Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 5.666
Filtrar
1.
Development ; 151(10)2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38738602

RESUMEN

Visual circuit development is characterized by subdivision of neuropils into layers that house distinct sets of synaptic connections. We find that, in the Drosophila medulla, this layered organization depends on the axon guidance regulator Plexin A. In Plexin A null mutants, synaptic layers of the medulla neuropil and arborizations of individual neurons are wider and less distinct than in controls. Analysis of semaphorin function indicates that Semaphorin 1a, acting in a subset of medulla neurons, is the primary partner for Plexin A in medulla lamination. Removal of the cytoplasmic domain of endogenous Plexin A has little effect on the formation of medulla layers; however, both null and cytoplasmic domain deletion mutations of Plexin A result in an altered overall shape of the medulla neuropil. These data suggest that Plexin A acts as a receptor to mediate morphogenesis of the medulla neuropil, and as a ligand for Semaphorin 1a to subdivide it into layers. Its two independent functions illustrate how a few guidance molecules can organize complex brain structures by each playing multiple roles.


Asunto(s)
Proteínas de Drosophila , Morfogénesis , Proteínas del Tejido Nervioso , Neurópilo , Lóbulo Óptico de Animales no Mamíferos , Receptores de Superficie Celular , Semaforinas , Animales , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Semaforinas/metabolismo , Semaforinas/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/genética , Morfogénesis/genética , Neurópilo/metabolismo , Lóbulo Óptico de Animales no Mamíferos/metabolismo , Lóbulo Óptico de Animales no Mamíferos/embriología , Receptores de Superficie Celular/metabolismo , Receptores de Superficie Celular/genética , Drosophila melanogaster/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/embriología , Neuronas/metabolismo , Drosophila/metabolismo , Drosophila/embriología , Mutación/genética
2.
PLoS Genet ; 20(4): e1010891, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38683842

RESUMEN

Transcriptional cis-regulatory modules, e.g., enhancers, control the time and location of metazoan gene expression. While changes in enhancers can provide a powerful force for evolution, there is also significant deep conservation of enhancers for developmentally important genes, with function and sequence characteristics maintained over hundreds of millions of years of divergence. Not well understood, however, is how the overall regulatory composition of a locus evolves, with important outstanding questions such as how many enhancers are conserved vs. novel, and to what extent are the locations of conserved enhancers within a locus maintained? We begin here to address these questions with a comparison of the respective single-minded (sim) loci in the two dipteran species Drosophila melanogaster (fruit fly) and Aedes aegypti (mosquito). sim encodes a highly conserved transcription factor that mediates development of the arthropod embryonic ventral midline. We identify two enhancers in the A. aegypti sim locus and demonstrate that they function equivalently in both transgenic flies and transgenic mosquitoes. One A. aegypti enhancer is highly similar to known Drosophila counterparts in its activity, location, and autoregulatory capability. The other differs from any known Drosophila sim enhancers with a novel location, failure to autoregulate, and regulation of expression in a unique subset of midline cells. Our results suggest that the conserved pattern of sim expression in the two species is the result of both conserved and novel regulatory sequences. Further examination of this locus will help to illuminate how the overall regulatory landscape of a conserved developmental gene evolves.


Asunto(s)
Aedes , Drosophila melanogaster , Elementos de Facilitación Genéticos , Regulación del Desarrollo de la Expresión Génica , Animales , Aedes/genética , Aedes/embriología , Drosophila melanogaster/genética , Drosophila melanogaster/embriología , Secuencia Conservada , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Animales Modificados Genéticamente , Evolución Molecular , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo
3.
PLoS Biol ; 22(4): e3002611, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38683880

