Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Annu Rev Neurosci ; 44: 153-172, 2021 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-33556251

RESUMEN

During the approximately 5 days of Drosophila neurogenesis (late embryogenesis to the beginning of pupation), a limited number of neural stem cells produce approximately 200,000 neurons comprising hundreds of cell types. To build a functional nervous system, neuronal types need to be produced in the proper places, appropriate numbers, and correct times. We discuss how neural stem cells (neuroblasts) obtain so-called area codes for their positions in the nervous system (spatial patterning) and how they keep time to sequentially produce neurons with unique fates (temporal patterning). We focus on specific examples that demonstrate how a relatively simple patterning system (Notch) can be used reiteratively to generate different neuronal types. We also speculate on how different modes of temporal patterning that operate over short versus long time periods might be linked. We end by discussing how specification programs are integrated and lead to the terminal features of different neuronal types.


Asunto(s)
Proteínas de Drosophila , Células-Madre Neurales , Animales , Drosophila , Proteínas de Drosophila/genética , Neurogénesis , Neuronas
2.
Development ; 150(2)2023 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-36537580

RESUMEN

Temporal identity factors regulate competence of neural progenitors to generate specific cell types in a time-dependent manner, but how they operate remains poorly defined. In the developing mouse retina, the Ikaros zinc-finger transcription factor Ikzf1 regulates production of early-born cell types, except cone photoreceptors. In this study we show that, during early stages of retinal development, another Ikaros family protein, Ikzf4, functions redundantly with Ikzf1 to regulate cone photoreceptor production. Using CUT&RUN and functional assays, we show that Ikzf4 binds and represses genes involved in late-born rod photoreceptor specification, hence favoring cone production. At late stages, when Ikzf1 is no longer expressed in progenitors, we show that Ikzf4 re-localizes to target genes involved in gliogenesis and is required for Müller glia production. We report that Ikzf4 regulates Notch signaling genes and is sufficient to activate the Hes1 promoter through two Ikzf GGAA-binding motifs, suggesting a mechanism by which Ikzf4 may influence gliogenesis. These results uncover a combinatorial role for Ikaros family members during nervous system development and provide mechanistic insights on how they temporally regulate cell fate output.


Asunto(s)
Factor de Transcripción Ikaros , Retina , Ratones , Animales , Retina/metabolismo , Factor de Transcripción Ikaros/genética , Factor de Transcripción Ikaros/metabolismo , Células Fotorreceptoras Retinianas Conos/metabolismo , Células Fotorreceptoras Retinianas Bastones/metabolismo , Diferenciación Celular/genética
3.
Semin Cell Dev Biol ; 142: 54-66, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-35738966

RESUMEN

The nervous system consists of a vast diversity of neurons and glia that are accurately assembled into functional circuits. What are the mechanisms that generate these diverse cell types? During development, an epithelial sheet with neurogenic potential is initially regionalised into spatially restricted domains of gene expression. From this, pools of neural stem cells (NSCs) with distinct molecular profiles and the potential to generate different neuron types, are specified. These NSCs then divide asymmetrically to self-renew and generate post-mitotic neurons or glia. As NSCs age, they experience transitions in gene expression, which further allows them to generate different neurons or glia over time. Versions of this general template of spatial and temporal patterning operate during the development of different parts of different nervous systems. Here, I cover our current knowledge of Drosophila brain and optic lobe development as well as the development of the vertebrate cortex and spinal cord within the framework of this above template. I highlight where our knowledge is lacking, where mechanisms beyond these might operate, and how the emergence of new technologies might help address unanswered questions.


Asunto(s)
Células-Madre Neurales , Animales , Células-Madre Neurales/metabolismo , Neuronas/metabolismo , Drosophila/genética , Vertebrados , Encéfalo
4.
Semin Cell Dev Biol ; 142: 13-22, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-35623984

RESUMEN

The generation of neuronal diversity involves temporal patterning mechanisms by which a given progenitor sequentially produces multiple cell types. Several parallels are evident between the brain development programs of Drosophila and vertebrates, such as the successive emergence of specific cell types and the use of combinations of transcription factors to specify cell fates. Furthermore, cell-extrinsic cues such as hormones and signaling pathways have also been shown to be regulatory modules of temporal patterning. Recently, transcriptomic and epigenomic studies using large single-cell sequencing datasets have provided insights into the transcriptional dynamics of neurogenesis in the Drosophila and mammalian central nervous systems. We review these commonalities in the specification of neuronal identity and highlight the conserved or convergent strategies of brain development by discussing temporal patterning mechanisms found in flies and vertebrates.


