locomotor activity, including how these molecular clocks might influence neuronal function. Finally, we address a range of behaviors and physiological systems regulated by circadian clocks, including discussion of specific peripheral oscillators and key molecular effectors where they have been described. These studies reveal a remarkable complexity to circadian pathways in this "simple" model organism.


Asunto(s)
Conducta Animal/fisiología , Ritmo Circadiano/genética , Ritmo Circadiano/fisiología , Drosophila/fisiología , Animales , Relojes Biológicos/genética , Relojes Biológicos/fisiología , Ritmo Circadiano/inmunología , Drosophila/inmunología , Humanos , Locomoción/fisiología , Red Nerviosa/fisiología
18.
J Biol Rhythms ; 39(1): 5-19, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37978840

RESUMEN

Collegiate athletes must satisfy the academic obligations common to all undergraduates, but they have the additional structural and social stressors of extensive practice time, competition schedules, and frequent travel away from their home campus. Clearly such stressors can have negative impacts on both their academic and athletic performances as well as on their health. These concerns are made more acute by recent proposals and decisions to reorganize major collegiate athletic conferences. These rearrangements will require more multi-day travel that interferes with the academic work and personal schedules of athletes. Of particular concern is additional east-west travel that results in circadian rhythm disruptions commonly called jet lag that contribute to the loss of amount as well as quality of sleep. Circadian misalignment and sleep deprivation and/or sleep disturbances have profound effects on physical and mental health and performance. We, as concerned scientists and physicians with relevant expertise, developed this white paper to raise awareness of these challenges to the wellbeing of our student-athletes and their co-travelers. We also offer practical steps to mitigate the negative consequences of collegiate travel schedules. We discuss the importance of bedtime protocols, the availability of early afternoon naps, and adherence to scheduled lighting exposure protocols before, during, and after travel, with support from wearables and apps. We call upon departments of athletics to engage with sleep and circadian experts to advise and help design tailored implementation of these mitigating practices that could contribute to the current and long-term health and wellbeing of their students and their staff members.


Asunto(s)
Ritmo Circadiano , Sueño , Humanos , Síndrome Jet Lag , Atletas , Estudiantes , Viaje
19.
bioRxiv ; 2024 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-38293065

RESUMEN

A catalog of transcription factor (TF) binding sites in the genome is critical for deciphering regulatory relationships. Here we present the culmination of the modERN (model organism Encyclopedia of Regulatory Networks) consortium that systematically assayed TF binding events in vivo in two major model organisms, Drosophila melanogaster (fly) and Caenorhabditis elegans (worm). We describe key features of these datasets, comprising 604 TFs identifying 3.6M sites in the fly and 350 TFs identifying 0.9 M sites in the worm. Applying a machine learning model to these data identifies sets of TFs with a prominent role in promoting target gene expression in specific cell types. TF binding data are available through the ENCODE Data Coordinating Center and at https://epic.gs.washington.edu/modERNresource, which provides access to processed and summary data, as well as widgets to probe cell type-specific TF-target relationships. These data are a rich resource that should fuel investigations into TF function during development.

20.
Proc Natl Acad Sci U S A ; 107(30): 13497-502, 2010 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-20624977

RESUMEN

To compare circadian gene expression within highly discrete neuronal populations, we separately purified and characterized two adjacent but distinct groups of Drosophila adult circadian neurons: the 8 small and 10 large PDF-expressing ventral lateral neurons (s-LNvs and l-LNvs, respectively). The s-LNvs are the principal circadian pacemaker cells, whereas recent evidence indicates that the l-LNvs are involved in sleep and light-mediated arousal. Although half of the l-LNv-enriched mRNA population, including core clock mRNAs, is shared between the l-LNvs and s-LNvs, the other half is l-LNv- and s-LNv-specific. The distribution of four specific mRNAs is consistent with prior characterization of the four encoded proteins, and therefore indicates successful purification of the two neuronal types. Moreover, an octopamine receptor mRNA is selectively enriched in l-LNvs, and only these neurons respond to in vitro application of octopamine. Dissection and purification of l-LNvs from flies collected at different times indicate that these neurons contain cycling clock mRNAs with higher circadian amplitudes as well as at least a 10-fold higher fraction of oscillating mRNAs than all previous analyses of head RNA. Many of these cycling l-LNv mRNAs are well expressed but do not cycle or cycle much less well elsewhere in heads. The results suggest that RNA cycling is much more prominent in circadian neurons than elsewhere in heads and may be particularly important for the functioning of these neurons.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Perfilación de la Expresión Génica , Neuronas/metabolismo , Neuropéptidos/genética , Animales , Relojes Biológicos/genética , Ritmo Circadiano/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Femenino , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Masculino , Microscopía Fluorescente , Neuronas/citología , Neuropéptidos/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Receptores de Amina Biogénica/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Núcleos Talámicos Ventrales/citología , Núcleos Talámicos Ventrales/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA