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










Base de dados
Intervalo de ano de publicação
1.
Curr Biol ; 33(2): 351-363.e3, 2023 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-36610393

RESUMO

Circadian clocks align various behaviors such as locomotor activity, sleep/wake, feeding, and mating to times of day that are most adaptive. How rhythmic information in pacemaker circuits is translated to neuronal outputs is not well understood. Here, we used brain-wide, 24-h in vivo calcium imaging in the Drosophila brain and searched for circadian rhythmic activity among identified clusters of dopaminergic (DA) and peptidergic neurosecretory (NS) neurons. Such rhythms were widespread and imposed by the PERIOD-dependent clock activity within the ∼150-cell circadian pacemaker network. The rhythms displayed either a morning (M), evening (E), or mid-day (MD) phase. Different subgroups of circadian pacemakers imposed neural activity rhythms onto different downstream non-clock neurons. Outputs from the canonical M and E pacemakers converged to regulate DA-PPM3 and DA-PAL neurons. E pacemakers regulate the evening-active DA-PPL1 neurons. In addition to these canonical M and E oscillators, we present evidence for a third dedicated phase occurring at mid-day: the l-LNv pacemakers present the MD activity peak, and they regulate the MD-active DA-PPM1/2 neurons and three distinct NS cell types. Thus, the Drosophila circadian pacemaker network is a polyphasic rhythm generator. It presents dedicated M, E, and MD phases that are functionally transduced as neuronal outputs to organize diverse daily activity patterns in downstream circuits.


Assuntos
Relógios Circadianos , Proteínas de Drosophila , Animais , Drosophila melanogaster/fisiologia , Atividade Motora/fisiologia , Ritmo Circadiano/fisiologia , Drosophila/fisiologia , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Neurônios Dopaminérgicos/metabolismo
2.
PLoS One ; 17(11): e0275410, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36318573

RESUMO

To support studies of neuropeptide neuromodulation, I have studied beta-arrestin binding sites (BBS's) by evaluating the incidence of BBS sequences among the C terminal tails (CTs) of each of the 49 Drosophila melanogaster neuropeptide GPCRs. BBS were identified by matches with a prediction derived from structural analysis of rhodopsin:arrestin and vasopressin receptor: arrestin complexes [1]. To increase the rigor of the identification, I determined the conservation of BBS sequences between two long-diverged species D. melanogaster and D. virilis. There is great diversity in the profile of BBS's in this group of GPCRs. I present evidence for conserved BBS's in a majority of the Drosophila neuropeptide GPCRs; notably some have no conserved BBS sequences. In addition, certain GPCRs display numerous conserved compound BBS's, and many GPCRs display BBS-like sequences in their intracellular loop (ICL) domains as well. Finally, 20 of the neuropeptide GPCRs are expressed as protein isoforms that vary in their CT domains. BBS profiles are typically different across related isoforms suggesting a need to diversify and regulate the extent and nature of GPCR:arrestin interactions. This work provides the initial basis to initiate future in vivo, genetic analyses in Drosophila to evaluate the roles of arrestins in neuropeptide GPCR desensitization, trafficking and signaling.


Assuntos
Drosophila melanogaster , Neuropeptídeos , Animais , beta-Arrestinas/metabolismo , Drosophila melanogaster/metabolismo , Arrestina/metabolismo , Drosophila/metabolismo , Incidência , beta-Arrestina 1/metabolismo , Sítios de Ligação , Neuropeptídeos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , beta-Arrestina 2/metabolismo
3.
PLoS Genet ; 18(5): e1010013, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35605015

RESUMO

Each day and in conjunction with ambient daylight conditions, neuropeptide PDF regulates the phase and amplitude of locomotor activity rhythms in Drosophila through its receptor, PDFR, a Family B G protein-coupled receptor (GPCR). We studied the in vivo process by which PDFR signaling turns off, by converting as many as half of the 28 potential sites of phosphorylation in its C terminal tail to a non-phosphorylatable residue (alanine). We report that many such sites are conserved evolutionarily, and their conversion creates a specific behavioral syndrome opposite to loss-of-function phenotypes previously described for pdfr. That syndrome includes increases in the amplitudes of both Morning and Evening behavioral peaks, as well as multi-hour delays of the Evening phase. The precise behavioral effects were dependent on day-length, and most effects mapped to conversion of only a few, specific serine residues near the very end of the protein and specific to its A isoform. Behavioral phase delays of the Evening activity under entraining conditions predicted the phase of activity cycles under constant darkness. The behavioral phenotypes produced by the most severe PDFR variant were ligand-dependent in vivo, and not a consequence of changes to their pharmacological properties, nor of changes in their surface expression, as measured in vitro. The mechanisms underlying termination of PDFR signaling are complex, subject to regulation that is modified by season, and central to a better understanding of the peptidergic modulation of behavior.


Assuntos
Proteínas de Drosophila , Neuropeptídeos , Animais , Ritmo Circadiano/genética , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Neurônios/metabolismo , Neuropeptídeos/metabolismo
4.
Proc Natl Acad Sci U S A ; 119(17): e2109969119, 2022 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-35446620

RESUMO

Circadian pacemaker neurons in the Drosophila brain display daily rhythms in the levels of intracellular calcium. These calcium rhythms are driven by molecular clocks and are required for normal circadian behavior. To study their biological basis, we employed genetic manipulations in conjunction with improved methods of in vivo light-sheet microscopy to measure calcium dynamics in individual pacemaker neurons over complete 24-h durations at sampling frequencies as high as 5 Hz. This technological advance unexpectedly revealed cophasic daily rhythms in basal calcium levels and in high-frequency calcium fluctuations. Further, we found that the rhythms of basal calcium levels and of fast calcium fluctuations reflect the activities of two proteins that mediate distinct forms of calcium fluxes. One is the inositol trisphosphate receptor (ITPR), a channel that mediates calcium fluxes from internal endoplasmic reticulum calcium stores, and the other is a T-type voltage-gated calcium channel, which mediates extracellular calcium influx. These results suggest that Drosophila molecular clocks regulate ITPR and T-type channels to generate two distinct but coupled rhythms in basal calcium and in fast calcium fluctuations. We propose that both internal and external calcium fluxes are essential for circadian pacemaker neurons to provide rhythmic outputs and thereby, regulate the activities of downstream brain centers.


Assuntos
Relógios Circadianos , Proteínas de Drosophila , Animais , Relógios Biológicos/fisiologia , Cálcio , Ritmo Circadiano/fisiologia , Drosophila/fisiologia , Proteínas de Drosophila/genética , Neurônios/fisiologia
5.
Nat Commun ; 13(1): 772, 2022 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-35140224

RESUMO

Embryogenesis depends on a tightly regulated balance between mitosis, differentiation, and morphogenesis. Understanding how the embryo uses a relatively small number of proteins to transition between growth and morphogenesis is a central question of developmental biology, but the mechanisms controlling mitosis and differentiation are considered to be fundamentally distinct. Here we show the mitotic kinase Polo, which regulates all steps of mitosis in Drosophila, also directs cellular morphogenesis after cell cycle exit. In mitotic cells, the Aurora kinases activate Polo to control a cytoskeletal regulatory module that directs cytokinesis. We show that in the post-mitotic mesoderm, the control of Polo activity transitions from the Aurora kinases to the uncharacterized kinase Back Seat Driver (Bsd), where Bsd and Polo cooperate to regulate muscle morphogenesis. Polo and its effectors therefore direct mitosis and cellular morphogenesis, but the transition from growth to morphogenesis is determined by the spatiotemporal expression of upstream activating kinases.


Assuntos
Drosophila/metabolismo , Mitose , Morfogênese/fisiologia , Fosfotransferases/metabolismo , Animais , Aurora Quinases/metabolismo , Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , Divisão do Núcleo Celular , Citocinese , Drosophila/genética , Proteínas de Drosophila , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Associadas aos Microtúbulos , Morfogênese/genética , Fosfotransferases/genética , Fuso Acromático/metabolismo
6.
Neuron ; 102(4): 843-857.e4, 2019 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-30981533

RESUMO

Many animals exhibit morning and evening peaks of locomotor behavior. In Drosophila, two corresponding circadian neural oscillators-M (morning) cells and E (evening) cells-exhibit a corresponding morning or evening neural activity peak. Yet we know little of the neural circuitry by which distinct circadian oscillators produce specific outputs to precisely control behavioral episodes. Here, we show that ring neurons of the ellipsoid body (EB-RNs) display spontaneous morning and evening neural activity peaks in vivo: these peaks coincide with the bouts of locomotor activity and result from independent activation by M and E pacemakers. Further, M and E cells regulate EB-RNs via identified PPM3 dopaminergic neurons, which project to the EB and are normally co-active with EB-RNs. These in vivo findings establish the fundamental elements of a circadian neuronal output pathway: distinct circadian oscillators independently drive a common pre-motor center through the agency of specific dopaminergic interneurons.


Assuntos
Ritmo Circadiano/fisiologia , Neurônios Dopaminérgicos/fisiologia , Interneurônios/fisiologia , Locomoção/fisiologia , Animais , Neurônios Dopaminérgicos/metabolismo , Drosophila melanogaster , Interneurônios/metabolismo , Neurônios/metabolismo , Neurônios/fisiologia
7.
Neuron ; 94(6): 1173-1189.e4, 2017 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-28552314

RESUMO

We studied the Drosophila circadian neural circuit using whole-brain imaging in vivo. Five major groups of pacemaker neurons display synchronized molecular clocks, yet each exhibits a distinct phase of daily Ca2+ activation. Light and neuropeptide pigment dispersing factor (PDF) from morning cells (s-LNv) together delay the phase of the evening (LNd) group by ∼12 hr; PDF alone delays the phase of the DN3 group by ∼17 hr. Neuropeptide sNPF, released from s-LNv and LNd pacemakers, produces Ca2+ activation in the DN1 group late in the night. The circuit also features negative feedback by PDF to truncate the s-LNv Ca2+ wave and terminate PDF release. Both PDF and sNPF suppress basal Ca2+ levels in target pacemakers with long durations by cell-autonomous actions. Thus, light and neuropeptides act dynamically at distinct hubs of the circuit to produce multiple suppressive events that create the proper tempo and sequence of circadian pacemaker neuronal activities.


Assuntos
Cálcio/metabolismo , Ritmo Circadiano/genética , Proteínas de Drosophila/genética , Luz , Neurônios/metabolismo , Neuropeptídeos/genética , Animais , Animais Geneticamente Modificados , Drosophila , Proteínas de Drosophila/metabolismo , Retroalimentação Fisiológica , Locomoção , Modelos Teóricos , Neurônios/fisiologia , Neuropeptídeos/metabolismo , Imagem Óptica
8.
Neuron ; 90(4): 781-794, 2016 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-27161526

RESUMO

The neuropeptide PDF promotes the normal sequencing of circadian behavioral rhythms in Drosophila, but its signaling mechanisms are not well understood. We report daily rhythmicity in responsiveness to PDF in critical pacemakers called small LNvs. There is a daily change in potency, as great as 10-fold higher, around dawn. The rhythm persists in constant darkness and does not require endogenous ligand (PDF) signaling or rhythmic receptor gene transcription. Furthermore, rhythmic responsiveness reflects the properties of the pacemaker cell type, not the receptor. Dopamine responsiveness also cycles, in phase with that of PDF, in the same pacemakers, but does not cycle in large LNv. The activity of RalA GTPase in s-LNv regulates PDF responsiveness and behavioral locomotor rhythms. Additionally, cell-autonomous PDF signaling reversed the circadian behavioral effects of lowered RalA activity. Thus, RalA activity confers high PDF responsiveness, providing a daily gate around the dawn hours to promote functional PDF signaling.


Assuntos
Comportamento Animal/fisiologia , Relógios Biológicos/fisiologia , Encéfalo/metabolismo , Ritmo Circadiano/fisiologia , Atividade Motora/fisiologia , Neurônios/metabolismo , Animais , Animais Geneticamente Modificados , Escuridão , Proteínas de Drosophila/genética , Drosophila melanogaster , Neuropeptídeos/metabolismo , Transdução de Sinais/fisiologia
9.
Science ; 351(6276): 976-81, 2016 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-26917772

RESUMO

In Drosophila, molecular clocks control circadian rhythmic behavior through a network of ~150 pacemaker neurons. To explain how the network's neuronal properties encode time, we performed brainwide calcium imaging of groups of pacemaker neurons in vivo for 24 hours. Pacemakers exhibited daily rhythmic changes in intracellular Ca(2+) that were entrained by environmental cues and timed by molecular clocks. However, these rhythms were not synchronous, as each group exhibited its own phase of activation. Ca(2+) rhythms displayed by pacemaker groups that were associated with the morning or evening locomotor activities occurred ~4 hours before their respective behaviors. Loss of the receptor for the neuropeptide PDF promoted synchrony of Ca(2+) waves. Thus, neuropeptide modulation is required to sequentially time outputs from a network of synchronous molecular pacemakers.


Assuntos
Sinalização do Cálcio/fisiologia , Cálcio/metabolismo , Relógios Circadianos , Ritmo Circadiano , Drosophila melanogaster/fisiologia , Neurônios/fisiologia , Animais , Comportamento Animal , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Atividade Motora , Neurônios/metabolismo , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo
10.
Mol Pharmacol ; 88(3): 534-5, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26162863

RESUMO

In August 2014, an international group of researchers gathered for 5 days at the Lorentz Center in Leiden, The Netherlands, to explore the technical and conceptual issues associated with the analysis of G protein-coupled receptor functions utilizing information from crystal structure models to the use of model organisms. This collection of review articles evolved from the 5-day meeting, with brief presentations and structured discussion periods that were designed to identify key questions remaining in understanding G protein-coupled receptor function and to propose novel strategies by integrating scientific disciplines to guide future research.


Assuntos
Congressos como Assunto , Receptores Acoplados a Proteínas G/metabolismo , Animais , Humanos , Receptores Acoplados a Proteínas G/química
11.
G3 (Bethesda) ; 5(7): 1517-24, 2015 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-25999585

RESUMO

The endocrine system employs peptide hormone signals to translate environmental changes into physiological responses. The diffuse endocrine system embedded in the gastrointestinal barrier epithelium is one of the largest and most diverse endocrine tissues. Furthermore, it is the only endocrine tissue in direct physical contact with the microbial environment of the gut lumen. However, it remains unclear how this sensory epithelium responds to specific pathogenic challenges in a dynamic and regulated manner. We demonstrate that the enteroendocrine cells of the adult Drosophila melanogaster midgut display a transient, sensitive, and systemic induction of the prosecretory factor dimmed (dimm) in response to the Gram-negative pathogen Pseudomonas entomophila (Pe). In enteroendocrine cells, dimm controls the levels of the targets Phm, dcat-4, and the peptide hormone, Allatostatin A. Finally, we identify dimm as a host factor that protects against Pe infection and controls the expression of antimicrobial peptides. We propose that dimm provides "gain" in enteroendocrine output during the adaptive response to episodic pathogen exposure.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Proteínas de Drosophila/genética , Células Enteroendócrinas/citologia , Células Enteroendócrinas/metabolismo , Células Enteroendócrinas/microbiologia , Mucosa Intestinal/metabolismo , Intestinos/microbiologia , Intestinos/patologia , Microscopia Confocal , Oxigenases de Função Mista/genética , Oxigenases de Função Mista/metabolismo , Neuropeptídeos/metabolismo , Pseudomonas/fisiologia , Infecções por Pseudomonas/microbiologia , Infecções por Pseudomonas/patologia , Infecções por Pseudomonas/veterinária , Reação em Cadeia da Polimerase em Tempo Real
12.
Mol Pharmacol ; 88(3): 596-603, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25979002

RESUMO

The study of G protein-coupled receptors (GPCRs) has benefited greatly from experimental approaches that interrogate their functions in controlled, artificial environments. Working in vitro, GPCR receptorologists discovered the basic biologic mechanisms by which GPCRs operate, including their eponymous capacity to couple to G proteins; their molecular makeup, including the famed serpentine transmembrane unit; and ultimately, their three-dimensional structure. Although the insights gained from working outside the native environments of GPCRs have allowed for the collection of low-noise data, such approaches cannot directly address a receptor's native (in vivo) functions. An in vivo approach can complement the rigor of in vitro approaches: as studied in model organisms, it imposes physiologic constraints on receptor action and thus allows investigators to deduce the most salient features of receptor function. Here, we briefly discuss specific examples in which model organisms have successfully contributed to the elucidation of signals controlled through GPCRs and other surface receptor systems. We list recent examples that have served either in the initial discovery of GPCR signaling concepts or in their fuller definition. Furthermore, we selectively highlight experimental advantages, shortcomings, and tools of each model organism.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila/genética , Receptores Acoplados a Proteínas G/metabolismo , Animais , Drosophila/metabolismo , Proteínas de Drosophila/genética , Receptores Acoplados a Proteínas G/genética
13.
Nucleic Acids Res ; 43(4): 2199-215, 2015 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-25634895

RESUMO

Neuroendocrine (NE) cells use large dense core vesicles (LDCVs) to traffic, process, store and secrete neuropeptide hormones through the regulated secretory pathway. The dimmed (DIMM) basic helix-loop-helix transcription factor of Drosophila controls the level of regulated secretory activity in NE cells. To pursue its mechanisms, we have performed two independent genome-wide analyses of DIMM's activities: (i) in vivo chromatin immunoprecipitation (ChIP) to define genomic sites of DIMM occupancy and (ii) deep sequencing of purified DIMM neurons to characterize their transcriptional profile. By this combined approach, we showed that DIMM binds to conserved E-boxes in enhancers of 212 genes whose expression is enriched in DIMM-expressing NE cells. DIMM binds preferentially to certain E-boxes within first introns of specific gene isoforms. Statistical machine learning revealed that flanking regions of putative DIMM binding sites contribute to its DNA binding specificity. DIMM's transcriptional repertoire features at least 20 LDCV constituents. In addition, DIMM notably targets the pro-secretory transcription factor, creb-A, but significantly, DIMM does not target any neuropeptide genes. DIMM therefore prescribes the scale of secretory activity in NE neurons, by a systematic control of both proximal and distal points in the regulated secretory pathway.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteínas de Drosophila/metabolismo , Células Neuroendócrinas/metabolismo , Animais , Sequência de Bases , Sítios de Ligação , Imunoprecipitação da Cromatina , Sequência Conservada , Drosophila/genética , Drosophila/metabolismo , Elementos E-Box , Genoma de Inseto , Sequenciamento de Nucleotídeos em Larga Escala , Via Secretória/genética , Análise de Sequência de DNA , Transativadores/metabolismo , Transcriptoma
14.
J Neurosci ; 34(39): 13195-207, 2014 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-25253864

RESUMO

Bioactive peptides are packaged in large dense-core secretory vesicles, which mediate regulated secretion by exocytosis. In a variety of tissues, the regulated release of neurotransmitters and hormones is dependent on calcium levels and controlled by vesicle-associated synaptotagmin (SYT) proteins. Drosophila express seven SYT isoforms, of which two (SYT-α and SYT-ß) were previously found to be enriched in neuroendocrine cells. Here we show that SYT-α and SYT-ß tissue expression patterns are similar, though not identical. Furthermore, both display significant overlap with the bHLH transcription factor DIMM, a known neuroendocrine (NE) regulator. RNAi-mediated knockdown indicates that both SYT-α and SYT-ß functions are essential in identified NE cells as these manipulations phenocopy loss-of-function states for the indicated peptide hormones. In Drosophila cell culture, both SYT-α and neuropeptide cargo form DIMM-dependent fluorescent puncta that are coassociated by super-resolution microscopy. DIMM is required to maintain SYT-α and SYT-ß protein levels in DIMM-expressing cells in vivo. In neurons normally lacking all three proteins (DIMM(-)/SYT-α(-)/SYT-ß(-)), DIMM misexpression conferred accumulation of endogenous SYT-α and SYT-ß proteins. Furthermore transgenic SYT-α does not appreciably accumulate in nonpeptidergic neurons in vivo but does so if DIMM is comisexpressed. Among Drosophila syt genes, only syt-α and syt-ß RNA levels are upregulated by DIMM overexpression. Together, these data suggest that SYT-α and SYT-ß are important for NE cell physiology, that one or both are integral membrane components of the large dense-core vesicles, and that they are closely regulated by DIMM at a post-transcriptional level.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Vesículas Secretórias/metabolismo , Sinaptotagminas/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Linhagem Celular , Proteínas de Drosophila/genética , Neurônios/metabolismo , Neurônios/ultraestrutura , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transporte Proteico , Vesículas Secretórias/ultraestrutura , Sinaptotagminas/genética
15.
J Biol Rhythms ; 28(4): 239-48, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23929551

RESUMO

We used real-time imaging to detect cAMP levels in neurons of intact fly brains to study the mechanisms of circadian pacemaker synchronization by the neuropeptide pigment dispersing factor (PDF) in Drosophila. PDF receptor (PDF-R) is expressed by both M (sLNv) and E (LNd) pacemaker subclasses and is coupled to G(sα) in both cases. We previously reported that PDF-R in M pacemakers elevates cAMP levels by activating the ortholog of mammalian adenylate cyclase 3 (AC3) but that AC3 disruptions had no effect on E pacemaker sensitivity to PDF. Here, we show that PDF-R in E pacemakers activates a different AC isoform, AC78C, an ortholog of mammalian AC8. Knockdown of AC78C by transgenic RNAi substantially reduces, but does not completely abrogate, PDF responses in these E pacemakers. The knockdown effect is intact when restricted to mature stages, suggesting a physiological and not a development role for AC78C in E pacemakers. The AC78C phenotype is rescued by the overexpression of AC78C but not by overexpression of the rutabaga AC. AC78C overexpression does not disrupt PDF responses in these E pacemakers, and neither AC78C knockdown nor its overexpression disrupted locomotor rhythms. Finally, knockdown of 2 AKAPs, nervy and AKAP200, partially reduces LNd PDF responses. These findings begin to identify the components of E pacemaker PDF-R signalosomes and indicate that they are distinct from PDF-R signalosomes in M pacemakers: we propose they contain AC78C and at least 1 other AC.


Assuntos
Ritmo Circadiano/fisiologia , Proteínas de Drosophila/fisiologia , Drosophila/fisiologia , Neurônios/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Proteínas de Ancoragem à Quinase A/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Adenilil Ciclases/genética , Animais , Encéfalo/citologia , Encéfalo/fisiologia , Interpretação Estatística de Dados , Proteínas de Drosophila/genética , Transferência Ressonante de Energia de Fluorescência , Expressão Gênica/fisiologia , Processamento de Imagem Assistida por Computador , Masculino , Proteínas de Membrana/genética , Atividade Motora/genética , Atividade Motora/fisiologia , Neuroimagem , Interferência de RNA , Receptores Acoplados a Proteínas G/genética
16.
Neuron ; 76(1): 82-97, 2012 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-23040808

RESUMO

Neuropeptides modulate neural circuits controlling adaptive animal behaviors and physiological processes, such as feeding/metabolism, reproductive behaviors, circadian rhythms, central pattern generation, and sensorimotor integration. Invertebrate model systems have enabled detailed experimental analysis using combined genetic, behavioral, and physiological approaches. Here we review selected examples of neuropeptide modulation in crustaceans, mollusks, insects, and nematodes, with a particular emphasis on the genetic model organisms Drosophila melanogaster and Caenorhabditis elegans, where remarkable progress has been made. On the basis of this survey, we provide several integrating conceptual principles for understanding how neuropeptides modulate circuit function, and also propose that continued progress in this area requires increased emphasis on the development of richer, more sophisticated behavioral paradigms.


Assuntos
Comportamento Animal/fisiologia , Neurônios/fisiologia , Neuropeptídeos/fisiologia , Neurotransmissores/fisiologia , Animais , Modelos Biológicos
17.
PLoS Biol ; 10(6): e1001337, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22679392

RESUMO

The neuropeptide Pigment Dispersing Factor (PDF) is essential for normal circadian function in Drosophila. It synchronizes the phases of M pacemakers, while in E pacemakers it decelerates their cycling and supports their amplitude. The PDF receptor (PDF-R) is present in both M and subsets of E cells. Activation of PDF-R stimulates cAMP increases in vitro and in M cells in vivo. The present study asks: What is the identity of downstream signaling components that are associated with PDF receptor in specific circadian pacemaker neurons? Using live imaging of intact fly brains and transgenic RNAi, we show that adenylate cyclase AC3 underlies PDF signaling in M cells. Genetic disruptions of AC3 specifically disrupt PDF responses: they do not affect other Gs-coupled GPCR signaling in M cells, they can be rescued, and they do not represent developmental alterations. Knockdown of the Drosophila AKAP-like scaffolding protein Nervy also reduces PDF responses. Flies with AC3 alterations show behavioral syndromes consistent with known roles of M pacemakers as mediated by PDF. Surprisingly, disruption of AC3 does not alter PDF responses in E cells--the PDF-R(+) LNd. Within M pacemakers, PDF-R couples preferentially to a single AC, but PDF-R association with a different AC(s) is needed to explain PDF signaling in the E pacemakers. Thus critical pathways of circadian synchronization are mediated by highly specific second messenger components. These findings support a hypothesis that PDF signaling components within target cells are sequestered into "circadian signalosomes," whose compositions differ between E and M pacemaker cell types.


Assuntos
Adenilil Ciclases/genética , Relógios Biológicos/fisiologia , Ritmo Circadiano/fisiologia , Proteínas de Drosophila/genética , Neuropeptídeos/metabolismo , Transdução de Sinais , Adenilil Ciclases/metabolismo , Animais , Animais Geneticamente Modificados , Relógios Biológicos/genética , Encéfalo/citologia , Encéfalo/metabolismo , Células Cultivadas , Ritmo Circadiano/genética , Drosophila , Proteínas de Drosophila/metabolismo , Neurônios/metabolismo , Neuropeptídeos/genética , Fenótipo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Interferência de RNA , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo
18.
Peptides ; 36(2): 251-6, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22595312

RESUMO

Bioactive peptides are important therapeutic drugs, yet conventional methods of peptide synthesis are challenged to meet increasing demand. We developed a novel and efficient means of metabolic engineering: therapeutic peptide production in Drosophila and as a proof of concept, we demonstrate production of fully matured human insulin. This in vivo system offers an innovative means to produce valuable bioactive peptides for therapies, its inherent flexibility facilitates drug development, and its ease of producing fully processed peptides simplifies metabolic engineering of new peptide products.


Assuntos
Drosophila/metabolismo , Engenharia Metabólica/métodos , Peptídeos/metabolismo , Animais , Drosophila/genética , Humanos , Insulina/genética , Insulina/metabolismo , Peptídeos/genética
19.
Bioessays ; 34(1): 10-6, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22028036

RESUMO

Developing cells acquire mature fates in part by selective (i.e. qualitatively different) expression of a few cell-specific genes. However, all cells share the same basic repertoire of molecular and subcellular building blocks. Therefore, cells must also specialize according to quantitative differences in cell-specific distributions of those common molecular resources. Here we propose the novel hypothesis that evolutionarily-conserved transcription factors called scaling factors (SFs) regulate quantitative differences among mature cell types. SFs: (1) are induced during late stages of cell maturation; (2) are dedicated to specific subcellular domains; and, thus, (3) allow cells to emphasize specific subcellular features. We identify candidate SFs and discuss one in detail: MIST1 (BHLHA15, vertebrates)/DIMM (CG8667, Drosophila); professional secretory cells use this SF to scale up regulated secretion. Because cells use SFs to develop their mature properties and also to adapt them to ever-changing environmental conditions, SF aberrations likely contribute to diseases of adult onset.


Assuntos
Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteínas de Drosophila/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Choque Térmico/metabolismo , Organelas/metabolismo , Fatores de Transcrição/metabolismo , Animais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Compartimento Celular/fisiologia , Diferenciação Celular , Proteínas de Drosophila/genética , Drosophila melanogaster , Proteínas de Choque Térmico/genética , Humanos , Especificidade de Órgãos , Organelas/genética , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Fatores de Transcrição/genética , Transcrição Gênica , Ativação Transcricional
20.
Curr Biol ; 21(21): R894-6, 2011 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-22075431

RESUMO

Circadian (~24 hour) pacemaking mechanisms exist within single cells. Which cellular properties contrive to produce a precise clockworks, and which cell properties are downstream of it? The literature is conflicted as to whether membrane excitability contributes to the mechanism. Now, a new conditional genetic strategy argues excitability is largely dispensable.


Assuntos
Proteínas de Drosophila/fisiologia , Drosophila melanogaster/fisiologia , Drosophila/fisiologia , Neuropeptídeos/fisiologia , Proteínas Circadianas Period/fisiologia , Canais de Potássio Corretores do Fluxo de Internalização/fisiologia , Animais , Masculino
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...