Asunto(s)
Proteínas de Drosophila , Drosophila , Animales , Drosophila/metabolismo , Vertebrados/metabolismo , Neuronas/metabolismo , Sistema Nervioso Central/metabolismo , Proteínas de Drosophila/metabolismo , Regulación del Desarrollo de la Expresión Génica , Mamíferos/metabolismo
5.
Semin Cell Dev Biol ; 142: 23-35, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-35915026

RESUMEN

Proper functioning of the nervous system relies not only on the generation of a vast repertoire of distinct neural cell types but also on the precise neural circuitry within them. How the generation of highly diverse neural populations is regulated during development remains a topic of interest. Landmark studies in Drosophila have identified the genetic and temporal cues regulating neural diversity and thus have provided valuable insights into our understanding of temporal patterning of the central nervous system. The development of the Drosophila central complex, which is mostly derived from type II neural stem cell (NSC) lineages, showcases how a small pool of NSCs can give rise to vast and distinct progeny. Similar to the human outer subventricular zone (OSVZ) neural progenitors, type II NSCs generate intermediate neural progenitors (INPs) to expand and diversify lineages that populate higher brain centers. Each type II NSC has a distinct spatial identity and timely regulated expression of many transcription factors and mRNA binding proteins. Additionally, INPs derived from them show differential expression of genes depending on their birth order. Together type II NSCs and INPs display a combinatorial temporal patterning that expands neural diversity of the central brain lineages. We cover advances in current understanding of type II NSC temporal patterning and discuss similarities and differences in temporal patterning mechanisms of various NSCs with a focus on how cell-intrinsic and extrinsic hormonal cues regulate temporal transitions in NSCs during larval development. Cell extrinsic ligands activate conserved signaling pathways and extrinsic hormonal cues act as a temporal switch that regulate temporal progression of the NSCs. We conclude by elaborating on how a progenitor's temporal code regulates the fate specification and identity of distinct neural types. At the end, we also discuss open questions in linking developmental cues to neural identity, circuits, and underlying behaviors in the adult fly.


Asunto(s)
Proteínas de Drosophila , Células-Madre Neurales , Animales , Humanos , Señales (Psicología) , Células-Madre Neurales/metabolismo , Neuronas/metabolismo , Drosophila/genética , Sistema Nervioso Central/metabolismo , Proteínas de Drosophila/metabolismo
6.
Development ; 149(17)2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-36069896

RESUMEN

In the developing nervous system, neural stem cells (NSCs) use temporal patterning to generate a wide variety of different neuronal subtypes. In Drosophila, the temporal transcription factors, Hunchback, Kruppel, Pdm and Castor, are sequentially expressed by NSCs to regulate temporal identity during neurogenesis. Here, we identify a new temporal transcription factor that regulates the transition from the Pdm to Castor temporal windows. This factor, which we call Chronophage (or 'time-eater'), is homologous to mammalian CTIP1 (Bcl11a) and CTIP2 (Bcl11b). We show that Chronophage binds upstream of the castor gene and regulates its expression. Consistent with Chronophage promoting a temporal switch, chronophage mutants generate an excess of Pdm-specified neurons and are delayed in generating neurons associated with the Castor temporal window. In addition to promoting the Pdm to Castor transition, Chronophage also represses the production of neurons generated during the earlier Hunchback and Kruppel temporal windows. Genetic interactions with Hunchback and Kruppel indicate that Chronophage regulates NSC competence to generate Hunchback- and Kruppel-specified neurons. Taken together, our results suggest that Chronophage has a conserved role in temporal patterning and neuronal subtype specification.


Asunto(s)
Proteínas de Drosophila , Células-Madre Neurales , Animales , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Drosophila/genética , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Mamíferos/metabolismo , Células-Madre Neurales/metabolismo , Receptores de Superficie Celular/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo
7.
Dev Biol ; 481: 116-128, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34666024

RESUMEN

During development, neural progenitors undergo temporal patterning as they age to sequentially generate differently fated progeny. Temporal patterning of neural progenitors is relatively well-studied in Drosophila. Temporal cascades of transcription factors or opposing temporal gradients of RNA-binding proteins are expressed in neural progenitors as they age to control the fates of the progeny. The temporal progression is mostly driven by intrinsic mechanisms including cross-regulations between temporal genes, but environmental cues also play important roles in certain transitions. Vertebrate neural progenitors demonstrate greater plasticity in response to extrinsic cues. Recent studies suggest that vertebrate neural progenitors are also temporally patterned by a combination of transcriptional and post-transcriptional mechanisms in response to extracellular signaling to regulate neural fate specification. In this review, we summarize recent advances in the study of temporal patterning of neural progenitors in Drosophila and vertebrates. We also discuss the involvement of epigenetic mechanisms, specifically the Polycomb group complexes and ATP-dependent chromatin remodeling complexes, in the temporal patterning of neural progenitors.


Asunto(s)
Epigénesis Genética , Regulación del Desarrollo de la Expresión Génica , Células-Madre Neurales/metabolismo , Neurogénesis , Transducción de Señal , Animales , Drosophila melanogaster
8.
Development ; 147(18)2020 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-32878923

RESUMEN

Multipotent retinal progenitor cells (RPCs) generate various cell types in a precise chronological order, but how exactly cone photoreceptor production is restricted to early stages remains unclear. Here, we show that the POU-homeodomain factors Pou2f1/Pou2f2, the homologs of Drosophila temporal identity factors nub/pdm2, regulate the timely production of cones in mice. Forcing sustained expression of Pou2f1 or Pou2f2 in RPCs expands the period of cone production, whereas misexpression in late-stage RPCs triggers ectopic cone production at the expense of late-born fates. Mechanistically, we report that Pou2f1 induces Pou2f2 expression, which binds to a POU motif in the promoter of the rod-inducing factor Nrl to repress its expression. Conversely, conditional inactivation of Pou2f2 in RPCs increases Nrl expression and reduces cone production. Finally, we provide evidence that Pou2f1 is part of a cross-regulatory cascade with the other temporal identity factors Ikzf1 and Casz1. These results uncover Pou2f1/2 as regulators of the temporal window for cone genesis and, given their widespread expression in the nervous system, raise the possibility of a general role in temporal patterning.This article has an associated 'The people behind the papers' interview.


Asunto(s)
Proteínas del Ojo/metabolismo , Factor 1 de Transcripción de Unión a Octámeros/metabolismo , Factor 2 de Transcripción de Unión a Octámeros/metabolismo , Retina/metabolismo , Células Fotorreceptoras Retinianas Conos/metabolismo , Animales , Drosophila/metabolismo , Femenino , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas/genética , Células Fotorreceptoras Retinianas Bastones/metabolismo , Células Madre/metabolismo
9.
Dev Biol ; 464(1): 35-44, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32442418

RESUMEN

Proper neural function depends on the correct specification of individual neural fates, controlled by combinations of neuronal transcription factors. Different neural types are sequentially generated by neural progenitors in a defined order, and this temporal patterning process can be controlled by Temporal Transcription Factors (TTFs) that form temporal cascades in neural progenitors. The Drosophila medulla, part of the visual processing center of the brain, contains more than 70 neural types generated by medulla neuroblasts which sequentially express several TTFs, including Homothorax (Hth), eyeless (Ey), Sloppy paired 1 and 2 (Slp), Dichaete (D) and Tailless (Tll). However, it is not clear how such a small number of TTFs could give rise to diverse combinations of neuronal transcription factors that specify a large number of medulla neuron types. Here we report how temporal patterning specifies one neural type, the T1 neuron. We show that the T1 neuron is the only medulla neuron type that expresses the combination of three transcription factors Ocelliless (Oc or Otd), Sox102F and Ets65A. Using CRISPR-Cas9 system, we show that each transcription factor is required for the correct morphogenesis of T1 neurons. Interestingly, Oc, Sox102F and Ets65A initiate expression in neurons beginning at different temporal stages and last in a few subsequent temporal stages. Oc expressing neurons are generated in the Ey, Slp and D stages; Sox102F expressing neurons are produced in the Slp and D stages; while Ets65A is expressed in subsets of medulla neurons born in the D and later stages. The TTF Ey, Slp or D is required to initiate the expression of Oc, Sox102F or Ets65A in neurons, respectively. Thus, the neurons expressing all three transcription factors are born in the D stage and become T1 neurons. In neurons where the three transcription factors do not overlap, each of the three transcription factors can act in combination with other neuronal transcription factors to specify different neural fates. We show that this way of expression regulation of neuronal transcription factors by temporal patterning can generate more possible combinations of transcription factors in neural progeny to diversify neural fates.


Asunto(s)
Proteínas de Drosophila/metabolismo , Regulación del Desarrollo de la Expresión Génica , Neuronas/metabolismo , Factores de Transcripción/metabolismo , Animales , Proteínas de Drosophila/genética , Drosophila melanogaster , Neuronas/citología , Factores de Transcripción/genética
10.
Development ; 144(24): 4552-4562, 2017 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-29158446

RESUMEN

Drosophila neuroblasts are an excellent model for investigating how neuronal diversity is generated. Most brain neuroblasts generate a series of ganglion mother cells (GMCs) that each make two neurons (type I lineage), but 16 brain neuroblasts generate a series of intermediate neural progenitors (INPs) that each produce 4-6 GMCs and 8-12 neurons (type II lineage). Thus, type II lineages are similar to primate cortical lineages, and may serve as models for understanding cortical expansion. Yet the origin of type II neuroblasts remains mysterious: do they form in the embryo or larva? If they form in the embryo, do their progeny populate the adult central complex, as do the larval type II neuroblast progeny? Here, we present molecular and clonal data showing that all type II neuroblasts form in the embryo, produce INPs and express known temporal transcription factors. Embryonic type II neuroblasts and INPs undergo quiescence, and produce embryonic-born progeny that contribute to the adult central complex. Our results provide a foundation for investigating the development of the central complex, and tools for characterizing early-born neurons in central complex function.


Asunto(s)
Encéfalo/embriología , Drosophila melanogaster/embriología , Ganglios de Invertebrados/embriología , Células-Madre Neurales/metabolismo , Neurogénesis/fisiología , Neuronas/citología , Animales , Encéfalo/citología , Linaje de la Célula/fisiología , Proliferación Celular , Femenino , Larva/citología , Masculino
11.
Int J Mol Sci ; 21(20)2020 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-33050604

RESUMEN

The complexity of brain structure and function is rooted in the precise spatial and temporal regulation of selective developmental events. During neurogenesis, both vertebrates and invertebrates generate a wide variety of specialized cell types through the expansion and specification of a restricted set of neuronal progenitors. Temporal patterning of neural progenitors rests on fine regulation between cell-intrinsic and cell-extrinsic mechanisms. The rapid emergence of high-throughput single-cell technologies combined with elaborate computational analysis has started to provide us with unprecedented biological insights related to temporal patterning in the developing central nervous system (CNS). Here, we present an overview of recent advances in Drosophila and vertebrates, focusing both on cell-intrinsic mechanisms and environmental influences. We then describe the various multi-omics approaches that have strongly contributed to our current understanding and discuss perspectives on the various -omics approaches that hold great potential for the future of temporal patterning research.


Asunto(s)
Tipificación del Cuerpo/genética , Genómica , Metabolómica , Neurogénesis/genética , Proteómica , Análisis de la Célula Individual , Lóbulo Temporal/embriología , Lóbulo Temporal/metabolismo , Animales , Regulación del Desarrollo de la Expresión Génica , Genómica/métodos , Proteómica/métodos , Análisis de la Célula Individual/métodos
12.
Int J Audiol ; 58(7): 393-400, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30888882

RESUMEN

Objective: To determine the efficacy of a targeted auditory training programme as a remediation approach for children diagnosed with a temporal patterning disorder. Design: Temporal Patterning scores were determined at two-time points pre-intervention and again post-training. Measures were then repeated in half of the participants after a further non-training period to determine the sustainability of effects. Cognitive skills and outcome measures were assessed at all time-points. Study Sample: Ten children aged between 7 and 12 years of age, diagnosed with a temporal patterning processing disorder, were enrolled in the training programme. Results: A group mean of 9.2 hours training was completed. Conclusion: Significant, sustainable improvements on the Frequency Pattern Test were found (2.5 SD increase in score relative to mean of age matched-peers) at the completion of training. Duration of training did not predict the degree of improvement. Cognitive skills did not show significant changes in ability. Significant, sustainable improvements in temporal patterning ability were seen after completion of the training programme. No associated changes in cognitive skills were seen, suggesting independence of the skills. Deficit-specific approaches are available across the traditional test battery, however, determining the appropriate management plan for a child diagnosed with an auditory processing disorder requires a patient-centric approach.


Asunto(s)
Percepción Auditiva , Trastornos de la Percepción Auditiva/terapia , Cognición , Remediación Cognitiva/métodos , Patrones de Reconocimiento Fisiológico , Trastornos de la Percepción Auditiva/psicología , Niño , Femenino , Humanos , Masculino , Resultado del Tratamiento
13.
Cereb Cortex ; 27(2): 1491-1500, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-26759484

RESUMEN

Voluntary movement initiation involves the engagement of large populations of motor cortical neurons around movement onset. Despite knowledge of the temporal dynamics that lead to movement, the spatial structure of these dynamics across the cortical surface remains unknown. In data from 4 rhesus macaques, we show that the timing of attenuation of beta frequency local field potential oscillations, a correlate of locally activated cortex, forms a spatial gradient across primary motor cortex (MI). We show that these spatio-temporal dynamics are recapitulated in the engagement order of ensembles of MI neurons. We demonstrate that these patterns are unique to movement onset and suggest that movement initiation requires a precise spatio-temporal sequential activation of neurons in MI.


Asunto(s)
Potenciales de Acción/fisiología , Corteza Motora/fisiología , Movimiento/fisiología , Animales , Conducta Animal , Mapeo Encefálico/métodos , Macaca mulatta , Neuronas Motoras/fisiología
14.
J Neurogenet ; 28(3-4): 233-49, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24912777

RESUMEN

The brain areas that endow insects with the ability to see consist of remarkably complex neural circuits. Reiterated arrays of many diverse neuron subtypes are assembled into modular yet coherent functional retinotopic maps. Tremendous progress in developing genetic tools and cellular markers over the past years advanced our understanding of the mechanisms that control the stepwise production and differentiation of neurons in the visual system of Drosophila melanogaster. The postembryonic optic lobe utilizes at least two modes of neurogenesis that are distinct from other parts of the fly central nervous system. In the first optic ganglion, the lamina, neuroepithelial cells give rise to precursor cells, whose proliferation and differentiation depend on anterograde signals from photoreceptor axons. In the second optic ganglion, the medulla, the coordinated activity of four signaling pathways orchestrates the gradual conversion of neuroepithelial cells into neuroblasts, while a specific cascade of temporal identity transcription factors controls subtype diversification of their progeny.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila/genética , Células-Madre Neurales/citología , Neurogénesis/genética , Lóbulo Óptico de Animales no Mamíferos/citología , Animales , Regulación del Desarrollo de la Expresión Génica , Células-Madre Neurales/metabolismo , Neuronas/citología , Neuronas/metabolismo , Lóbulo Óptico de Animales no Mamíferos/metabolismo
15.
J Sleep Res ; 23(5): 499-507, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24730977

RESUMEN

Whether subjects with insomnia exhibit good sleep on some interval basis is unclear. Prior research suggests that patients with insomnia are highly variable with respect to night-to-night sleep continuity, that more than 40% of patients exhibit temporal patterning of good sleep, and that nearly 90% of patients exhibit better than average sleep following 1 to 3 nights of relatively poor sleep. The aim of the present study was to replicate and extend the above-noted findings utilizing: (i) a large sample studied over an extended time interval (ii) absolute standards for 'good' and 'poor' sleep; and (iii) a formal statistical methodology to assess temporal patterning and the association of time in bed with bout duration of poor or average sleep. Thirty-three subjects with insomnia and 33 good sleepers completed sleep diaries over the course of 110 days. It was found that subjects with insomnia (compared to good sleepers) had more poor nights (e.g. about 39 versus 7% of the assessed nights), a higher probability of a having a poor night on any given occasion (60% greater probability than good sleepers) and more consecutive nights of poor sleep between good sleep nights (median bout duration of approximately three versus one night). Lastly, it was found that (as would be predicted by both the Spielman model and the two-process model) time in bed moderated bout duration in the insomnia group. That is, longer times in bed were associated with longer bouts of poor sleep.


Asunto(s)
Trastornos del Inicio y del Mantenimiento del Sueño/fisiopatología , Sueño/fisiología , Adulto , Femenino , Humanos , Masculino , Autoinforme , Factores de Tiempo
16.
FEBS J ; 291(4): 663-671, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37943156

RESUMEN

Developing tissues are patterned in space and time; this enables them to differentiate their cell types and form complex structures to support different body plans. Although space and time are two independent entities, there are many examples of spatial patterns that originate from temporal ones. The most prominent example is the expression of the genes hunchback, Krüppel, pdm, and castor, which are expressed temporally in the neural stem cells of the Drosophila ventral nerve cord and spatially along the anteroposterior axis of the blastoderm stage embryo. In this Viewpoint, we investigate the relationship between space and time in specific examples of spatial and temporal patterns with the aim of gaining insight into the evolutionary history of patterning.


Asunto(s)
Proteínas de Drosophila , Células-Madre Neurales , Animales , Regulación del Desarrollo de la Expresión Génica , Drosophila/genética , Proteínas de Drosophila/genética , Blastodermo , Tipificación del Cuerpo/genética
17.
Elife ; 122024 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-38391176

RESUMEN

Neuroblasts in Drosophila divide asymmetrically, sequentially expressing a series of intrinsic factors to generate a diversity of neuron types. These intrinsic factors known as temporal factors dictate timing of neuroblast transitions in response to steroid hormone signaling and specify early versus late temporal fates in neuroblast neuron progeny. After completing their temporal programs, neuroblasts differentiate or die, finalizing both neuron number and type within each neuroblast lineage. From a screen aimed at identifying genes required to terminate neuroblast divisions, we identified Notch and Notch pathway components. When Notch is knocked down, neuroblasts maintain early temporal factor expression longer, delay late temporal factor expression, and continue dividing into adulthood. We find that Delta, expressed in cortex glia, neuroblasts, and after division, their GMC progeny, regulates neuroblast Notch activity. We also find that Delta in neuroblasts is expressed high early, low late, and is controlled by the intrinsic temporal program: early factor Imp promotes Delta, late factors Syp/E93 reduce Delta. Thus, in addition to systemic steroid hormone cues, forward lineage progression is controlled by local cell-cell signaling between neuroblasts and their cortex glia/GMC neighbors: Delta transactivates Notch in neuroblasts bringing the early temporal program and early temporal factor expression to a close.


Asunto(s)
Proteínas de Drosophila , Drosophila , Animales , Drosophila/genética , Proteínas de Drosophila/metabolismo , Neurogénesis/genética , Hormonas/metabolismo , Esteroides/metabolismo , Drosophila melanogaster/genética , Regulación del Desarrollo de la Expresión Génica
18.
Curr Biol ; 34(3): 473-488.e6, 2024 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-38181792

RESUMEN

Complex behaviors depend on the precise developmental specification of neuronal circuits, but the relationship between genetic programs for neural development, circuit structure, and behavioral output is often unclear. The central complex (CX) is a conserved sensory-motor integration center in insects, which governs many higher-order behaviors and largely derives from a small number of type II neural stem cells (NSCs). Here, we show that Imp, a conserved IGF-II mRNA-binding protein expressed in type II NSCs, plays a role in specifying essential components of CX olfactory navigation circuitry. We show the following: (1) that multiple components of olfactory navigation circuitry arise from type II NSCs. (2) Manipulating Imp expression in type II NSCs alters the number and morphology of many of these circuit elements, with the most potent effects on neurons targeting the ventral layers of the fan-shaped body (FB). (3) Imp regulates the specification of Tachykinin-expressing ventral FB input neurons. (4) Imp is required in type II NSCs for establishing proper morphology of the CX neuropil structures. (5) Loss of Imp in type II NSCs abolishes upwind orientation to attractive odor while leaving locomotion and odor-evoked regulation of movement intact. Taken together, our findings establish that a temporally expressed gene can regulate the expression of a complex behavior by developmentally regulating the specification of multiple circuit components and provides a first step toward a developmental dissection of the CX and its roles in behavior.


Asunto(s)
Proteínas de Drosophila , Drosophila melanogaster , Células-Madre Neurales , Proteínas de Unión al ARN , Olfato , Navegación Espacial , Animales , Drosophila melanogaster/genética , Drosophila melanogaster/fisiología , Células-Madre Neurales/metabolismo , Neuronas/fisiología , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/fisiología , Proteínas de Drosophila/genética , Proteínas de Drosophila/fisiología
19.
Cell Rep ; 42(3): 112144, 2023 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-36821439

RESUMEN

How axon guidance pathways are utilized in coordination with temporal and spatial patterning of neural progenitors to regulate neuropil assembly is not well understood. We study this question in the Drosophila medulla using the transmedullary (Tm) projection neurons that target lobula through the inner optic chiasm (IOC). We demonstrate that the Netrin pathway plays multiple roles in guidance of Tm axons and that temporal patterning of medulla neuroblasts determines pioneer versus follower Tm neurons during this process. Loss of Frazzled (Fra) in early-born pioneer Tm neurons leads to IOC defects, while loss of Fra from follower neurons does not affect the IOC. In the follower projection neurons, Fra is required in other targeting steps including lobula branch extension and layer-specific targeting. Furthermore, different from other identified scenarios of Netrin/Fra involved axon guidance in Drosophila, we demonstrate that diffusible Netrin is required for the correct axon targeting and optic lobe organization.


Asunto(s)
Proteínas de Drosophila , Drosophila , Netrina-1 , Animales , Axones/metabolismo , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , Receptores de Netrina/metabolismo , Netrina-1/genética , Netrina-1/metabolismo , Netrinas/genética , Netrinas/metabolismo , Neuronas/metabolismo , Receptores de Superficie Celular/metabolismo
20.
Dev Cell ; 58(22): 2563-2579.e8, 2023 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-37643611

RESUMEN

Development relies on the exquisite control of both the timing and the levels of gene expression to achieve robust developmental transitions. How cis- and trans-acting factors control both aspects simultaneously is unclear. We show that transcriptional pulses of the temporal patterning microRNA (miRNA) lin-4 are generated by two nuclear hormone receptors (NHRs) in C. elegans, NHR-85 and NHR-23, whose mammalian orthologs, Rev-Erb and ROR, function in the circadian clock. Although Rev-Erb and ROR antagonize each other to control once-daily transcription in mammals, NHR-85/NHR-23 heterodimers bind cooperatively to lin-4 regulatory elements to induce a single pulse of expression during each larval stage. Each pulse's timing, amplitude, and duration are dictated by the phased expression of these NHRs and the C. elegans Period ortholog, LIN-42, that binds to and represses NHR-85. Therefore, during nematode temporal patterning, an evolutionary rewiring of circadian clock components couples the timing of gene expression to the control of transcriptional dosage.


Asunto(s)
Proteínas de Caenorhabditis elegans , MicroARNs , Animales , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Redes Reguladoras de Genes , Regulación del Desarrollo de la Expresión Génica , Receptores Citoplasmáticos y Nucleares/metabolismo , Mamíferos/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA