Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 41(3): 111495, 2022 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-36261011

RESUMEN

Somatic adult stem cell lineages in high-turnover tissues are under tight gene regulatory control. Like its mammalian counterpart, the Drosophila intestine precisely adjusts the rate of stem cell division with the onset of differentiation based on physiological demand. Although Notch signaling is indispensable for these decisions, the regulation of Notch activity that drives the differentiation of stem cell progenies into functional, mature cells is not well understood. Here, we report that commitment to the terminally differentiated enterocyte (EC) cell fate is under microRNA control. We show that an intestinally enriched microRNA, miR-956, fine-tunes Notch signaling activity specifically in intermediate, enteroblast (EB) progenitor cells to control EC differentiation. We further identify insensitive mRNA as a target of miR-956 that regulates EB/EC ratios by repressing Notch activity in EBs. In summary, our study highlights a post-transcriptional gene-regulatory mechanism for controlling differentiation in an adult intestinal stem cell lineage.


Asunto(s)
Proteínas de Drosophila , MicroARNs , Animales , Drosophila/genética , Proteínas de Drosophila/genética , Receptores Notch/genética , Drosophila melanogaster/fisiología , MicroARNs/genética , Intestinos , ARN Mensajero , Mamíferos/genética
2.
Front Cell Dev Biol ; 10: 909212, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35784477

RESUMEN

Differential processing is a hallmark of clustered microRNAs (miRNAs) and the role of position and order of miRNAs in a cluster together with the contribution of stem-base and terminal loops has not been explored extensively within the context of a polycistronic transcript. To elucidate the structural attributes of a polycistronic transcript that contribute towards the differences in efficiencies of processing of the co-transcribed miRNAs, we constructed a series of chimeric variants of Drosophila let-7-Complex that encodes three evolutionary conserved and differentially expressed miRNAs (miR-100, let-7 and miR-125) and examined the expression and biological activity of the encoded miRNAs. The kinetic effects of Drosha and Dicer processing on the chimeric precursors were examined by in vitro processing assays. Our results highlight the importance of stem-base and terminal loop sequences in differential expression of polycistronic miRNAs and provide evidence that processing of a particular miRNA in a polycistronic transcript is in part determined by the kinetics of processing of adjacent miRNAs in the same cluster. Overall, this analysis provides specific guidelines for achieving differential expression of a particular miRNA in a cluster by structurally induced changes in primary miRNA (pri-miRNA) sequences.

3.
Genetics ; 222(2)2022 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-35762963

RESUMEN

The regulation of stem cell survival, self-renewal, and differentiation is critical for the maintenance of tissue homeostasis. Although the involvement of signaling pathways and transcriptional control mechanisms in stem cell regulation have been extensively investigated, the role of post-transcriptional control is still poorly understood. Here, we show that the nuclear activity of the RNA-binding protein Second Mitotic Wave Missing is critical for Drosophila melanogaster intestinal stem cells and their daughter cells, enteroblasts, to maintain their progenitor cell properties and functions. Loss of swm causes intestinal stem cells and enteroblasts to stop dividing and instead detach from the basement membrane, resulting in severe progenitor cell loss. swm loss is further characterized by nuclear accumulation of poly(A)+ RNA in progenitor cells. Second Mitotic Wave Missing associates with transcripts involved in epithelial cell maintenance and adhesion, and the loss of swm, while not generally affecting the levels of these Second Mitotic Wave Missing-bound mRNAs, leads to elevated expression of proteins encoded by some of them, including the fly ortholog of Filamin. Taken together, this study indicates a nuclear role for Second Mitotic Wave Missing in adult stem cell maintenance, raising the possibility that nuclear post-transcriptional regulation of mRNAs encoding cell adhesion proteins ensures proper attachment of progenitor cells.


Asunto(s)
Proteínas de Drosophila , Drosophila melanogaster , Animales , Diferenciación Celular/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Filaminas/metabolismo , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Células Madre/metabolismo
4.
Curr Biol ; 32(2): 386-397.e6, 2022 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-34875230

RESUMEN

The role of processing bodies (P-bodies), key sites of post-transcriptional control, in adult stem cells remains poorly understood. Here, we report that adult Drosophila intestinal stem cells, but not surrounding differentiated cells such as absorptive enterocytes (ECs), harbor P-bodies that contain Drosophila orthologs of mammalian P-body components DDX6, EDC3, EDC4, and LSM14A/B. A targeted RNAi screen in intestinal progenitor cells identified 39 previously known and 64 novel P-body regulators, including Patr-1, a gene necessary for P-body assembly. Loss of Patr-1-dependent P-bodies leads to a loss of stem cells that is associated with inappropriate expression of EC-fate gene nubbin. Transcriptomic analysis of progenitor cells identifies a cadre of such weakly transcribed pro-differentiation transcripts that are elevated after P-body loss. Altogether, this study identifies a P-body-dependent repression activity that coordinates with known transcriptional repression programs to maintain a population of in vivo stem cells in a state primed for differentiation.


Asunto(s)
Proteínas de Drosophila , Drosophila , Animales , Diferenciación Celular/genética , Drosophila/genética , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Intestinos , Mamíferos , Células Madre/metabolismo
5.
Elife ; 102021 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-34100717

RESUMEN

Dietary restriction (DR) extends healthy lifespan in diverse species. Age and nutrient-related changes in the abundance of microRNAs (miRNAs) and their processing factors have been linked to organismal longevity. However, the mechanisms by which they modulate lifespan and the tissue-specific role of miRNA-mediated networks in DR-dependent enhancement of lifespan remains largely unexplored. We show that two neuronally enriched and highly conserved microRNAs, miR-125 and let-7 mediate the DR response in Drosophila melanogaster. Functional characterization of miR-125 demonstrates its role in neurons while its target chinmo acts both in neurons and the fat body to modulate fat metabolism and longevity. Proteomic analysis revealed that Chinmo exerts its DR effects by regulating the expression of FATP, CG2017, CG9577, CG17554, CG5009, CG8778, CG9527, and FASN1. Our findings identify miR-125 as a conserved effector of the DR pathway and open the avenue for this small RNA molecule and its downstream effectors to be considered as potential drug candidates for the treatment of late-onset diseases and biomarkers for healthy aging in humans.


Asunto(s)
Restricción Calórica , Proteínas de Drosophila/metabolismo , Longevidad/fisiología , MicroARNs/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Animales , Línea Celular , Drosophila , Proteínas de Drosophila/análisis , Proteínas de Drosophila/química , Embrión no Mamífero , Femenino , Transducción de Señal/fisiología
6.
Proc Natl Acad Sci U S A ; 118(21)2021 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-34016750

RESUMEN

Adult organisms must sense and adapt to environmental fluctuations. In high-turnover tissues such as the intestine, these adaptive responses require rapid changes in gene expression that, in turn, likely involve posttranscriptional gene control. However, intestinal-tissue-specific microRNA (miRNA)-mediated regulatory pathways remain unexplored. Here, we report the role of an intestinal-specific miRNA, miR-958, that non-cell autonomously regulates stem cell numbers during tissue homeostasis and regeneration in the Drosophila adult midgut. We identify its downstream target cabut, the Drosophila ortholog of mammalian KLF10/11 transcription factors, which mediates this miR-958 function by promoting paracrine enterocyte-to-stem-cell bone morphogenetic protein (BMP) signaling. We also show that mature miR-958 levels transiently decrease in response to stress and that this decrease is required for proper stem cell expansion during tissue regeneration. In summary, we have identified a posttranscriptional mechanism that modulates BMP signaling activity within Drosophila adult intestinal tissue during both normal homeostasis and tissue regeneration to regulate intestinal stem cell numbers.


Asunto(s)
Proteínas Morfogenéticas Óseas/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Enterocitos/metabolismo , MicroARNs/genética , Células Madre/metabolismo , Factores de Transcripción/genética , Animales , Bleomicina/farmacología , Proteínas Morfogenéticas Óseas/metabolismo , Recuento de Células , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Enterocitos/citología , Regulación de la Expresión Génica , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Homeostasis/genética , MicroARNs/metabolismo , Regeneración/genética , Transducción de Señal , Células Madre/citología , Factores de Transcripción/metabolismo
7.
Development ; 148(1)2021 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-33246929

RESUMEN

The adult Drosophila intestinal epithelium is a model system for stem cell biology, but its utility is limited by current biochemical methods that lack cell type resolution. Here, we describe a new proximity-based profiling method that relies upon a GAL4 driver, termed intestinal-kickout-GAL4 (I-KCKT-GAL4), that is exclusively expressed in intestinal progenitor cells. This method uses UV crosslinked whole animal frozen powder as its starting material to immunoprecipitate the RNA cargoes of transgenic epitope-tagged RNA binding proteins driven by I-KCKT-GAL4 When applied to the general mRNA-binder, poly(A)-binding protein, the RNA profile obtained by this method identifies 98.8% of transcripts found after progenitor cell sorting, and has low background noise despite being derived from whole animal lysate. We also mapped the targets of the more selective RNA binder, Fragile X mental retardation protein (FMRP), using enhanced crosslinking and immunoprecipitation (eCLIP), and report for the first time its binding motif in Drosophila cells. This method will therefore enable the RNA profiling of wild-type and mutant intestinal progenitor cells from intact flies exposed to normal and altered environments, as well as the identification of RNA-protein interactions crucial for stem cell function.


Asunto(s)
Envejecimiento/fisiología , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Técnicas Genéticas , Intestinos/citología , ARN/metabolismo , Células Madre/metabolismo , Factores de Transcripción/metabolismo , Animales , Secuencia de Bases , Elementos de Facilitación Genéticos/genética , Femenino , Regulación de la Expresión Génica , Especificidad de Órganos , Proteínas de Unión a Poli(A)/metabolismo , ARN/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo
8.
G3 (Bethesda) ; 10(11): 4271-4285, 2020 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-32972999

RESUMEN

Balancers are rearranged chromosomes used in Drosophila melanogaster to maintain deleterious mutations in stable populations, preserve sets of linked genetic elements and construct complex experimental stocks. Here, we assess the phenotypes associated with breakpoint-induced mutations on commonly used third chromosome balancers and show remarkably few deleterious effects. We demonstrate that a breakpoint in p53 causes loss of radiation-induced apoptosis and a breakpoint in Fucosyltransferase A causes loss of fucosylation in nervous and intestinal tissue-the latter study providing new markers for intestinal cell identity and challenging previous conclusions about the regulation of fucosylation. We also describe thousands of potentially harmful mutations shared among X or third chromosome balancers, or unique to specific balancers, including an Ankyrin2 mutation present on most TM3 balancers, and reiterate the risks of using balancers as experimental controls. We used long-read sequencing to confirm or refine the positions of two inversions with breakpoints lying in repetitive sequences and provide evidence that one of the inversions, In(2L)Cy, arose by ectopic recombination between foldback transposon insertions and the other, In(3R)C, cleanly separates subtelomeric and telomeric sequences and moves the subtelomeric sequences to an internal chromosome position. In addition, our characterization of In(3R)C shows that balancers may be polymorphic for terminal deletions. Finally, we present evidence that extremely distal mutations on balancers can add to the stability of stocks whose purpose is to maintain homologous chromosomes carrying mutations in distal genes. Overall, these studies add to our understanding of the structure, diversity and effectiveness of balancer chromosomes.


Asunto(s)
Cromosomas , Drosophila melanogaster , Animales , Inversión Cromosómica , Drosophila melanogaster/genética , Mutación , Fenotipo
9.
Genetics ; 216(4): 891-903, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32988987

RESUMEN

The Drosophila adult midgut is a model epithelial tissue composed of a few major cell types with distinct regional identities. One of the limitations to its analysis is the lack of tools to manipulate gene expression based on these regional identities. To overcome this obstacle, we applied the intersectional split-GAL4 system to the adult midgut and report 653 driver combinations that label cells by region and cell type. We first identified 424 split-GAL4 drivers with midgut expression from ∼7300 drivers screened, and then evaluated the expression patterns of each of these 424 when paired with three reference drivers that report activity specifically in progenitor cells, enteroendocrine cells, or enterocytes. We also evaluated a subset of the drivers expressed in progenitor cells for expression in enteroblasts using another reference driver. We show that driver combinations can define novel cell populations by identifying a driver that marks a distinct subset of enteroendocrine cells expressing genes usually associated with progenitor cells. The regional cell type patterns associated with the entire set of driver combinations are documented in a freely available website, providing information for the design of thousands of additional driver combinations to experimentally manipulate small subsets of intestinal cells. In addition, we show that intestinal enhancers identified with the split-GAL4 system can confer equivalent expression patterns on other transgenic reporters. Altogether, the resource reported here will enable more precisely targeted gene expression for studying intestinal processes, epithelial cell functions, and diseases affecting self-renewing tissues.


Asunto(s)
Proteínas de Drosophila/genética , Elementos de Facilitación Genéticos , Marcación de Gen/métodos , Ingeniería Genética/métodos , Mucosa Intestinal/citología , Factores de Transcripción/genética , Animales , Drosophila melanogaster , Células Enteroendocrinas/metabolismo , Mucosa Intestinal/metabolismo , Regiones Promotoras Genéticas
10.
Trends Microbiol ; 28(6): 430-432, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32396824

RESUMEN

Interactions between the eukaryotic host, microbiome members, and invading pathogens help to shape disease outcomes. Using the Drosophila model, Fast et al. identified that Vibrio cholerae acts to inhibit epithelial renewal through complex interactions between the type VI secretion system of V. cholerae and the microbial community of the fly.


Asunto(s)
Cólera , Microbiota , Sistemas de Secreción Tipo VI , Vibrio cholerae , Animales , Drosophila
11.
J Cell Sci ; 133(10)2020 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-32265270

RESUMEN

Stressed cells downregulate translation initiation and assemble membrane-less foci termed stress granules (SGs). Although SGs have been extensively characterized in cultured cells, the existence of such structures in stressed adult stem cell pools remains poorly characterized. Here, we report that the Drosophila orthologs of the mammalian SG components AGO1, ATX2, CAPRIN, eIF4E, FMRP, G3BP, LIN-28, PABP and TIAR are enriched in adult fly intestinal progenitor cells, where they accumulate in small cytoplasmic messenger ribonucleoprotein complexes (mRNPs). Treatment with sodium arsenite or rapamycin reorganized these mRNPs into large cytoplasmic granules. Formation of these intestinal progenitor stress granules (IPSGs) depended on polysome disassembly, led to translational downregulation and was reversible. Although the canonical SG nucleators ATX2 and G3BP were sufficient for IPSG formation in the absence of stress, neither of them, nor TIAR, either individually or collectively, were required for stress-induced IPSG formation. This work therefore finds that IPSGs do not assemble via a canonical mechanism, raising the possibility that other stem cell populations employ a similar stress-response mechanism.


Asunto(s)
Proteínas de Drosophila , Drosophila , Animales , Proteínas Argonautas , Línea Celular , Células Cultivadas , Gránulos Citoplasmáticos , Proteínas de Drosophila/genética , Polirribosomas , Proteínas de Unión al ARN
12.
Development ; 146(17)2019 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-31399469

RESUMEN

The dramatic growth that occurs during Drosophila larval development requires rapid conversion of nutrients into biomass. Many larval tissues respond to these biosynthetic demands by increasing carbohydrate metabolism and lactate dehydrogenase (LDH) activity. The resulting metabolic program is ideally suited for synthesis of macromolecules and mimics the manner by which cancer cells rely on aerobic glycolysis. To explore the potential role of Drosophila LDH in promoting biosynthesis, we examined how Ldh mutations influence larval development. Our studies unexpectedly found that Ldh mutants grow at a normal rate, indicating that LDH is dispensable for larval biomass production. However, subsequent metabolomic analyses suggested that Ldh mutants compensate for the inability to produce lactate by generating excess glycerol-3-phosphate (G3P), the production of which also influences larval redox balance. Consistent with this possibility, larvae lacking both LDH and G3P dehydrogenase (GPDH1) exhibit growth defects, synthetic lethality and decreased glycolytic flux. Considering that human cells also generate G3P upon inhibition of lactate dehydrogenase A (LDHA), our findings hint at a conserved mechanism in which the coordinate regulation of lactate and G3P synthesis imparts metabolic robustness to growing animal tissues.


Asunto(s)
Drosophila melanogaster/fisiología , Glicerolfosfato Deshidrogenasa/metabolismo , L-Lactato Deshidrogenasa/metabolismo , Larva/crecimiento & desarrollo , Larva/metabolismo , Azúcares/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Animales Modificados Genéticamente , Femenino , Glicerolfosfato Deshidrogenasa/genética , Glucólisis/genética , Homeostasis/genética , L-Lactato Deshidrogenasa/genética , Ácido Láctico/biosíntesis , Masculino , Mutación , NAD/metabolismo , Oxidación-Reducción
13.
PLoS Genet ; 12(8): e1006247, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27508495

RESUMEN

Messenger RNAs (mRNAs) often contain binding sites for multiple, different microRNAs (miRNAs). However, the biological significance of this feature is unclear, since such co-targeting miRNAs could function coordinately, independently, or redundantly with one another. Here, we show that two co-transcribed Drosophila miRNAs, let-7 and miR-125, non-redundantly regulate a common target, the transcription factor Chronologically Inappropriate Morphogenesis (Chinmo). We first characterize novel adult phenotypes associated with loss of both let-7 and miR-125, which are derived from a common, polycistronic transcript that also encodes a third miRNA, miR-100. Consistent with the coordinate upregulation of all three miRNAs in aging flies, these phenotypes include brain degeneration and shortened lifespan. However, transgenic rescue analysis reveal separable roles for these miRNAs: adult miR-125 but not let-7 mutant phenotypes are associated with ectopic Chinmo expression in adult brains and are suppressed by chinmo reduction. In contrast, let-7 is predominantly responsible for regulating chinmo during nervous system formation. These results indicate that let-7 and miR-125 function during two distinct stages, development and adulthood, rather than acting at the same time. These different activities are facilitated by an increased rate of processing of let-7 during development and a lower rate of decay of the accumulated miR-125 in the adult nervous system. Thus, this work not only establishes a key role for the highly conserved miR-125 in aging. It also demonstrates that two co-transcribed miRNAs function independently during distinct stages to regulate a common target, raising the possibility that such biphasic control may be a general feature of clustered miRNAs.


Asunto(s)
Proteínas de Drosophila/genética , Longevidad/genética , MicroARNs/genética , Proteínas del Tejido Nervioso/genética , Envejecimiento/genética , Envejecimiento/patología , Animales , Sitios de Unión , Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Drosophila/genética , Drosophila/crecimiento & desarrollo , Proteínas de Drosophila/biosíntesis , Regulación del Desarrollo de la Expresión Génica , MicroARNs/biosíntesis , Morfogénesis/genética , Proteínas del Tejido Nervioso/biosíntesis , Sistema Nervioso/crecimiento & desarrollo , Sistema Nervioso/patología , Neuronas/metabolismo
14.
Elife ; 52016 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-27296804

RESUMEN

Pediatric neural tumors are often initiated during early development and can undergo very rapid transformation. However, the molecular basis of this early malignant susceptibility remains unknown. During Drosophila development, neural stem cells (NSCs) divide asymmetrically and generate intermediate progenitors that rapidly differentiate in neurons. Upon gene inactivation, these progeny can dedifferentiate and generate malignant tumors. Here, we find that intermediate progenitors are prone to malignancy only when born during an early window of development while expressing the transcription factor Chinmo, and the mRNA-binding proteins Imp/IGF2BP and Lin-28. These genes compose an oncogenic module that is coopted upon dedifferentiation of early-born intermediate progenitors to drive unlimited tumor growth. In late larvae, temporal transcription factor progression in NSCs silences the module, thereby limiting mitotic potential and terminating the window of malignant susceptibility. Thus, this study identifies the gene regulatory network that confers malignant potential to neural tumors with early developmental origins.


Asunto(s)
Carcinogénesis , Diferenciación Celular , Proliferación Celular , Susceptibilidad a Enfermedades , Drosophila/embriología , Células-Madre Neurales/fisiología , Animales , Proteínas de Drosophila/biosíntesis , Proteínas del Tejido Nervioso/biosíntesis , Proteínas de Unión al ARN/biosíntesis , Factores de Tiempo
15.
Nucleic Acids Res ; 42(8): 5245-55, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24561617

RESUMEN

Adenosine deaminases acting on RNAs (ADARs) convert adenosine residues to inosines in primary microRNA (pri-miRNA) transcripts to alter the structural conformation of these precursors and the subsequent functions of the encoded microRNAs (miRNAs). Here we show that RNA editing by Drosophila ADAR modulates the expression of three co-transcribed miRNAs encoded by the evolutionarily conserved let-7-Complex (let-7-C) locus. For example, a single A-to-I change at the -6 residue of pri-miR-100, the first miRNA in this let-7-C polycistronic transcript, leads to enhanced miRNA processing by Drosha and consequently enhanced functional miR-100 both in vitro as well as in vivo. In contrast, other editing events, including one at the +43 residue of the pri-miR-125, destabilize the primary transcript and reduce the levels of all three encoded miRNAs. Consequently, loss of adar in vivo leads to reduced miR-100 but increased miR-125. In wild-type animals, the destabilizing editing events in pri-let-7-C increase during the larval-to-adult transition and are critical for the normal downregulation of all three miRNAs seen late in metamorphosis. These findings unravel a new regulatory role for ADAR and raise the possibility that ADAR mediates the differential expression characteristic of many polycistronic miRNA clusters.


Asunto(s)
Adenosina Desaminasa/metabolismo , Proteínas de Drosophila/metabolismo , MicroARNs/metabolismo , Edición de ARN , Animales , Drosophila melanogaster/genética , Drosophila melanogaster/crecimiento & desarrollo , Metamorfosis Biológica/genética
16.
Proc Natl Acad Sci U S A ; 111(4): 1421-6, 2014 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-24474768

RESUMEN

Cleavage of microRNAs and mRNAs by Drosha and its cofactor Pasha/DGCR8 is required for animal development, but whether these proteins also have independent roles in development has been unclear. Known phenotypes associated with loss of either one of these two proteins are very similar and consistent with their joint function, even though both cofactors are involved with additional distinct RNA biogenesis pathways. Here, we report clear phenotypic differences between drosha and pasha/dgcr8 null alleles in two postembryonic lineages in the Drosophila brain: elimination of pasha/dgcr8 leads to defects that are not shared by drosha null mutations in the morphology of gamma neurons in the mushroom body lineage, as well as many neurons in the anterodorsal projection neuron lineage. These morphological defects are not detected in neurons that are genetically depleted of two additional microRNA pathway components, dicer-1 and argonaute1, indicating that they are not due to loss of microRNA activity. They are, however, phenocopied by a newly identified recessive gain-of-function allele in drosha that probably interferes with the microRNA independent functions of Pasha/DGCR8. These data therefore identify a general Drosha-independent DGCR8/Pasha pathway that promotes proper morphology in multiple neuronal lineages. Given that reduction of human DGCR8/Pasha may contribute to the cognitive and behavioral characteristics of DiGeorge syndrome patients, disruption of this newly described pathway could underlie human neurological disease.


Asunto(s)
Proteínas de Drosophila/fisiología , Morfogénesis , Neuronas/citología , Proteínas de Unión al ARN/fisiología , Ribonucleasa III/fisiología , Alelos , Animales , Proteínas de Drosophila/genética , MicroARNs/genética , Fenotipo , Procesamiento Postranscripcional del ARN , Proteínas de Unión al ARN/genética , Ribonucleasa III/genética
17.
Curr Top Dev Biol ; 105: 97-123, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23962840

RESUMEN

MicroRNAs (miRNAs) ensure progression through development by synchronizing cell fate transitions in response to environmental cues. These cues are mediated at least in part by steroid hormones. Emerging evidence indicates that miRNAs are also components of additional systemic signaling pathways, including insulin, stress, immune, and circadian pathways. Thus, the roles that miRNAs play during development are reflected in their post-developmental functions, where they similarly function to coordinate cell behavior in response to environmental cues. In this review, we summarize current work highlighting the role of miRNAs in systemic signaling pathways in Drosophila melanogaster as a way of synthesizing the underlying roles of miRNAs in both animal developmental transitions and adult physiology.


Asunto(s)
Ritmo Circadiano/fisiología , Drosophila melanogaster/fisiología , Insulina/metabolismo , Proteínas de la Membrana/fisiología , Redes y Vías Metabólicas/fisiología , MicroARNs/metabolismo , Modelos Biológicos , Serina Endopeptidasas/fisiología , Animales , Ecdisona/metabolismo , Hormonas Juveniles/metabolismo , MicroARNs/biosíntesis
18.
Dev Cell ; 23(1): 202-9, 2012 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-22814608

RESUMEN

Many neural lineages display a temporal pattern, but the mechanisms controlling the ordered production of neuronal subtypes remain unclear. Here, we show that Drosophila let-7 and miR-125, cotranscribed from the let-7-Complex (let-7-C) locus, regulate the transcription factor chinmo to control temporal cell fate in the mushroom body (MB) lineage. We find that let-7-C is activated in postmitotic neurons born during the larval-to-pupal transition, when transitions among three MB subtypes occur. Loss or increase of let-7-C delays or accelerates these transitions, respectively, and leads to cell fate transformations. Consistent with our identification of let-7 and miR-125 sites in a recently identified ∼6 kb extension of the chinmo 3' UTR, Chinmo is elevated in let-7-C mutant MBs. In addition, we show that let-7-C acts upstream of chinmo and that let-7-C phenotypes are caused by elevated chinmo. Thus, these heterochronic miRNAs, originally identified in C. elegans, underlie progenitor cell multipotency during the development of diverse bilateria.


Asunto(s)
Proteínas de Drosophila/fisiología , MicroARNs/fisiología , Cuerpos Pedunculados/citología , Cuerpos Pedunculados/fisiología , Proteínas del Tejido Nervioso/fisiología , Neuronas/fisiología , Animales , Linaje de la Célula/fisiología , Células Cultivadas , Proteínas de Drosophila/genética , Drosophila melanogaster , Regulación del Desarrollo de la Expresión Génica/fisiología , Sitios Genéticos , Proteínas del Tejido Nervioso/genética
19.
Curr Opin Genet Dev ; 22(4): 368-73, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22578317

RESUMEN

The lin-4/miR-125 and let-7 microRNAs are at the heart of the heterochronic pathway, which controls temporal cell fate determination during Caenorhabditis elegans development. These small temporal RNAs are clustered along with a third microRNA, miR-100, in the genomes of most animals. Their conserved temporal and neural expression profile suggests a general role in cell fate determination during nervous system differentiation. By triggering consecutive differentiation programs, these microRNAs probably help to determine birth-order dependent temporal identity and thereby contribute to neural stem cell multipotency.


Asunto(s)
MicroARNs/genética , Animales , Evolución Molecular , Regulación del Desarrollo de la Expresión Génica , Humanos , Filogenia , Esteroides/metabolismo
20.
Development ; 139(10): 1788-97, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22510985

RESUMEN

Steroid hormones and their nuclear receptors drive developmental transitions in diverse organisms, including mammals. In this study, we show that the Drosophila steroid hormone 20-hydroxyecdysone (20E) and its nuclear receptor directly activate transcription of the evolutionarily conserved let-7-complex (let-7-C) locus, which encodes the co-transcribed microRNAs miR-100, let-7 and miR-125. These small RNAs post-transcriptionally regulate the expression of target genes, and are required for the remodeling of the Drosophila neuromusculature during the larval-to-adult transition. Deletion of three 20E responsive elements located in the let-7-C locus results in reduced levels of let-7-C microRNAs, leading to neuromuscular and behavioral defects in adults. Given the evolutionary conservation of let-7-C microRNA sequences and temporal expression profiles, these findings indicate that steroid hormone-coupled control of let-7-C microRNAs is part of an ancestral pathway controlling the transition from larval-to-reproductive animal forms.


Asunto(s)
MicroARNs/metabolismo , Receptores de Esteroides/metabolismo , Animales , Northern Blotting , Línea Celular , Drosophila melanogaster , Ecdisterona/farmacología , Ensayo de Cambio de Movilidad Electroforética , MicroARNs/genética , Receptores de Esteroides/genética , Elementos de Respuesta/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...