RESUMEN

As tissues grow and change shape during animal development, they physically pull and push on each other, and these mechanical interactions can be important for morphogenesis. During Drosophila gastrulation, mesoderm invagination temporally overlaps with the convergence and extension of the ectodermal germband; the latter is caused primarily by Myosin II-driven polarised cell intercalation. Here, we investigate the impact of mesoderm invagination on ectoderm extension, examining possible mechanical and mechanotransductive effects on Myosin II recruitment and polarised cell intercalation. We find that the germband ectoderm is deformed by the mesoderm pulling in the orthogonal direction to germband extension (GBE), showing mechanical coupling between these tissues. However, we do not find a significant change in Myosin II planar polarisation in response to mesoderm invagination, nor in the rate of junction shrinkage leading to neighbour exchange events. We conclude that the main cellular mechanism of axis extension, polarised cell intercalation, is robust to the mesoderm invagination pull. We find, however, that mesoderm invagination slows down the rate of anterior-posterior cell elongation that contributes to axis extension, counteracting the tension from the endoderm invagination, which pulls along the direction of GBE.


Asunto(s)
Drosophila melanogaster , Ectodermo , Gastrulación , Mesodermo , Miosina Tipo II , Animales , Mesodermo/embriología , Mesodermo/citología , Gastrulación/fisiología , Ectodermo/citología , Ectodermo/embriología , Ectodermo/metabolismo , Miosina Tipo II/metabolismo , Drosophila melanogaster/embriología , Polaridad Celular , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Embrión no Mamífero , Morfogénesis , Tipificación del Cuerpo/fisiología , Drosophila/embriología
4.
Development ; 151(10)2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38639390

RESUMEN

The planar orientation of cell division (OCD) is important for epithelial morphogenesis and homeostasis. Here, we ask how mechanics and antero-posterior (AP) patterning combine to influence the first divisions after gastrulation in the Drosophila embryonic epithelium. We analyse hundreds of cell divisions and show that stress anisotropy, notably from compressive forces, can reorient division directly in metaphase. Stress anisotropy influences the OCD by imposing metaphase cell elongation, despite mitotic rounding, and overrides interphase cell elongation. In strongly elongated cells, the mitotic spindle adapts its length to, and hence its orientation is constrained by, the cell long axis. Alongside mechanical cues, we find a tissue-wide bias of the mitotic spindle orientation towards AP-patterned planar polarised Myosin-II. This spindle bias is lost in an AP-patterning mutant. Thus, a patterning-induced mitotic spindle orientation bias overrides mechanical cues in mildly elongated cells, whereas in strongly elongated cells the spindle is constrained close to the high stress axis.


Asunto(s)
División Celular , Polaridad Celular , Drosophila melanogaster , Células Epiteliales , Metafase , Huso Acromático , Estrés Mecánico , Animales , Metafase/fisiología , Células Epiteliales/citología , Células Epiteliales/metabolismo , Huso Acromático/metabolismo , Drosophila melanogaster/embriología , Drosophila melanogaster/citología , Polaridad Celular/fisiología , Tipificación del Cuerpo , Miosina Tipo II/metabolismo , Embrión no Mamífero/citología , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Gastrulación/fisiología
5.
Development ; 151(10)2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38646822

RESUMEN

The precise assembly of tissues and organs relies on spatiotemporal regulation of gene expression to coordinate the collective behavior of cells. In Drosophila embryos, the midgut musculature is formed through collective migration of caudal visceral mesoderm (CVM) cells, but how gene expression changes as cells migrate is not well understood. Here, we have focused on ten genes expressed in the CVM and the cis-regulatory sequences controlling their expression. Although some genes are continuously expressed, others are expressed only early or late during migration. Late expression relates to cell cycle progression, as driving string/Cdc25 causes earlier division of CVM cells and accelerates the transition to late gene expression. In particular, we found that the cell cycle effector transcription factor E2F1 is a required input for the late gene CG5080. Furthermore, whereas late genes are broadly expressed in all CVM cells, early gene transcripts are polarized to the anterior or posterior ends of the migrating collective. We show this polarization requires transcription factors Snail, Zfh1 and Dorsocross. Collectively, these results identify two sequential gene expression programs bridged by cell division that support long-distance directional migration of CVM cells.


Asunto(s)
División Celular , Movimiento Celular , Proteínas de Drosophila , Regulación del Desarrollo de la Expresión Génica , Animales , Movimiento Celular/genética , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , División Celular/genética , Mesodermo/metabolismo , Mesodermo/citología , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Drosophila melanogaster/embriología , Factor de Transcripción E2F1/metabolismo , Factor de Transcripción E2F1/genética , Embrión no Mamífero/metabolismo , Embrión no Mamífero/citología , Drosophila/genética , Drosophila/metabolismo , Drosophila/embriología , Factores de Transcripción de la Familia Snail/metabolismo , Factores de Transcripción de la Familia Snail/genética
6.
G3 (Bethesda) ; 14(5)2024 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-38366558

RESUMEN

In Drosophila, the signaling pathway activated by the ligand Folded gastrulation (Fog) is among the few known G protein-coupled receptor (GPCR) pathways to regulate cell shape change with a well-characterized role in gastrulation. However, an understanding of the spectrum of morphogenetic events regulated by Fog signaling is still lacking. Here, we present an analysis of the expression pattern and regulation of fog using a genome-engineered Fog::sfGFP line. We show that Fog expression is widespread and in tissues previously not associated with the signaling pathway including germ cells, trachea, and amnioserosa. In the central nervous system (CNS), we find that the ligand is expressed in multiple types of glia indicating a prominent role in the development of these cells. Consistent with this, we have identified 3 intronic enhancers whose expression in the CNS overlaps with Fog::sfGFP. Further, we show that enhancer-1, (fogintenh-1) located proximal to the coding exon is responsive to AbdA. Supporting this, we find that fog expression is downregulated in abdA mutants. Together, our study highlights the broad scope of Fog-GPCR signaling during embryogenesis and identifies Hox gene AbdA as a novel regulator of fog expression.


Asunto(s)
Proteínas de Drosophila , Gastrulación , Regulación del Desarrollo de la Expresión Génica , Animales , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Gastrulación/genética , Transducción de Señal , Morfogénesis/genética , Elementos de Facilitación Genéticos , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Drosophila melanogaster/genética , Drosophila melanogaster/embriología , Drosophila melanogaster/metabolismo , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/embriología , Embrión no Mamífero/metabolismo
7.
Nature ; 626(7997): 207-211, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38086418

RESUMEN

Enhancers control gene expression and have crucial roles in development and homeostasis1-3. However, the targeted de novo design of enhancers with tissue-specific activities has remained challenging. Here we combine deep learning and transfer learning to design tissue-specific enhancers for five tissues in the Drosophila melanogaster embryo: the central nervous system, epidermis, gut, muscle and brain. We first train convolutional neural networks using genome-wide single-cell assay for transposase-accessible chromatin with sequencing (ATAC-seq) datasets and then fine-tune the convolutional neural networks with smaller-scale data from in vivo enhancer activity assays, yielding models with 13% to 76% positive predictive value according to cross-validation. We designed and experimentally assessed 40 synthetic enhancers (8 per tissue) in vivo, of which 31 (78%) were active and 27 (68%) functioned in the target tissue (100% for central nervous system and muscle). The strategy of combining genome-wide and small-scale functional datasets by transfer learning is generally applicable and should enable the design of tissue-, cell type- and cell state-specific enhancers in any system.


Asunto(s)
Aprendizaje Profundo , Drosophila melanogaster , Embrión no Mamífero , Elementos de Facilitación Genéticos , Redes Neurales de la Computación , Especificidad de Órganos , Animales , Cromatina/genética , Cromatina/metabolismo , Conjuntos de Datos como Asunto , Drosophila melanogaster/embriología , Drosophila melanogaster/genética , Embrión no Mamífero/embriología , Embrión no Mamífero/metabolismo , Elementos de Facilitación Genéticos/genética , Especificidad de Órganos/genética , Reproducibilidad de los Resultados , Análisis de la Célula Individual , Transposasas/metabolismo , Biología Sintética/métodos
8.
Development ; 150(16)2023 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-37526610

RESUMEN

Drosophila is an important model for studying heart development and disease. Yet, single-cell transcriptomic data of its developing heart have not been performed. Here, we report single-cell profiling of the entire fly heart using ∼3000 Hand-GFP embryos collected at five consecutive developmental stages, ranging from bilateral migrating rows of cardiac progenitors to a fused heart tube. The data revealed six distinct cardiac cell types in the embryonic fly heart: cardioblasts, both Svp+ and Tin+ subtypes; and five types of pericardial cell (PC) that can be distinguished by four key transcription factors (Eve, Odd, Ct and Tin) and include the newly described end of the line PC. Notably, the embryonic fly heart combines transcriptional signatures of the mammalian first and second heart fields. Using unique markers for each heart cell type, we defined their number and location during heart development to build a comprehensive 3D cell map. These data provide a resource to track the expression of any gene in the developing fly heart, which can serve as a reference to study genetic perturbations and cardiac diseases.


Asunto(s)
Drosophila melanogaster , Drosophila melanogaster/citología , Drosophila melanogaster/embriología , Corazón/embriología , Análisis de Expresión Génica de una Sola Célula , Ganglios Linfáticos/citología , Ganglios Linfáticos/embriología , Embrión no Mamífero , Desarrollo Embrionario , Biomarcadores , Organogénesis
9.
Curr Biol ; 33(14): 2839-2850.e7, 2023 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-37116484

RESUMEN

The genes that drive development each typically have many different enhancers. Properly coordinating the action of these different enhancers is crucial to correctly specifying cell-fate decisions, yet it remains poorly understood how their activity is choregraphed in time. To shed light on this question, we used recently developed single-cell live imaging tools to dissect the regulation of Fushi tarazu (Ftz) in Drosophila melanogaster embryos. Ftz is a transcription factor that is expressed in asymmetric stripes by two distinct enhancers: autoregulatory and zebra. The anterior edge of each stripe needs to be sharply defined to specify essential lineage boundaries. Here, we tracked how boundary cells commit to either a high-Ftz or low-Ftz fate by measuring Ftz protein traces in real time and simultaneously quantifying transcription from the endogenous locus and individual enhancers. This revealed that the autoregulatory enhancer does not establish this fate choice. Instead, it perpetuates the decision defined by zebra. This is contrary to the prevailing view that autoregulation drives the fate decision by causing bi-stable Ftz expression. Furthermore, we showed that the autoregulatory enhancer is not activated based on a Ftz-concentration threshold but through a timing-based mechanism. We hypothesize that this is regulated by several ubiquitously expressed pioneer-like transcription factors, which have recently been shown to act as timers in the embryo. Our work provides new insight into how precisely timed enhancer activity can directly regulate the dynamics of gene regulatory networks, which may be a general mechanism for ensuring that embryogenesis runs like clockwork.


Asunto(s)
Proteínas de Drosophila , Drosophila melanogaster , Animales , Drosophila melanogaster/embriología , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Elementos de Facilitación Genéticos , Regulación de la Expresión Génica , Proteínas de Homeodominio/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
10.
Cell Syst ; 14(4): 258-272.e4, 2023 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-37080162

RESUMEN

Combinatorial regulation of gene expression by transcription factors (TFs) may in part arise from kinetic synergy-wherein TFs regulate different steps in the transcription cycle. Kinetic synergy requires that TFs play distinguishable kinetic roles. Here, we used live imaging to determine the kinetic roles of three TFs that activate transcription in the Drosophila embryo-Zelda, Bicoid, and Stat92E-by introducing their binding sites into the even-skipped stripe 2 enhancer. These TFs influence different sets of kinetic parameters, and their influence can change over time. All three TFs increased the fraction of transcriptionally active nuclei; Zelda also shortened the first-passage time into transcription and regulated the interval between transcription events. Stat92E also increased the lifetimes of active transcription. Different TFs can therefore play distinct kinetic roles in activating the transcription. This has consequences for understanding the composition and flexibility of regulatory DNA sequences and the biochemical function of TFs. A record of this paper's transparent peer review process is included in the supplemental information.


Asunto(s)
Drosophila melanogaster , Animales , Drosophila melanogaster/embriología , Drosophila melanogaster/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Regiones Promotoras Genéticas , Transcripción Genética , Regulación del Desarrollo de la Expresión Génica , Cinética
11.
Nat Commun ; 14(1): 1357, 2023 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-36914655

RESUMEN

In most eukaryotes, biparentally inherited nuclear genomes and maternally inherited cytoplasmic genomes have different evolutionary interests. Strongly female-biased sex ratios that are repeatedly observed in various arthropods often result from the male-specific lethality (male-killing) induced by maternally inherited symbiotic bacteria such as Spiroplasma and Wolbachia. However, despite some plausible case reports wherein viruses are raised as male-killers, it is not well understood how viruses, having much smaller genomes than bacteria, are capable of inducing male-killing. Here we show that a maternally inherited double-stranded RNA (dsRNA) virus belonging to the family Partitiviridae (designated DbMKPV1) induces male-killing in Drosophila. DbMKPV1 localizes in the cytoplasm and possesses only four genes, i.e., one gene in each of the four genomic segments (dsRNA1-dsRNA4), in contrast to ca. 1000 or more genes possessed by Spiroplasma or Wolbachia. We also show that a protein (designated PVMKp1; 330 amino acids in size), encoded by a gene on the dsRNA4 segment, is necessary and sufficient for inducing male-killing. Our results imply that male-killing genes can be easily acquired by symbiotic viruses through reassortment and that symbiotic viruses are hidden players in arthropod evolution. We anticipate that host-manipulating genes possessed by symbiotic viruses can be utilized for controlling arthropods.


Asunto(s)
Drosophila melanogaster , Genes Virales , Virus de Insectos , Razón de Masculinidad , Simbiosis , Drosophila melanogaster/embriología , Drosophila melanogaster/virología , Virus de Insectos/genética , Genes Virales/fisiología , Masculino , Animales , Desarrollo Embrionario , ARN Viral/fisiología , ARN Bicatenario/fisiología , Evolución Molecular , Sistemas de Lectura Abierta/genética , Caracteres Sexuales , Femenino
12.
Science ; 378(6621): eabg3679, 2022 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-36395225

RESUMEN

The Hippo signaling pathway is widely considered a master regulator of organ growth because of the prominent overgrowth phenotypes caused by experimental manipulation of its activity. Contrary to this model, we show here that removing Hippo transcriptional output did not impair the ability of the mouse liver and Drosophila eyes to grow to their normal size. Moreover, the transcriptional activity of the Hippo pathway effectors Yap/Taz/Yki did not correlate with cell proliferation, and hyperactivation of these effectors induced gene expression programs that did not recapitulate normal development. Concordantly, a functional screen in Drosophila identified several Hippo pathway target genes that were required for ectopic overgrowth but not normal growth. Thus, Hippo signaling does not instruct normal growth, and the Hippo-induced overgrowth phenotypes are caused by the activation of abnormal genetic programs.


Asunto(s)
Drosophila melanogaster , Ojo , Regulación del Desarrollo de la Expresión Génica , Vía de Señalización Hippo , Hígado , Transcripción Genética , Proteínas Coactivadoras Transcripcionales con Motivo de Unión a PDZ , Proteínas Señalizadoras YAP , Animales , Ratones , Drosophila melanogaster/embriología , Drosophila melanogaster/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Ojo/embriología , Vía de Señalización Hippo/genética , Hígado/embriología , Tamaño de los Órganos , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Transactivadores/genética , Proteínas Coactivadoras Transcripcionales con Motivo de Unión a PDZ/metabolismo , Proteínas Señalizadoras YAP/metabolismo
13.
Science ; 377(6606): eabn5800, 2022 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-35926038

RESUMEN

Drosophila melanogaster is a powerful, long-standing model for metazoan development and gene regulation. We profiled chromatin accessibility in almost 1 million and gene expression in half a million nuclei from overlapping windows spanning the entirety of embryogenesis. Leveraging developmental asynchronicity within embryo collections, we applied deep neural networks to infer the age of each nucleus, resulting in continuous, multimodal views of molecular and cellular transitions in absolute time. We identify cell lineages; infer their developmental relationships; and link dynamic changes in enhancer usage, transcription factor (TF) expression, and the accessibility of TFs' cognate motifs. With these data, the dynamics of enhancer usage and gene expression can be explored within and across lineages at the scale of minutes, including for precise transitions like zygotic genome activation.


Asunto(s)
Proteínas de Drosophila , Drosophila melanogaster , Desarrollo Embrionario , Regulación del Desarrollo de la Expresión Génica , Animales , Linaje de la Célula/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citología , Drosophila melanogaster/embriología , Drosophila melanogaster/genética , Desarrollo Embrionario/genética , Elementos de Facilitación Genéticos , Redes Neurales de la Computación , Análisis de la Célula Individual
14.
Proc Natl Acad Sci U S A ; 119(26): e2200780119, 2022 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-35733247

RESUMEN

In the metazoan S phase, coordinated firing of clusters of origins replicates different parts of the genome in a temporal program. Despite advances, neither the mechanism controlling timing nor that coordinating firing of multiple origins is fully understood. Rif1, an evolutionarily conserved inhibitor of DNA replication, recruits protein phosphatase 1 (PP1) and counteracts firing of origins by S-phase kinases. During the midblastula transition (MBT) in Drosophila embryos, Rif1 forms subnuclear hubs at each of the large blocks of satellite sequences and delays their replication. Each Rif1 hub disperses abruptly just prior to the replication of the associated satellite sequences. Here, we show that the level of activity of the S-phase kinase, DDK, accelerated this dispersal program, and that the level of Rif1-recruited PP1 retarded it. Further, Rif1-recruited PP1 supported chromatin association of nearby Rif1. This influence of nearby Rif1 can create a "community effect" counteracting kinase-induced dissociation such that an entire hub of Rif1 undergoes switch-like dispersal at characteristic times that shift in response to the balance of Rif1-PP1 and DDK activities. We propose a model in which the spatiotemporal program of late replication in the MBT embryo is controlled by self-stabilizing Rif1-PP1 hubs, whose abrupt dispersal synchronizes firing of associated late origins.


Asunto(s)
Proteínas Portadoras , Replicación del ADN , Proteínas de Drosophila , Drosophila melanogaster , Proteína Fosfatasa 1 , Origen de Réplica , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/embriología , Drosophila melanogaster/genética , Proteína Fosfatasa 1/genética , Proteína Fosfatasa 1/metabolismo , Fase S/genética , Proteínas de Unión a Telómeros/genética , Proteínas de Unión a Telómeros/metabolismo
15.
J Cell Biol ; 221(6)2022 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-35404399

RESUMEN

During tissue morphogenesis, the changes in cell shape, resulting from cell-generated forces, often require active regulation of intracellular trafficking. How mechanical stimuli influence intracellular trafficking and how such regulation impacts tissue mechanics are not fully understood. In this study, we identify an actomyosin-dependent mechanism involving Rab11-mediated trafficking in regulating apical constriction in the Drosophila embryo. During Drosophila mesoderm invagination, apical actin and Myosin II (actomyosin) contractility induces apical accumulation of Rab11-marked vesicle-like structures ("Rab11 vesicles") by promoting a directional bias in dynein-mediated vesicle transport. At the apical domain, Rab11 vesicles are enriched near the adherens junctions (AJs). The apical accumulation of Rab11 vesicles is essential to prevent fragmented apical AJs, breaks in the supracellular actomyosin network, and a reduction in the apical constriction rate. This Rab11 function is separate from its role in promoting apical Myosin II accumulation. These findings suggest a feedback mechanism between actomyosin activity and Rab11-mediated intracellular trafficking that regulates the force generation machinery during tissue folding.


Asunto(s)
Actomiosina , Proteínas de Drosophila , Drosophila melanogaster , Proteínas de Unión al GTP rab , Actomiosina/metabolismo , Animales , Constricción , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/embriología , Drosophila melanogaster/metabolismo , Morfogénesis , Miosina Tipo II/metabolismo , Proteínas de Unión al GTP rab/metabolismo
16.
Sci Rep ; 12(1): 2056, 2022 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-35136137

RESUMEN

Drosophila melanogaster tumor models are growing in popularity, driven by the high degree of genetic as well as functional conservation to humans. The most common method to measure the effects of a tumor on distant organs of a human cancer patient is to use computed tomography (CT), often used in diagnosing cachexia, a debilitating cancer-induced syndrome most visibly characterized by loss of muscle mass. Successful application of high resolution micro-CT scanning of D. melanogaster was recently reported and we here present the segmentation of all visible larval organs at several stages of tumor development. We previously showed the strong expected reduction in muscle mass as the tumor develops, and we here report a surprisingly strong reduction also in gut and Malpighian tubules (kidney) volume. Time-point of tumor development was found to have a stronger correlation to cachectic organ volume loss than tumor volume, giving support to the previously proposed idea that tumor size does not directly determine degree of cachexia.


Asunto(s)
Caquexia/patología , Drosophila melanogaster/genética , Tracto Gastrointestinal/patología , Túbulos de Malpighi/patología , Neoplasias/patología , Animales , Modelos Animales de Enfermedad , Drosophila melanogaster/embriología , Drosophila melanogaster/crecimiento & desarrollo , Humanos , Larva/crecimiento & desarrollo , Tamaño de los Órganos/fisiología , Microtomografía por Rayos X
17.
G3 (Bethesda) ; 12(4)2022 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-35143618

RESUMEN

MicroRNAs can have subtle and combinatorial effects on the levels of the targets and pathways they act on. Studying the consequences of a single microRNA knockout often proves difficult as many such knockouts exhibit phenotypes only under stress conditions. This has often led to the hypothesis that microRNAs buffer the effects of intrinsic and environmental stochasticity on gene expression. Observing and understanding this buffering effect entails quantitative analysis of microRNA and target expression in single cells. To this end, we have employed single-molecule fluorescence in situ hybridization, immunofluorescence, and high-resolution confocal microscopy to investigate the effects of miR-9a loss on the expression of the serine-protease Rhomboid in Drosophila melanogaster early embryos. Our single-cell quantitative approach shows that spatially, the rhomboid mRNA pattern is identical in WT and miR-9a knockout embryos. However, we find that the number of mRNA molecules per cell is higher when miR-9a is absent, and the level and temporal accumulation of rhomboid protein shows a more dramatic increase in the miR-9a knockout. Specifically, we see accumulation of rhomboid protein in miR-9a mutants by stage 5, much earlier than in WT. The data, therefore, show that miR-9a functions in the regulation of rhomboid mRNA and protein levels. While further work is required to establish whether rhomboid is a direct target of miR-9 in Drosophila, our results further establish the miR-9 family microRNAs as conserved regulators of timing in neurogenic processes. This study shows the power of single-cell quantification as an experimental tool to study phenotypic consequences of microRNA mis-regulation.


Asunto(s)
Proteínas de Drosophila , MicroARNs , Animales , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/embriología , Drosophila melanogaster/metabolismo , Regulación del Desarrollo de la Expresión Génica , Hibridación Fluorescente in Situ , MicroARNs/genética , ARN Mensajero/genética
18.
Nat Commun ; 13(1): 859, 2022 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-35165263

RESUMEN

Maternal RNA degradation is critical for embryogenesis and is tightly controlled by maternal RNA-binding proteins. Fragile X mental-retardation protein (FMR1) binds target mRNAs to form ribonucleoprotein (RNP) complexes/granules that control various biological processes, including early embryogenesis. However, how FMR1 recognizes target mRNAs and how FMR1-RNP granule assembly/disassembly regulates FMR1-associated mRNAs remain elusive. Here we show that Drosophila FMR1 preferentially binds mRNAs containing m6A-marked "AGACU" motif with high affinity to contributes to maternal RNA degradation. The high-affinity binding largely depends on a hydrophobic network within FMR1 KH2 domain. Importantly, this binding greatly induces FMR1 granule condensation to efficiently recruit unmodified mRNAs. The degradation of maternal mRNAs then causes granule de-condensation, allowing normal embryogenesis. Our findings reveal that sequence-specific mRNAs instruct FMR1-RNP granules to undergo a dynamic phase-switch, thus contributes to maternal mRNA decay. This mechanism may represent a general principle that regulated RNP-granules control RNA processing and normal development.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/embriología , Desarrollo Embrionario/genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Metiltransferasas/metabolismo , Estabilidad del ARN/genética , Animales , Gránulos Citoplasmáticos/metabolismo , Embrión no Mamífero/embriología , Metilación , Dominios Proteicos/genética , ARN Mensajero/genética , Proteínas de Unión al ARN/metabolismo
19.
Cells ; 11(2)2022 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-35053396

RESUMEN

Patients with Alzheimer's disease suffer from a decrease in brain mass and a prevalence of amyloid-ß plaques. These plaques are thought to play a role in disease progression, but their exact role is not entirely established. We developed an optogenetic model to induce amyloid-ß intracellular oligomerization to model distinct disease etiologies. Here, we examine the effect of Wnt signaling on amyloid in an optogenetic, Drosophila gut stem cell model. We observe that Wnt activation rescues the detrimental effects of amyloid expression and oligomerization. We analyze the gene expression changes downstream of Wnt that contribute to this rescue and find changes in aging related genes, protein misfolding, metabolism, and inflammation. We propose that Wnt expression reduces inflammation through repression of Toll activating factors. We confirm that chronic Toll activation reduces lifespan, but a decrease in the upstream activator Persephone extends it. We propose that the protective effect observed for lithium treatment functions, at least in part, through Wnt activation and the inhibition of inflammation.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Drosophila melanogaster/metabolismo , Intestinos/patología , Células Madre/patología , Vía de Señalización Wnt , Animales , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/efectos de los fármacos , Drosophila melanogaster/embriología , Embrión no Mamífero/efectos de los fármacos , Embrión no Mamífero/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Longevidad/efectos de los fármacos , Optogenética , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Vía de Señalización Wnt/efectos de los fármacos , Vía de Señalización Wnt/genética
20.
Sci Rep ; 12(1): 292, 2022 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-34997175

RESUMEN

Neurogenesis in the Drosophila central brain progresses dynamically in order to generate appropriate numbers of neurons during different stages of development. Thus, a central challenge in neurobiology is to reveal the molecular and genetic mechanisms of neurogenesis timing. Here, we found that neurogenesis is significantly impaired when a novel mutation, Nuwa, is induced at early but not late larval stages. Intriguingly, when the Nuwa mutation is induced in neuroblasts of olfactory projection neurons (PNs) at the embryonic stage, embryonic-born PNs are generated, but larval-born PNs of the same origin fail to be produced. Through molecular characterization and transgenic rescue experiments, we determined that Nuwa is a loss-of-function mutation in Drosophila septin interacting protein 1 (sip1). Furthermore, we found that SIP1 expression is enriched in neuroblasts, and RNAi knockdown of sip1 using a neuroblast driver results in formation of small and aberrant brains. Finally, full-length SIP1 protein and truncated SIP1 proteins lacking either the N- or C-terminus display different subcellular localization patterns, and only full-length SIP1 can rescue the Nuwa-associated neurogenesis defect. Taken together, these results suggest that SIP1 acts as a crucial factor for specific neurogenesis programs in the early developing larval brain.


Asunto(s)
Encéfalo/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Neurogénesis , Neuronas/metabolismo , Animales , Animales Modificados Genéticamente , Encéfalo/embriología , Proteínas de Drosophila/genética , Drosophila melanogaster/embriología , Drosophila melanogaster/genética , Regulación del Desarrollo de la Expresión Génica , Larva/genética , Larva/metabolismo , Mutación con Pérdida de Función , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA