Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 137
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 165(3): 742-53, 2016 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-27040499

RESUMEN

RNA transcripts are bound and regulated by RNA-binding proteins (RBPs). Current methods for identifying in vivo targets of an RBP are imperfect and not amenable to examining small numbers of cells. To address these issues, we developed TRIBE (targets of RNA-binding proteins identified by editing), a technique that couples an RBP to the catalytic domain of the Drosophila RNA-editing enzyme ADAR and expresses the fusion protein in vivo. RBP targets are marked with novel RNA editing events and identified by sequencing RNA. We have used TRIBE to identify the targets of three RBPs (Hrp48, dFMR1, and NonA). TRIBE compares favorably to other methods, including CLIP, and we have identified RBP targets from as little as 150 specific fly neurons. TRIBE can be performed without an antibody and in small numbers of specific cells.


Asunto(s)
Adenosina Desaminasa/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/enzimología , Técnicas Genéticas , Edición de ARN , Regiones no Traducidas 3' , Animales , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Proteínas de Unión al ARN
2.
Genes Dev ; 37(9-10): 432-448, 2023 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-37164645

RESUMEN

A wide range of sequencing methods has been developed to assess nascent RNA transcription and resolve the single-nucleotide position of RNA polymerase genome-wide. These techniques are often burdened with high input material requirements and lengthy protocols. We leveraged the template-switching properties of thermostable group II intron reverse transcriptase (TGIRT) and developed Butt-seq (bulk analysis of nascent transcript termini sequencing), which can produce libraries from purified nascent RNA in 6 h and from as few as 10,000 cells-an improvement of at least 10-fold over existing techniques. Butt-seq shows that inhibition of the superelongation complex (SEC) causes promoter-proximal pausing to move upstream in a fashion correlated with subnucleosomal fragments. To address transcriptional regulation in a tissue, Butt-seq was used to measure the circadian regulation of transcription from fly heads. All the results indicate that Butt-seq is a simple and powerful technique to analyze transcription at a high level of resolution.


Asunto(s)
ADN Polimerasa Dirigida por ARN , ARN , ARN/genética , ADN Polimerasa Dirigida por ARN/genética , ADN Polimerasa Dirigida por ARN/metabolismo , Regulación de la Expresión Génica , ARN Polimerasa II/metabolismo , Intrones , Análisis de Secuencia de ARN/métodos , Transcripción Genética/genética
3.
Mol Cell ; 82(2): 389-403, 2022 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-34739873

RESUMEN

RNA binding proteins (RBPs) regulate nearly all post-transcriptional processes within cells. To fully understand RBP function, it is essential to identify their in vivo targets. Standard techniques for profiling RBP targets, such as crosslinking immunoprecipitation (CLIP) and its variants, are limited or suboptimal in some situations, e.g. when compatible antibodies are not available and when dealing with small cell populations such as neuronal subtypes and primary stem cells. This review summarizes and compares several genetic approaches recently designed to identify RBP targets in such circumstances. TRIBE (targets of RNA binding proteins identified by editing), RNA tagging, and STAMP (surveying targets by APOBEC-mediated profiling) are new genetic tools useful for the study of post-transcriptional regulation and RBP identification. We describe the underlying RNA base editing technology, recent applications, and therapeutic implications.


Asunto(s)
Técnicas Genéticas , Edición de ARN , Procesamiento Postranscripcional del ARN , Proteínas de Unión al ARN/genética , ARN/genética , Animales , Sitios de Unión , Humanos , Unión Proteica , ARN/metabolismo , Proteínas de Unión al ARN/metabolismo
4.
Cell ; 171(6): 1241-1245, 2017 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-29195068
5.
Proc Natl Acad Sci U S A ; 121(15): e2321338121, 2024 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-38568969

RESUMEN

To address the contribution of transcriptional regulation to Drosophila clock gene expression and to behavior, we generated a series of CRISPR-mediated deletions within two regions of the circadian gene timeless (tim), an intronic E-box region and an upstream E-box region that are both recognized by the key transcription factor Clock (Clk) and its heterodimeric partner Cycle. The upstream deletions but not an intronic deletion dramatically impact tim expression in fly heads; the biggest upstream deletion reduces peak RNA levels and tim RNA cycling amplitude to about 15% of normal, and there are similar effects on tim protein (TIM). The cycling amplitude of other clock genes is also strongly reduced, in these cases due to increases in trough levels. These data underscore the important contribution of the upstream E-box enhancer region to tim expression and of TIM to clock gene transcriptional repression in fly heads. Surprisingly, tim expression in clock neurons is only modestly affected by the biggest upstream deletion and is similarly affected by a deletion of the intronic E-box region. This distinction between clock neurons and glia is paralleled by a dramatically enhanced accessibility of the intronic enhancer region within clock neurons. This distinctive feature of tim chromatin was revealed by ATAC-seq (assay for transposase-accessible chromatin with sequencing) assays of purified neurons and glia as well as of fly heads. The enhanced cell type-specific accessibility of the intronic enhancer region explains the resilience of clock neuron tim expression and circadian behavior to deletion of the otherwise more prominent upstream tim E-box region.


Asunto(s)
Proteínas de Drosophila , Drosophila , Animales , Cromatina/metabolismo , Ritmo Circadiano/genética , Proteínas CLOCK/genética , ADN/metabolismo , Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Regulación de la Expresión Génica , ARN/metabolismo
7.
Proc Natl Acad Sci U S A ; 120(29): e2303779120, 2023 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-37428902

RESUMEN

Circadian behavioral rhythms in Drosophila melanogaster are regulated by about 75 pairs of brain neurons. They all express the core clock genes but have distinct functions and gene expression profiles. To understand the importance of these distinct molecular programs, neuron-specific gene manipulations are essential. Although RNAi based methods are standard to manipulate gene expression in a cell-specific manner, they are often ineffective, especially in assays involving smaller numbers of neurons or weaker Gal4 drivers. We and others recently exploited a neuron-specific CRISPR-based method to mutagenize genes within circadian neurons. Here, we further explore this approach to mutagenize three well-studied clock genes: the transcription factor gene vrille, the photoreceptor gene Cryptochrome (cry), and the neuropeptide gene Pdf (pigment dispersing factor). The CRISPR-based strategy not only reproduced their known phenotypes but also assigned cry function for different light-mediated phenotypes to discrete, different subsets of clock neurons. We further tested two recently published methods for temporal regulation in adult neurons, inducible Cas9 and the auxin-inducible gene expression system. The results were not identical, but both approaches successfully showed that the adult-specific knockout of the neuropeptide Pdf reproduces the canonical loss-of-function mutant phenotypes. In summary, a CRISPR-based strategy is a highly effective, reliable, and general method to temporally manipulate gene function in specific adult neurons.


Asunto(s)
Relojes Circadianos , Proteínas de Drosophila , Neuropéptidos , Animales , Drosophila melanogaster/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Ritmo Circadiano/genética , Neuropéptidos/genética , Neuropéptidos/metabolismo , Criptocromos/genética , Criptocromos/metabolismo , Neuronas/metabolismo , Relojes Circadianos/genética
8.
RNA ; 29(8): 1230-1242, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37169395

RESUMEN

RNA binding proteins (RBPs) perform a myriad of functions and are implicated in numerous neurological diseases. To identify the targets of RBPs in small numbers of cells, we developed TRIBE, in which the catalytic domain of the RNA editing enzyme ADAR (ADARcd) is fused to an RBP. When the RBP binds to an mRNA, ADAR catalyzes A to G modifications in the target mRNA that can be easily identified in standard RNA sequencing. In STAMP, the concept is the same except the ADARcd is replaced by the RNA editing enzyme APOBEC. Here we compared TRIBE and STAMP side-by-side in human and Drosophila cells. The goal is to learn the pros and cons of each method so that researchers can choose the method best suited to their RBP and system. In human cells, TRIBE and STAMP were performed using the RBP TDP-43. Although they both identified TDP-43 target mRNAs, combining the two methods more successfully identified high-confidence targets. In Drosophila cells, RBP-APOBEC fusions generated only low numbers of editing sites, comparable to the level of control editing. This was true for two different RBPs, Hrp48 and Thor (Drosophila EIF4E-BP), indicating that STAMP does not work well in Drosophila.


Asunto(s)
Proteínas de Drosophila , Drosophila , Animales , Humanos , Drosophila/genética , Proteínas de Drosophila/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ADN/metabolismo
10.
Proc Natl Acad Sci U S A ; 119(34): e2206066119, 2022 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-35969763

RESUMEN

The metronome-like circadian regulation of sleep timing must still adapt to an uncertain environment. Recent studies in Drosophila indicate that neuromodulation not only plays a key role in clock neuron synchronization but also affects interactions between the clock network and brain sleep centers. We show here that the targets of neuromodulators, G Protein Coupled Receptors (GPCRs), are highly enriched in the fly brain circadian clock network. Single-cell sequencing indicates that they are not only enriched but also differentially expressed and contribute to clock neuron identity. We generated a comprehensive guide library to mutagenize individual GPCRs in specific neurons and verified the strategy by introducing a targeted sequencing approach. Combined with a behavioral screen, the mutagenesis strategy revealed a role of dopamine in sleep regulation by identifying two dopamine receptors and a clock neuron subpopulation that gate the timing of sleep.


Asunto(s)
Ritmo Circadiano , Dopamina , Proteínas de Drosophila , Neuronas , Receptores Acoplados a Proteínas G , Animales , Relojes Circadianos/genética , Ritmo Circadiano/genética , Dopamina/genética , Dopamina/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Neuronas/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Sueño/genética
11.
PLoS Genet ; 18(3): e1010024, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35239675

RESUMEN

Genetic mechanisms that repress transposable elements (TEs) in young animals decline during aging, as reflected by increased TE expression in aged animals. Does increased TE expression during aging lead to more genomic TE copies in older animals? To address this question, we quantified TE Landscapes (TLs) via whole genome sequencing of young and aged Drosophila strains of wild-type and mutant backgrounds. We quantified TLs in whole flies and dissected brains and validated the feasibility of our approach in detecting new TE insertions in aging Drosophila genomes when small RNA and RNA interference (RNAi) pathways are compromised. We also describe improved sequencing methods to quantify extra-chromosomal DNA circles (eccDNAs) in Drosophila as an additional source of TE copies that accumulate during aging. Lastly, to combat the natural progression of aging-associated TE expression, we show that knocking down PAF1, a conserved transcription elongation factor that antagonizes RNAi pathways, may bolster suppression of TEs during aging and extend lifespan. Our study suggests that in addition to a possible influence by different genetic backgrounds, small RNA and RNAi mechanisms may mitigate genomic TL expansion despite the increase in TE transcripts during aging.


Asunto(s)
Elementos Transponibles de ADN , Drosophila , Envejecimiento/genética , Animales , Elementos Transponibles de ADN/genética , Drosophila/genética , Genómica/métodos , ARN
12.
Proc Natl Acad Sci U S A ; 117(21): 11760-11769, 2020 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-32393629

RESUMEN

Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) are two related neurodegenerative diseases that present with similar TDP-43 pathology in patient tissue. TDP-43 is an RNA-binding protein which forms aggregates in neurons of ALS and FTD patients as well as in a subset of patients diagnosed with other neurodegenerative diseases. Despite our understanding that TDP-43 is essential for many aspects of RNA metabolism, it remains obscure how TDP-43 dysfunction contributes to neurodegeneration. Interestingly, altered neuronal dendritic morphology is a common theme among several neurological disorders and is thought to precede neurodegeneration. We previously found that both TDP-43 overexpression (OE) and knockdown (KD) result in reduced dendritic branching of cortical neurons. In this study, we used TRIBE (targets of RNA-binding proteins identified by editing) as an approach to identify signaling pathways that regulate dendritic branching downstream of TDP-43. We found that TDP-43 RNA targets are enriched for pathways that signal to the CREB transcription factor. We further found that TDP-43 dysfunction inhibits CREB activation and CREB transcriptional output, and restoring CREB signaling rescues defects in dendritic branching. Finally, we demonstrate, using RNA sequencing, that TDP-43 OE and KD cause similar changes in the abundance of specific messenger RNAs, consistent with their ability to produce similar morphological defects. Our data therefore provide a mechanism by which TDP-43 dysfunction interferes with dendritic branching, and may define pathways for therapeutic intervention in neurodegenerative diseases.


Asunto(s)
Proteína de Unión a Elemento de Respuesta al AMP Cíclico , Proteínas de Unión al ADN , Dendritas , Regulación de la Expresión Génica/genética , Transducción de Señal , Animales , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Dendritas/metabolismo , Dendritas/patología , Células HEK293 , Humanos , ARN Mensajero/metabolismo , Ratas , Transducción de Señal/genética , Transducción de Señal/fisiología , Proteinopatías TDP-43
13.
Nature ; 536(7616): 292-7, 2016 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-27479324

RESUMEN

Little is known about the ability of Drosophila circadian neurons to promote sleep. Here we show, using optogenetic manipulation and video recording, that a subset of dorsal clock neurons (DN1s) are potent sleep-promoting cells that release glutamate to directly inhibit key pacemaker neurons. The pacemakers promote morning arousal by activating these DN1s, implying that a late-day feedback circuit drives midday siesta and night-time sleep. To investigate more plastic aspects of the sleep program, we used a calcium assay to monitor and compare the real-time activity of DN1 neurons in freely behaving males and females. Our results revealed that DN1 neurons were more active in males than in females, consistent with the finding that male flies sleep more during the day. DN1 activity is also enhanced by elevated temperature, consistent with the ability of higher temperatures to increase sleep. These new approaches indicate that DN1s have a major effect on the fly sleep-wake profile and integrate environmental information with the circadian molecular program.


Asunto(s)
Ritmo Circadiano/fisiología , Drosophila melanogaster/citología , Drosophila melanogaster/fisiología , Retroalimentación Fisiológica , Neuronas/fisiología , Sueño/fisiología , Vigilia/fisiología , Animales , Relojes Biológicos/fisiología , Calcio/metabolismo , Femenino , Ácido Glutámico/metabolismo , Masculino , Actividad Motora , Optogenética , Caracteres Sexuales , Temperatura , Grabación en Video
14.
Genes Dev ; 28(1): 8-13, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-24395244

RESUMEN

The mammalian circadian clock relies on the master genes CLOCK and BMAL1 to drive rhythmic gene expression and regulate biological functions under circadian control. Here we show that rhythmic CLOCK:BMAL1 DNA binding promotes rhythmic chromatin opening. Mechanisms include CLOCK:BMAL1 binding to nucleosomes and rhythmic chromatin modification; e.g., incorporation of the histone variant H2A.Z. This rhythmic chromatin remodeling mediates the rhythmic binding of other transcription factors adjacent to CLOCK:BMAL1, suggesting that the activity of these other transcription factors contributes to the genome-wide CLOCK:BMAL1 heterogeneous transcriptional output. These data therefore indicate that the clock regulation of transcription relies on the rhythmic regulation of chromatin accessibility and suggest that the concept of pioneer function extends to acute gene regulation.


Asunto(s)
Factores de Transcripción ARNTL/metabolismo , Proteínas CLOCK/metabolismo , Ritmo Circadiano/fisiología , Factores de Transcripción/metabolismo , Factores de Transcripción ARNTL/genética , Animales , Proteínas CLOCK/genética , Ensamble y Desensamble de Cromatina , Regulación de la Expresión Génica , Ratones , Nucleosomas/metabolismo , Unión Proteica , Factores de Transcripción/genética
15.
Angew Chem Int Ed Engl ; 60(16): 8650-8666, 2021 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-33634573

RESUMEN

Circadian rhythms are present in most if not all animals, plants, and even photosynthetic cyanobacteria. These cyanobacterial clocks as well as plant clocks are very different from those of animals, with no credible homology between the different clock proteins. Therefore circadian rhythms probably emerged multiple times in evolution, which underscores their importance.


Asunto(s)
Ritmo Circadiano/genética , Retroalimentación Fisiológica , Animales , Humanos
16.
J Neurosci ; 39(9): 1621-1630, 2019 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-30606757

RESUMEN

To provide organisms with a fitness advantage, circadian clocks have to react appropriately to changes in their environment. High-intensity (HI) light plays an essential role in the adaptation to hot summer days, which especially endanger insects of desiccation or prey visibility. Here, we show that solely increasing light intensity leads to an increased midday siesta in Drosophila behavior. Interestingly, this change is independent of the fly's circadian photoreceptor cryptochrome and is solely caused by a small visual organ, the Hofbauer-Buchner eyelets. Using receptor knock-downs, immunostaining, and recently developed calcium tools, we show that the eyelets activate key core clock neurons, namely the s-LNvs, at HI. This activation delays the decrease of PERIOD (PER) in the middle of the day and propagates to downstream target clock neurons that prolong the siesta. We show a new pathway for integrating light-intensity information into the clock network, suggesting new network properties and surprising parallels between Drosophila and the mammalian system.SIGNIFICANCE STATEMENT The ability of animals to adapt to their ever-changing environment plays an important role in their fitness. A key player in this adaptation is the circadian clock. For animals to predict the changes of day and night, they must constantly monitor, detect and incorporate changes in the environment. The appropriate incorporation and reaction to high-intensity (HI) light is of special importance for insects because they might suffer from desiccation during hot summer days. We show here that different photoreceptors have specialized functions to integrate low-intensity, medium-intensity, or HI light into the circadian system in Drosophila These results show surprising parallels to mammalian mechanisms, which also use different photoreceptor subtypes to respond to different light intensities.


Asunto(s)
Adaptación Fisiológica , Relojes Circadianos , Vías Visuales/fisiología , Animales , Criptocromos/genética , Criptocromos/metabolismo , Drosophila , Neuronas/metabolismo , Neuronas/fisiología , Proteínas Circadianas Period/genética , Proteínas Circadianas Period/metabolismo , Vías Visuales/metabolismo
17.
RNA ; 24(2): 173-182, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29127211

RESUMEN

We previously developed TRIBE, a method for the identification of cell-specific RNA-binding protein targets. TRIBE expresses an RBP of interest fused to the catalytic domain (cd) of the RNA-editing enzyme ADAR and performs adenosine-to-inosine editing on RNA targets of the RBP. However, target identification is limited by the low editing efficiency of the ADARcd. Here we describe HyperTRIBE, which carries a previously characterized hyperactive mutation (E488Q) of the ADARcd. HyperTRIBE identifies dramatically more editing sites, many of which are also edited by TRIBE but at a much lower editing frequency. HyperTRIBE therefore more faithfully recapitulates the known binding specificity of its RBP than TRIBE. In addition, separating RNA binding from the enhanced editing activity of the HyperTRIBE ADAR catalytic domain sheds light on the mechanism of ADARcd editing as well as the enhanced activity of the HyperADARcd.


Asunto(s)
Adenosina Desaminasa/genética , Proteínas de Drosophila/genética , Proteínas de Unión al ARN/metabolismo , Adenosina/metabolismo , Animales , Sitios de Unión , Dominio Catalítico , Proteínas de Drosophila/metabolismo , Ribonucleoproteínas Nucleares Heterogéneas/genética , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Inosina/metabolismo , Mutación , Edición de ARN , Proteínas Recombinantes de Fusión/metabolismo , Análisis de Secuencia de ARN
18.
Mol Cell ; 47(1): 27-37, 2012 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-22658416

RESUMEN

The RNA editing enzyme ADAR chemically modifies adenosine (A) to inosine (I), which is interpreted by the ribosome as a guanosine. Here we assess cotranscriptional A-to-I editing in Drosophila by isolating nascent RNA from adult fly heads and subjecting samples to high throughput sequencing. There are a large number of edited sites within nascent exons. Nascent RNA from an ADAR-null strain was also sequenced, indicating that almost all A-to-I events require ADAR. Moreover, mRNA editing levels correlate with editing levels within the cognate nascent RNA sequence, indicating that the extent of editing is set cotranscriptionally. Surprisingly, the nascent data also identify an excess of intronic over exonic editing sites. These intronic sites occur preferentially within introns that are poorly spliced cotranscriptionally, suggesting a link between editing and splicing. We conclude that ADAR-mediated editing is more widespread than previously indicated and largely occurs cotranscriptionally.


Asunto(s)
Adenosina Desaminasa/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Edición de ARN , Transcripción Genética , Animales , Sitios de Unión/genética , Drosophila/clasificación , Drosophila/genética , Evolución Molecular , Exones/genética , Expresión Génica , Intrones/genética , Mutación , Precursores del ARN/genética , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
19.
Proc Natl Acad Sci U S A ; 114(41): E8780-E8787, 2017 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-28973886

RESUMEN

There are no general methods for reliably assessing the firing properties or even calcium profiles of specific neurons in freely moving flies. To this end, we adapted a GFP-based calcium reporter to luciferase that was expressed in small subsets of circadian neurons. This Tric-LUC reporter allowed a direct comparison of luciferase activity with locomotor activity, which was assayed in the same flies with video recording. The LUC profile from activity-promoting E cells paralleled evening locomotor activity, and the LUC profile from sleep-promoting glutamatergic DN1s (gDN1s) paralleled daytime sleep. Similar profiles were generated by novel reporters recently identified based on transcription factor activation. As E cell and gDN1 activity is necessary and sufficient for normal evening locomotor activity and daytime sleep profiles, respectively, we suggest that their luciferase profiles reflect their neuronal calcium and in some cases firing profiles in wake-behaving flies.


Asunto(s)
Calcio/metabolismo , Ritmo Circadiano/fisiología , Drosophila melanogaster/fisiología , Retroalimentación Fisiológica , Locomoción/fisiología , Neuronas/fisiología , Sueño/fisiología , Animales , Drosophila melanogaster/citología , Masculino , Actividad Motora , Neuronas/citología , Optogenética
20.
PLoS Genet ; 13(2): e1006613, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28182648

RESUMEN

Locomotor activity rhythms are controlled by a network of ~150 circadian neurons within the adult Drosophila brain. They are subdivided based on their anatomical locations and properties. We profiled transcripts "around the clock" from three key groups of circadian neurons with different functions. We also profiled a non-circadian outgroup, dopaminergic (TH) neurons. They have cycling transcripts but fewer than clock neurons as well as low expression and poor cycling of clock gene transcripts. This suggests that TH neurons do not have a canonical circadian clock and that their gene expression cycling is driven by brain systemic cues. The three circadian groups are surprisingly diverse in their cycling transcripts and overall gene expression patterns, which include known and putative novel neuropeptides. Even the overall phase distributions of cycling transcripts are distinct, indicating that different regulatory principles govern transcript oscillations. This surprising cell-type diversity parallels the functional heterogeneity of the different neurons.


Asunto(s)
Relojes Circadianos/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Neuronas/metabolismo , Neuropéptidos/genética , Animales , Animales Modificados Genéticamente , Encéfalo/citología , Encéfalo/metabolismo , Neuronas Dopaminérgicas/metabolismo , Drosophila melanogaster/citología , Drosophila melanogaster/metabolismo , Perfilación de la Expresión Génica/métodos , Microscopía Fluorescente , Análisis de Secuencia por Matrices de Oligonucleótidos , Análisis de Secuencia de ARN/métodos , Factores de Tiempo , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA