Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42.685
Filtrar
1.
Nat Commun ; 15(1): 6993, 2024 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-39143098

RESUMEN

RNA interference (RNAi) is a gene-silencing mechanism triggered by the cytosolic entry of double-stranded RNAs (dsRNAs). Many animal cells internalize extracellular dsRNAs via endocytosis for RNAi induction. However, it is not clear how the endocytosed dsRNAs are translocated into the cytosol across the endo/lysosomal membrane. Herein, we show that in Drosophila S2 cells, endocytosed dsRNAs induce lysosomal membrane permeabilization (LMP) that allows cytosolic dsRNA translocation. LMP mediated by dsRNAs requires the lysosomal Cl-/H+ antiporter ClC-b/DmOstm1. In clc-b or dmostm1 knockout S2 cells, extracellular dsRNAs are endocytosed and reach the lysosomes normally but fail to enter the cytosol. Pharmacological induction of LMP restores extracellular dsRNA-directed RNAi in clc-b or dmostm1-knockout cells. Furthermore, clc-b or dmostm1 mutant flies are defective in extracellular dsRNA-directed RNAi and its associated antiviral immunity. Therefore, endocytosed dsRNAs have an intrinsic ability to induce ClC-b/DmOstm1-dependent LMP that allows cytosolic dsRNA translocation for RNAi responses in Drosophila cells.


Asunto(s)
Citosol , Proteínas de Drosophila , Endocitosis , Lisosomas , Interferencia de ARN , ARN Bicatenario , Animales , ARN Bicatenario/metabolismo , Lisosomas/metabolismo , Citosol/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/metabolismo , Drosophila melanogaster/genética , Canales de Cloruro/metabolismo , Canales de Cloruro/genética , Línea Celular , Membranas Intracelulares/metabolismo , Permeabilidad , Drosophila/metabolismo , Drosophila/genética
2.
Sci Rep ; 14(1): 19867, 2024 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-39191909

RESUMEN

Drosophila suzukii lay eggs in soft-skinned, ripening fruits, making this insect a serious threat to berry production. Since its 2008 introduction into North America, growers have used insecticides, such as pyrethroids and spinosads, as the primary approach for D. suzukii management, resulting in development of insecticide resistance in this pest. This study sought to identify the molecular mechanisms conferring insecticide resistance in these populations. We sequenced the transcriptomes of two pyrethroid- and two spinosad-resistant isofemale lines. In both pyrethroid-resistant lines and one spinosad-resistant line, we identified overexpression of metabolic genes that are implicated in resistance in other insect pests. In the other spinosad-resistant line, we observed an overexpression of cuticular genes that have been linked to resistance. Our findings enabled the development of molecular diagnostics that we used to confirm persistence of insecticide resistance in California, U.S.A. To validate these findings, we leveraged D. melanogaster mutants with reduced expression of metabolic or cuticular genes that were found to be upregulated in resistant D. suzukii to demonstrate that these genes are involved in promoting resistance. This study is the first to characterize the molecular mechanisms of insecticide resistance in D. suzukii and provides insights into how current management practices can be optimized.


Asunto(s)
Drosophila , Combinación de Medicamentos , Perfilación de la Expresión Génica , Resistencia a los Insecticidas , Insecticidas , Macrólidos , Piretrinas , Animales , Resistencia a los Insecticidas/genética , Macrólidos/farmacología , Piretrinas/farmacología , Drosophila/genética , Insecticidas/farmacología , Transcriptoma
3.
Nat Commun ; 15(1): 7041, 2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-39147786

RESUMEN

The evolutionary expansion of sensory neuron populations detecting important environmental cues is widespread, but functionally enigmatic. We investigated this phenomenon through comparison of homologous olfactory pathways of Drosophila melanogaster and its close relative Drosophila sechellia, an extreme specialist for Morinda citrifolia noni fruit. D. sechellia has evolved species-specific expansions in select, noni-detecting olfactory sensory neuron (OSN) populations, through multigenic changes. Activation and inhibition of defined proportions of neurons demonstrate that OSN number increases contribute to stronger, more persistent, noni-odour tracking behaviour. These expansions result in increased synaptic connections of sensory neurons with their projection neuron (PN) partners, which are conserved in number between species. Surprisingly, having more OSNs does not lead to greater odour-evoked PN sensitivity or reliability. Rather, pathways with increased sensory pooling exhibit reduced PN adaptation, likely through weakened lateral inhibition. Our work reveals an unexpected functional impact of sensory neuron population expansions to explain ecologically-relevant, species-specific behaviour.


Asunto(s)
Drosophila melanogaster , Odorantes , Neuronas Receptoras Olfatorias , Animales , Neuronas Receptoras Olfatorias/fisiología , Drosophila melanogaster/fisiología , Drosophila/fisiología , Olfato/fisiología , Adaptación Fisiológica , Vías Olfatorias/fisiología , Especificidad de la Especie , Morinda , Femenino , Conducta Animal/fisiología , Evolución Biológica
4.
Arch Insect Biochem Physiol ; 116(4): e22080, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39148444

RESUMEN

Spotted-wing drosophila, Drosophila suzukii (Matsumura), is an invasive vinegar fly that is a major threat to the small fruits industries globally. Insect capa genes encode multiple neuropeptides, including CAPA-periviscerokinin (CAPA-PVK) peptides, that are specifically known to cause diuresis or anti-diuresis in various organisms. Here we identified and characterized a corresponding G protein-coupled receptor (GPCR) of the D. suzukii CAPA-PVK peptides: CAPA receptor (CAPA-R). To better characterize the behavior of D. suzukii CAPA-R, we used insect cell-based functional expression assays to evaluate responses of CAPA-R against D. suzukii CAPA-PVKs, CAPA-PVKs from five species in Insecta, one species from Mollusca, modified CAPA-PVK peptides, and some PRXamide family peptides: pyrokinin (PK), diapause hormone (DH), and ecdysis-triggering hormone (ETH). Functional studies revealed that the D. suzukii CAPA-R is strongly activated by both of its own natural D. suzukii CAPA-PVKs, and interestingly, it was strongly activated by other CAPA-PVK peptides from Frankliniella occidentallis (Thysanoptera), Solenopsis invicta (Hymenoptera), Helicoverpa zea (Lepidoptera) and Plutella xylostella (Lepidoptera). However, D. suzukii CAPA-R was not activated by Mollusca CAPA-PVK or the other PRXamide peptides. Gene expression analyses showed that the CAPA-R was highly expressed in the Malpighian tubules and moderately in hindgut compared to other digestive organs or the rest of body, supporting diuretic/antidiuretic functionality. When compared across life stages of D. suzukii, expression of CAPA-R was approximately 1.5x greater in the third instar than the other stages and minimally detected in the eggs, 4-day old pupae and 3-day old adults. Our results functionally characterized the D. suzukii CAPA-R and a few short peptides were identified as potential biological targets to exploit the CAPA-R for D. suzukii management.


Asunto(s)
Proteínas de Drosophila , Drosophila , Neuropéptidos , Animales , Femenino , Secuencia de Aminoácidos , Drosophila/metabolismo , Drosophila/genética , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Tracto Gastrointestinal/metabolismo , Hormonas de Insectos/metabolismo , Larva/crecimiento & desarrollo , Larva/metabolismo , Larva/genética , Neuropéptidos/metabolismo , Neuropéptidos/genética , Pupa/crecimiento & desarrollo , Pupa/metabolismo , Pupa/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/genética
5.
Development ; 151(16)2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-39140265

RESUMEN

Keaton Schuster completed his PhD in the lab of Rachel Smith-Bolton at the University of Illinois, USA, investigating Drosophila wing disc regeneration before joining Lionel Christiaen's lab at New York University, USA, for his postdoc studying heart regeneration in the chordate tunicate Ciona robusta (formerly Ciona intestinalis type A). Keaton is part of the second cohort of Development's Pathway to Independence Programme fellows and we spoke to him over Teams to learn more about his career to date and his future plans for starting his own group continuing to use emerging model systems to study cardiac regeneration.


Asunto(s)
Ciona intestinalis , Animales , Historia del Siglo XXI , Ciona intestinalis/fisiología , Regeneración/fisiología , Historia del Siglo XX , Biología Evolutiva/historia , Drosophila , Corazón/fisiología
6.
BMC Biol ; 22(1): 174, 2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-39148049

RESUMEN

BACKGROUND: Venoms have repeatedly evolved over 100 occasions throughout the animal tree of life, making them excellent systems for exploring convergent evolutionary novelty. Growing evidence supports that venom evolution is predominantly driven by prey or host-related selection pressures, and the expression patterns of venom glands reflect adaptive evolution. However, it remains elusive whether the evolution of expression patterns in venom glands is likewise a convergent evolution driven by their prey/host species. RESULTS: We utilized parasitoid wasps that had independently adapted to Drosophila hosts as models to investigate the convergent evolution of venom gland transcriptomes in 19 hymenopteran species spanning ~ 200 million years of evolution. Comparative transcriptome analysis reveals that the global expression patterns among the venom glands of Drosophila parasitoid wasps do not achieve higher similarity compared to non-Drosophila parasitoid wasps. Further evolutionary analyses of expression patterns at the single gene, orthogroup, and Gene Ontology (GO) term levels indicate that some orthogroups/GO terms show correlation with the Drosophila parasitoid wasps. However, these groups rarely include genes highly expressed in venom glands or putative venom genes in the Drosophila parasitoid wasps. CONCLUSIONS: Our study suggests that convergent evolution may not play a predominant force shaping gene expression levels in the venom gland of the Drosophila parasitoid wasps, offering novel insights into the co-evolution between venom and prey/host.


Asunto(s)
Evolución Molecular , Transcriptoma , Venenos de Avispas , Avispas , Animales , Avispas/genética , Avispas/fisiología , Venenos de Avispas/genética , Drosophila/genética , Drosophila/parasitología , Interacciones Huésped-Parásitos/genética , Evolución Biológica
7.
PLoS One ; 19(8): e0308652, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39150918

RESUMEN

The fruit fly Drosophila is a major discovery platform in the biology of ageing due to its balance of relatively short lifespan and relatively complex physiology and behaviour. Previous studies have suggested that some important phenotypes of ageing, for instance increasingly fragmented sleep, are shared from humans to Drosophila and can be useful measures of behavioural change with age: these phenotypes therefore hold potential as readouts of healthy ageing for genetic or pharmacological interventions aimed at the underpinning biology of ageing. However, some age-related phenotypes in Drosophila show differing results among studies, leading to questions regarding the source of discrepancies among experiments. In this study, I have tested females and males from three common laboratory strains of Drosophila to determine the extent to which sex and background strain influence age-related behavioural changes in sleep and activity patterns. Surprisingly, I find that some phenotypes-including age-related changes in total activity, total sleep, and sleep fragmentation-depend strongly on sex and strain, to the extent that some phenotypes show opposing age-related changes in different sexes or strains. Conversely, I identify other phenotypes, including age-related decreases in morning and evening anticipation, that are more uniform across sexes and strains. These results reinforce the importance of controlling for background strain in both behavioural and ageing experiments, and they imply that caution should be used when drawing conclusions from studies on a single sex or strain of Drosophila. At the same time, these findings also offer suggestions for behavioural measures that merit further investigation as potentially more consistent phenotypes of ageing.


Asunto(s)
Envejecimiento , Sueño , Animales , Femenino , Masculino , Sueño/fisiología , Envejecimiento/fisiología , Fenotipo , Drosophila/fisiología , Factores Sexuales , Drosophila melanogaster/fisiología , Conducta Animal/fisiología , Actividad Motora/fisiología , Caracteres Sexuales
8.
Sci Rep ; 14(1): 17770, 2024 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-39090271

RESUMEN

Wolbachia is an obligate intracellular α-proteobacterium, which commonly infects arthropods and filarial nematodes. Different strains of Wolbachia are capable of a wide range of regulatory manipulations in their diverse hosts, including the modulation of host cellular differentiation to influence host reproduction. The genetic basis for the majority of these phenotypes is unknown. The wWil strain from the neotropical fruit fly, Drosophila willistoni, exhibits a remarkably high affinity for host germline-derived cells relative to the somatic cells. This trait could be leveraged for understanding how Wolbachia influences the host germline and for controlling host populations in the field. To further the use of this strain in biological and biomedical research, we sequenced the genome of the wWil strain isolated from host cell culture cells. Here, we present the first high quality Nanopore assembly of wWil, the Wolbachia endosymbiont of D. willistoni. Our assembly resulted in a circular genome of 1.27 Mb with a BUSCO completeness score of 99.7%. Consistent with other insect-associated Wolbachia strains, comparative genomic analysis revealed that wWil has a highly mosaic genome relative to the closely related wMel and wAu strains from Drosophila melanogaster and Drosophila simulans, respectively.


Asunto(s)
Drosophila , Genoma Bacteriano , Simbiosis , Wolbachia , Wolbachia/genética , Animales , Drosophila/microbiología , Drosophila/genética , Simbiosis/genética , Filogenia , Secuenciación Completa del Genoma/métodos , Genómica/métodos
9.
BMC Genomics ; 25(1): 766, 2024 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-39107687

RESUMEN

BACKGROUND: Many common diseases exhibit uncontrolled mTOR signaling, prompting considerable interest in the therapeutic potential of mTOR inhibitors, such as rapamycin, to treat a range of conditions, including cancer, aging-related pathologies, and neurological disorders. Despite encouraging preclinical results, the success of mTOR interventions in the clinic has been limited by off-target side effects and dose-limiting toxicities. Improving clinical efficacy and mitigating side effects require a better understanding of the influence of key clinical factors, such as sex, tissue, and genomic background, on the outcomes of mTOR-targeting therapies. RESULTS: We assayed gene expression with and without rapamycin exposure across three distinct body parts (head, thorax, abdomen) of D. melanogaster flies, bearing either their native melanogaster mitochondrial genome or the mitochondrial genome from a related species, D. simulans. The fully factorial RNA-seq study design revealed a large number of genes that responded to the rapamycin treatment in a sex-dependent and tissue-dependent manner, and relatively few genes with the transcriptional response to rapamycin affected by the mitochondrial background. Reanalysis of an earlier study confirmed that mitochondria can have a temporal influence on rapamycin response. CONCLUSIONS: We found significant and wide-ranging effects of sex and body part, alongside a subtle, potentially time-dependent, influence of mitochondria on the transcriptional response to rapamycin. Our findings suggest a number of pathways that could be crucial for predicting potential side effects of mTOR inhibition in a particular sex or tissue. Further studies of the temporal response to rapamycin are necessary to elucidate the effects of the mitochondrial background on mTOR and its inhibition.


Asunto(s)
Mitocondrias , Sirolimus , Animales , Sirolimus/farmacología , Femenino , Masculino , Mitocondrias/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/genética , Drosophila melanogaster/genética , Drosophila melanogaster/efectos de los fármacos , Factores Sexuales , Serina-Treonina Quinasas TOR/metabolismo , Especificidad de Órganos/genética , Drosophila/genética , Drosophila/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Perfilación de la Expresión Génica
10.
Elife ; 132024 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-39110491

RESUMEN

The chromosomes in multicellular eukaryotes are organized into a series of topologically independent loops called TADs. In flies, TADs are formed by physical interactions between neighboring boundaries. Fly boundaries exhibit distinct partner preferences, and pairing interactions between boundaries are typically orientation-dependent. Pairing can be head-to-tail or head-to-head. The former generates a stem-loop TAD, while the latter gives a circle-loop TAD. The TAD that encompasses the Drosophila even skipped (eve) gene is formed by the head-to-tail pairing of the nhomie and homie boundaries. To explore the relationship between loop topology and the physical and regulatory landscape, we flanked the nhomie boundary region with two attP sites. The attP sites were then used to generate four boundary replacements: λ DNA, nhomie forward (WT orientation), nhomie reverse (opposite of WT orientation), and homie forward (same orientation as WT homie). The nhomie forward replacement restores the WT physical and regulatory landscape: in MicroC experiments, the eve TAD is a 'volcano' triangle topped by a plume, and the eve gene and its regulatory elements are sequestered from interactions with neighbors. The λ DNA replacement lacks boundary function: the endpoint of the 'new' eve TAD on the nhomie side is ill-defined, and eve stripe enhancers activate a nearby gene, eIF3j. While nhomie reverse and homie forward restore the eve TAD, the topology is a circle-loop, and this changes the local physical and regulatory landscape. In MicroC experiments, the eve TAD interacts with its neighbors, and the plume at the top of the eve triangle peak is converted to a pair of 'clouds' of contacts with the next-door TADs. Consistent with the loss of isolation afforded by the stem-loop topology, the eve enhancers weakly activate genes in the neighboring TADs. Conversely, eve function is partially disrupted.


Asunto(s)
Proteínas de Drosophila , Animales , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Proteínas de Homeodominio/metabolismo , Proteínas de Homeodominio/genética , Drosophila melanogaster/genética , Drosophila/genética
11.
Elife ; 132024 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-39110499

RESUMEN

Two different models have been proposed to explain how the endpoints of chromatin looped domains ('TADs') in eukaryotic chromosomes are determined. In the first, a cohesin complex extrudes a loop until it encounters a boundary element roadblock, generating a stem-loop. In this model, boundaries are functionally autonomous: they have an intrinsic ability to halt the movement of incoming cohesin complexes that is independent of the properties of neighboring boundaries. In the second, loops are generated by boundary:boundary pairing. In this model, boundaries are functionally non-autonomous, and their ability to form a loop depends upon how well they match with their neighbors. Moreover, unlike the loop-extrusion model, pairing interactions can generate both stem-loops and circle-loops. We have used a combination of MicroC to analyze how TADs are organized, and experimental manipulations of the even skipped TAD boundary, homie, to test the predictions of the 'loop-extrusion' and the 'boundary-pairing' models. Our findings are incompatible with the loop-extrusion model, and instead suggest that the endpoints of TADs in flies are determined by a mechanism in which boundary elements physically pair with their partners, either head-to-head or head-to-tail, with varying degrees of specificity. Although our experiments do not address how partners find each other, the mechanism is unlikely to require loop extrusion.


Asunto(s)
Drosophila , Animales , Drosophila/genética , Drosophila melanogaster/genética , Cromatina/química , Cromatina/metabolismo , Cohesinas , Proteínas Cromosómicas no Histona/metabolismo , Proteínas Cromosómicas no Histona/química , Proteínas Cromosómicas no Histona/genética , Estructuras Cromosómicas , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/química , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/química
12.
Anal Chem ; 96(32): 13007-13014, 2024 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-39101551

RESUMEN

Histone methylation is a key epigenetic modification that regulates the chromatin structure and gene expression for proper cellular and physiological processes. Aberrant histone methylation patterns are implicated in many diseases. Therefore, monitoring histone methylation dynamics in living cells and species is essential for elucidating its regulatory mechanisms and identifying potential therapeutic targets. However, current methods for detecting histone methylation are limited by their low sensitivity and specificity. To overcome this challenge, we have developed a genetically encoded biosensor named Phaser-Trim (Phase separation based genetically encoded reporter for H3K9 Trimethylation) to detect the dynamic changes of H3K9me3 in living cells and species through the generation and disappearance of phase-separated droplets. Phaser-Trim demonstrates advantages of clear phenotypic characteristics, convenient operation, quantitative accuracy, biocompatibility, high specificity, and superior imaging performance with high signal-to-background ratio (SBR) for in vivo animal imaging. Using Phaser-Trim, we have successfully detected the dynamics of the H3K9me3 level during the differentiation of neural stem cells in Drosophila. Furthermore, Phaser-Trim also holds promise for application in high-throughput screening systems to facilitate the discovery of novel anticancer drugs.


Asunto(s)
Histonas , Histonas/metabolismo , Histonas/química , Animales , Metilación , Humanos , Técnicas Biosensibles/métodos , Células-Madre Neurales/metabolismo , Células-Madre Neurales/citología , Drosophila , Diferenciación Celular , Separación de Fases
13.
Sci Rep ; 14(1): 19540, 2024 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-39174694

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a devastating paralytic disorder caused by the death of motoneurons. Several mutations in the KIF5A gene have been identified in patients with ALS. Some mutations affect the splicing sites of exon 27 leading to its deletion (Δ27 mutation). KIF5A Δ27 is aggregation-prone and pathogenic for motoneurons due to a toxic gain of function. Another mutation found to be enriched in ALS patients is a proline/leucine substitution at position 986 (P986L mutation). Bioinformatic analyses strongly suggest that this variant is benign. Our study aims to conduct functional studies in Drosophila to classify the KIF5A P986L variant. When expressed in motoneurons, KIF5A P986L does not modify the morphology of larval NMJ or the synaptic transmission. In addition, KIF5A P986L is uniformly distributed in axons and does not disturb mitochondria distribution. Locomotion at larval and adult stages is not affected by KIF5A P986L. Finally, both KIF5A WT and P986L expression in adult motoneurons extend median lifespan compared to control flies. Altogether, our data show that the KIF5A P986L variant is not pathogenic for motoneurons and may represent a hypomorphic allele, although it is not causative for ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral , Cinesinas , Neuronas Motoras , Animales , Cinesinas/genética , Cinesinas/metabolismo , Neuronas Motoras/metabolismo , Neuronas Motoras/patología , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/patología , Esclerosis Amiotrófica Lateral/metabolismo , Mutación , Humanos , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila , Unión Neuromuscular/metabolismo , Unión Neuromuscular/patología , Drosophila melanogaster/genética , Transmisión Sináptica/genética , Modelos Animales de Enfermedad , Axones/metabolismo , Axones/patología , Larva/genética , Larva/metabolismo
14.
Elife ; 122024 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-39177028

RESUMEN

Autosomal dominant optic atrophy (DOA) is a progressive form of blindness caused by degeneration of retinal ganglion cells and their axons, mainly caused by mutations in the OPA1 mitochondrial dynamin like GTPase (OPA1) gene. OPA1 encodes a dynamin-like GTPase present in the mitochondrial inner membrane. When associated with OPA1 mutations, DOA can present not only ocular symptoms but also multi-organ symptoms (DOA plus). DOA plus often results from point mutations in the GTPase domain, which are assumed to have dominant-negative effects. However, the presence of mutations in the GTPase domain does not always result in DOA plus. Therefore, an experimental system to distinguish between DOA and DOA plus is needed. In this study, we found that loss-of-function mutations of the dOPA1 gene in Drosophila can imitate the pathology of optic nerve degeneration observed in DOA. We successfully rescued this degeneration by expressing the human OPA1 (hOPA1) gene, indicating that hOPA1 is functionally interchangeable with dOPA1 in the fly system. However, mutations previously identified did not ameliorate the dOPA1 deficiency phenotype. By expressing both WT and DOA plus mutant hOPA1 forms in the optic nerve of dOPA1 mutants, we observed that DOA plus mutations suppressed the rescue, facilitating the distinction between loss-of-function and dominant-negative mutations in hOPA1. This fly model aids in distinguishing DOA from DOA plus and guides initial hOPA1 mutation treatment strategies.


Asunto(s)
Modelos Animales de Enfermedad , Proteínas de Drosophila , GTP Fosfohidrolasas , Atrofia Óptica Autosómica Dominante , Animales , Atrofia Óptica Autosómica Dominante/genética , Atrofia Óptica Autosómica Dominante/metabolismo , Atrofia Óptica Autosómica Dominante/patología , GTP Fosfohidrolasas/genética , GTP Fosfohidrolasas/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Humanos , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Mutación , Drosophila/genética , Proteínas de la Membrana
15.
Mol Biol Evol ; 41(8)2024 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-39117360

RESUMEN

Understanding the evolutionary potential of mutations in gene regulatory networks is essential to furthering the study of evolution and development. However, in multicellular systems, genetic manipulation of regulatory networks in a targeted and high-throughput way remains challenging. In this study, we designed TF-High-Evolutionary (HighEvo), a transcription factor (TF) fused with a base editor (activation-induced deaminase), to continuously induce germline mutations at TF-binding sites across regulatory networks in Drosophila. Populations of flies expressing TF-HighEvo in their germlines accumulated mutations at rates an order of magnitude higher than natural populations. Importantly, these mutations accumulated around the targeted TF-binding sites across the genome, leading to distinct morphological phenotypes consistent with the developmental roles of the tagged TFs. As such, this TF-HighEvo method allows the interrogation of the mutational space of gene regulatory networks at scale and can serve as a powerful reagent for experimental evolution and genetic screens focused on the regulatory genome.


Asunto(s)
Redes Reguladoras de Genes , Factores de Transcripción , Animales , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Genoma de los Insectos , Mutagénesis , Drosophila/genética , Evolución Molecular , Drosophila melanogaster/genética
16.
Sci Adv ; 10(32): eadp0860, 2024 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-39121221

RESUMEN

How complex 3D tissue shape emerges during animal development remains an important open question in biology and biophysics. Here, we discover a mechanism for 3D epithelial shape change based on active, in-plane cellular events that is analogous to inanimate "shape programmable" materials, which undergo blueprinted 3D shape transformations from in-plane gradients of spontaneous strains. We study eversion of the Drosophila wing disc pouch, when the epithelium transforms from a dome into a curved fold, quantifying 3D tissue shape changes and mapping spatial patterns of cellular behaviors on the evolving geometry using cellular topology. Using a physical model inspired by shape programming, we find that active cell rearrangements are the major contributor to pouch eversion and validate this conclusion using a knockdown of MyoVI, which reduces rearrangements and disrupts morphogenesis. This work shows that shape programming is a mechanism for animal tissue morphogenesis and suggests that patterns in nature could present design strategies for shape-programmable materials.


Asunto(s)
Morfogénesis , Alas de Animales , Animales , Alas de Animales/crecimiento & desarrollo , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/crecimiento & desarrollo , Drosophila , Modelos Biológicos , Discos Imaginales/metabolismo , Discos Imaginales/crecimiento & desarrollo
17.
Proc Natl Acad Sci U S A ; 121(36): e2411846121, 2024 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-39190348

RESUMEN

Optogenetic techniques provide genetically targeted, spatially and temporally precise approaches to correlate cellular activities and physiological outcomes. In the nervous system, G protein-coupled receptors (GPCRs) have essential neuromodulatory functions through binding extracellular ligands to induce intracellular signaling cascades. In this work, we develop and validate an optogenetic tool that disrupts Gαq signaling through membrane recruitment of a minimal regulator of G protein signaling (RGS) domain. This approach, Photo-induced Gα Modulator-Inhibition of Gαq (PiGM-Iq), exhibited potent and selective inhibition of Gαq signaling. Using PiGM-Iq we alter the behavior of Caenorhabditis elegans and Drosophila with outcomes consistent with GPCR-Gαq disruption. PiGM-Iq changes axon guidance in cultured dorsal root ganglia neurons in response to serotonin. PiGM-Iq activation leads to developmental deficits in zebrafish embryos and larvae resulting in altered neuronal wiring and behavior. Furthermore, by altering the minimal RGS domain, we show that this approach is amenable to Gαi signaling. Our unique and robust optogenetic Gα inhibiting approaches complement existing neurobiological tools and can be used to investigate the functional effects neuromodulators that signal through GPCR and trimeric G proteins.


Asunto(s)
Caenorhabditis elegans , Subunidades alfa de la Proteína de Unión al GTP Gq-G11 , Optogenética , Proteínas RGS , Transducción de Señal , Pez Cebra , Animales , Optogenética/métodos , Caenorhabditis elegans/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , Proteínas RGS/metabolismo , Proteínas RGS/genética , Pez Cebra/embriología , Neuronas/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/genética , Dominios Proteicos , Ganglios Espinales/metabolismo , Ganglios Espinales/citología , Drosophila/metabolismo
18.
Elife ; 132024 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-39190452

RESUMEN

Host-microbe interactions are virtually bidirectional, but how the host affects their microbiome is poorly understood. Here, we report that the host is a critical modulator to regulate the lifestyle switch and pathogenicity heterogeneity of the opportunistic pathogens Serratia marcescens utilizing the Drosophila and bacterium model system. First, we find that Drosophila larvae efficiently outcompete S. marcescens and typically drive a bacterial switch from pathogenicity to commensalism toward the fly. Furthermore, Drosophila larvae reshape the transcriptomic and metabolic profiles of S. marcescens characterized by a lifestyle switch. More importantly, the host alters pathogenicity and heterogeneity of S. marcescens in the single-cell resolution. Finally, we find that larvae-derived AMPs are required to recapitulate the response of S. marcescens to larvae. Altogether, our findings provide an insight into the pivotal roles of the host in harnessing the life history and heterogeneity of symbiotic bacterial cells, advancing knowledge of the reciprocal relationships between the host and pathogen.


Asunto(s)
Drosophila melanogaster , Interacciones Huésped-Patógeno , Larva , Serratia marcescens , Animales , Serratia marcescens/patogenicidad , Serratia marcescens/genética , Serratia marcescens/fisiología , Larva/microbiología , Drosophila melanogaster/microbiología , Análisis de la Célula Individual , Simbiosis , Drosophila/microbiología , Virulencia/genética
19.
RNA Biol ; 21(1): 11-22, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-39190491

RESUMEN

Fragile X Syndrome (FXS) is the most common inherited form of intellectual disability and is caused by mutations in the gene encoding the Fragile X messenger ribonucleoprotein (FMRP). FMRP is an evolutionarily conserved and neuronally enriched RNA-binding protein (RBP) with functions in RNA editing, RNA transport, and protein translation. Specific target RNAs play critical roles in neurodevelopment, including the regulation of neurite morphogenesis, synaptic plasticity, and cognitive function. The different biological functions of FMRP are modulated by its cooperative interaction with distinct sets of neuronal RNA and protein-binding partners. Here, we focus on interactions between FMRP and components of the microRNA (miRNA) pathway. Using the Drosophila S2 cell model system, we show that the Drosophila ortholog of FMRP (dFMRP) can repress translation when directly tethered to a reporter mRNA. This repression requires the activity of AGO1, GW182, and MOV10/Armitage, conserved proteins associated with the miRNA-containing RNA-induced silencing complex (miRISC). Additionally, we find that untagged dFMRP can interact with a short stem-loop sequence in the translational reporter, a prerequisite for repression by exogenous miR-958. Finally, we demonstrate that dFmr1 interacts genetically with GW182 to control neurite morphogenesis. These data suggest that dFMRP may recruit the miRISC to nearby miRNA binding sites and repress translation via its cooperative interactions with evolutionarily conserved components of the miRNA pathway.


Asunto(s)
Proteínas de Drosophila , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil , MicroARNs , Neuritas , Biosíntesis de Proteínas , Animales , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , MicroARNs/genética , MicroARNs/metabolismo , Neuritas/metabolismo , Morfogénesis/genética , Complejo Silenciador Inducido por ARN/metabolismo , Complejo Silenciador Inducido por ARN/genética , Drosophila/metabolismo , Drosophila/genética , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , Drosophila melanogaster/metabolismo , Drosophila melanogaster/genética , Unión Proteica
20.
Development ; 151(16)2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-39190553

RESUMEN

The size of subcellular structures must be tightly controlled to maintain normal cell function. Despite its importance, few studies have determined how the size of organelles or other structures is maintained during development, when cells are growing, dividing and rearranging. The developing Drosophila egg chamber is a powerful model in which to study the relative growth rates of subcellular structures. The egg chamber contains a cluster of 16 germline cells, which are connected through intercellular bridges called ring canals. As the egg chamber grows, the germline cells and the ring canals that connect them increase in size. Here, we demonstrate that ring canal size scaling is related to lineage; the largest, 'first-born' ring canals increase in size at a relatively slower rate than ring canals derived from subsequent mitotic divisions. This lineage-based scaling relationship is maintained even if directed transport is reduced, ring canal size is altered, or in egg chambers with twice as many germline cells. Analysis of lines that produce larger or smaller mature eggs reveals that different strategies could be used to alter final egg size.


Asunto(s)
Linaje de la Célula , Células Germinativas , Oogénesis , Animales , Oogénesis/fisiología , Femenino , Células Germinativas/citología , Drosophila melanogaster , Drosophila , Óvulo/citología , Tamaño de la Célula
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...