Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 52.209
Filtrer
1.
Fly (Austin) ; 18(1): 2398300, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-39239739

RÉSUMÉ

Talaromycosis, caused by Talaromyces marneffei (T. marneffei, formerly known as Penicillium marneffei), is an opportunistic invasive mycosis endemic in tropical and subtropical areas of Asia with high mortality rate. Despite various infection models established to study the immunological interaction between T. marneffei and the host, the pathogenicity of this fungus is not yet fully understood. So far, Drosophila melanogaster, a well-established genetic model organism to study innate immunity, has not been used in related research on T. marneffei. In this study, we provide the initial characterization of a systemic infection model of T. marneffei in the D. melanogaster host. Survival curves and fungal loads were tested as well as Toll pathway activation was quantified by RT-qPCR of several antimicrobial peptide (AMP) genes including Drosomycin, Metchnikowin, and Bomanin Short 1. We discovered that whereas most wild-type flies were able to overcome the infection, MyD88 or Toll mutant flies failed to prevent fungal dissemination and proliferation and ultimately succumbed to this challenge. Unexpectedly, the induction of classical Toll pathway activation readouts, Drosomycin and Bomanin Short 1, by live or killed T. marneffei was quite limited in wild-type flies, suggesting that the fungus largely escapes detection by the systemic immune system. This unusual situation of a poor systemic activation of the Toll pathway and a strong susceptibility phenotype of MyD88/Toll might be accounted for by a requirement for this host defence in only specific tissues, a hypothesis that remains to be rigorously tested.


Sujet(s)
Protéines de Drosophila , Drosophila melanogaster , Facteur de différenciation myéloïde-88 , Talaromyces , Récepteurs de type Toll , Animaux , Talaromyces/génétique , Talaromyces/métabolisme , Facteur de différenciation myéloïde-88/métabolisme , Facteur de différenciation myéloïde-88/génétique , Drosophila melanogaster/microbiologie , Drosophila melanogaster/immunologie , Protéines de Drosophila/métabolisme , Protéines de Drosophila/génétique , Récepteurs de type Toll/métabolisme , Récepteurs de type Toll/génétique , Mycoses/immunologie , Mycoses/microbiologie , Immunité innée , Transduction du signal , Antigènes de différenciation , Récepteurs immunologiques , Protéines adaptatrices de la transduction du signal
2.
PLoS Biol ; 22(9): e3002783, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39226305

RÉSUMÉ

Cell shape remodeling is a principal driver of epithelial tissue morphogenesis. While progress continues to be made in our understanding of the pathways that control the apical (top) geometry of epithelial cells, we know comparatively little about those that control cell basal (bottom) geometry. To examine this, we used the Drosophila ommatidium, which is the basic visual unit of the compound eye. The ommatidium is shaped as a hexagonal prism, and generating this 3D structure requires ommatidial cells to adopt specific apical and basal polygonal geometries. Using this model system, we find that generating cell type-specific basal geometries starts with patterning of the basal extracellular matrix, whereby Laminin accumulates at discrete locations across the basal surface of the retina. We find the Dystroglycan receptor complex (DGC) is required for this patterning by promoting localized Laminin accumulation at the basal surface of cells. Moreover, our results reveal that localized accumulation of Laminin and the DGC are required for directing Integrin adhesion. This induces cell basal geometry remodeling by anchoring the basal surface of cells to the extracellular matrix at specific, Laminin-rich locations. We propose that patterning of a basal extracellular matrix by generating discrete Laminin domains can direct Integrin adhesion to induce cell shape remodeling in epithelial morphogenesis.


Sujet(s)
Forme de la cellule , Protéines de Drosophila , Drosophila melanogaster , Dystroglycanes , Matrice extracellulaire , Intégrines , Laminine , Rétine , Animaux , Dystroglycanes/métabolisme , Laminine/métabolisme , Intégrines/métabolisme , Protéines de Drosophila/métabolisme , Protéines de Drosophila/génétique , Matrice extracellulaire/métabolisme , Rétine/métabolisme , Rétine/croissance et développement , Rétine/cytologie , Rétine/embryologie , Drosophila melanogaster/métabolisme , Drosophila melanogaster/croissance et développement , Drosophila melanogaster/génétique , Morphogenèse , Adhérence cellulaire , Drosophila/métabolisme , Drosophila/croissance et développement
3.
PLoS Genet ; 20(9): e1011387, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39226333

RÉSUMÉ

A programmed developmental switch to G / S endocycles results in tissue growth through an increase in cell size. Unscheduled, induced endocycling cells (iECs) promote wound healing but also contribute to cancer. Much remains unknown, however, about how these iECs affect tissue growth. Using the D. melanogaster wing disc as model, we find that populations of iECs initially increase in size but then subsequently undergo a heterogenous arrest that causes severe tissue undergrowth. iECs acquired DNA damage and activated a Jun N-terminal kinase (JNK) pathway, but, unlike other stressed cells, were apoptosis-resistant and not eliminated from the epithelium. Instead, iECs entered a JNK-dependent and reversible senescent-like arrest. Senescent iECs promoted division of diploid neighbors, but this compensatory proliferation did not rescue tissue growth. Our study has uncovered unique attributes of iECs and their effects on tissue growth that have important implications for understanding their roles in wound healing and cancer.


Sujet(s)
Altération de l'ADN , Drosophila melanogaster , Ailes d'animaux , Animaux , Ailes d'animaux/croissance et développement , Ailes d'animaux/métabolisme , Drosophila melanogaster/croissance et développement , Drosophila melanogaster/génétique , Prolifération cellulaire , Protéines de Drosophila/génétique , Protéines de Drosophila/métabolisme , Apoptose , Disques imaginaux/croissance et développement , Disques imaginaux/métabolisme , Cicatrisation de plaie/génétique , Vieillissement de la cellule , Système de signalisation des MAP kinases , JNK Mitogen-Activated Protein Kinases/métabolisme , JNK Mitogen-Activated Protein Kinases/génétique , Cycle cellulaire
4.
Nat Commun ; 15(1): 8053, 2024 Sep 14.
Article de Anglais | MEDLINE | ID: mdl-39277611

RÉSUMÉ

CRISPR homing gene drives can suppress pest populations by targeting female fertility genes, converting wild-type alleles into drive alleles in the germline of drive heterozygotes. fsRIDL (female-specific Release of Insects carrying a Dominant Lethal) is a self-limiting population suppression strategy involving continual release of transgenic males carrying female lethal alleles. Here, we propose an improved pest suppression system called "Release of Insects carrying a Dominant-sterile Drive" (RIDD), combining performance characteristics of homing drive and fsRIDL. We construct a split RIDD system in Drosophila melanogaster by creating a 3-gRNA drive disrupting the doublesex female exon. Drive alleles bias their inheritance in males, while drive alleles and resistance alleles formed by end-joining cause dominant female sterility. Weekly releases of RIDD males progressively suppressed and eventually eliminated cage populations. Modeling shows that RIDD is substantially stronger than SIT and fsRIDL. RIDD is also self-limiting, potentially allowing targeted population suppression.


Sujet(s)
Animal génétiquement modifié , Protéines de Drosophila , Drosophila melanogaster , Forçage génétique , Animaux , Femelle , Mâle , Drosophila melanogaster/génétique , Forçage génétique/méthodes , Protéines de Drosophila/génétique , Protéines de Drosophila/métabolisme , Allèles , Systèmes CRISPR-Cas , Gènes dominants , Lutte biologique contre les nuisibles/méthodes , Infertilité/génétique , Infertilité/thérapie , /génétique , Protéines de liaison à l'ADN
5.
Narra J ; 4(2): e898, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39280279

RÉSUMÉ

Enteropathy is a gastrointestinal disorder characterized by inflammation in the small intestine and one of the causes of enteropathy is the side effects of certain drugs, such as non-steroidal anti-inflammatory drugs (NSAIDs). The mechanism of NSAIDs, such as indomethacin, could inhibit prostaglandin synthesis, leading to a decrease in mucus production and small intestine integrity. To test the effects of a drug, it is necessary to undergo preclinical testing using animal models. Commonly used animal models such as mice and rats have several drawbacks including high cost, ethical issues, and long lifespan. Therefore, alternatives such as using invertebrate animals like Drosophila melanogaster as a more economical in vivo platform with genetic similarity to mammals and devoid of ethical concerns are needed. The aim of this study was to evaluate Drosophila melanogaster as an in vivo model organism in testing the side effects of pharmaceuticals that cause enteropathy. In this study, flies aged 3-5 days were starved and then placed into treatment vials comprising untreated control and indomethacin-treated (3.75 mM, 7.5 mM, and 15 mM). Survival analysis was conducted during the treatment period, followed by a Smurf assay test after seven days of treatment. Subsequently, the expression of pro-inflammatory cytokine-related genes (drs and totA), mitochondria stability-related genes (tom40), and endogenous antioxidant-related genes (sod1, sod2, and cat) was performed using reverse transcription-quantitative polymerase chain reaction (RT-qPCR). Our data indicated that indomethacin did not impact lifespan or cause intestinal damage. However, we observed increased expression of pro-inflammatory cytokine-related genes, including drs, and a twofold increase in totA gene expression. Furthermore, there was a significant upregulation of mitochondrial stability gene tom40, endogenous antioxidant genes sod1 and cat, and a threefold increase in sod2 at 15 mM indomethacin. Although no phenotypical changes in gut integrity were detected, the increased expression of pro-inflammatory cytokine genes suggests the occurrence of inflammation in the indomethacin-treated flies.


Sujet(s)
Anti-inflammatoires non stéroïdiens , Drosophila melanogaster , Indométacine , Maladies intestinales , Animaux , Drosophila melanogaster/effets des médicaments et des substances chimiques , Indométacine/effets indésirables , Indométacine/pharmacologie , Anti-inflammatoires non stéroïdiens/pharmacologie , Anti-inflammatoires non stéroïdiens/effets indésirables , Maladies intestinales/induit chimiquement , Maladies intestinales/anatomopathologie , Maladies intestinales/génétique , Maladies intestinales/traitement médicamenteux , Modèles animaux de maladie humaine , Superoxide dismutase/génétique , Superoxide dismutase/métabolisme
6.
Narra J ; 4(2): e818, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39280322

RÉSUMÉ

Drug repurposing is a promising approach to identify new pharmacological indications for drugs that have already been established. However, there is still a limitation in the availability of a high-throughput in vivo preclinical system that is suitable for screening and investigating new pharmacological indications. The aim of this study was to introduce the application of Drosophila larvae as an in vivo platform to screen drug candidates with anti-aging and immunomodulatory activities. To determine whether Drosophila larvae can be utilized for assessing anti-aging and immunomodulatory activities, phenotypical and molecular assays were conducted using wildtype and mutant lines of Drosophila. The utilization of mutant lines (PGRP-LBΔ and Psh[1];;ModSP[KO]) mimics the autoinflammatory and immunodeficient conditions in humans, thereby enabling a thorough investigation of the effects of various compounds. The phenotypical assay was carried out using survival and locomotor observation in Drosophila larvae and adult flies. Meanwhile, the molecular assay was conducted using the RT-qPCR method. In vivo survival analysis revealed that caffeine was relatively safe for Drosophila larvae and exhibited the ability to extend Drosophila lifespan compared to the untreated controls, suggesting its anti-aging properties. Further analysis using the RT-qPCR method demonstrated that caffeine treatment induced transcriptional changes in the Drosophila larvae, particularly in the downstream of NF-κB and JAK-STAT pathways, two distinct immune-related pathways homologue to humans. In addition, caffeine enhanced the survival of Drosophila autoinflammatory model, further implying its immunosuppressive activity. Nevertheless, this compound had minimal to no effect on the survival of Staphylococcus aureus-infected wildtype and immunodeficient Drosophila, refuting its antibacterial and immunostimulant activities. Overall, our results suggest that the anti-aging and immunosuppressive activities of caffeine observed in Drosophila larvae align with those reported in mammalian model systems, emphasizing the suitability of Drosophila larvae as a model organism in drug repurposing endeavors, particularly for the screening of newly discovered chemical entities to assess their immunomodulatory activities before proceedings to investigations in mammalian animal models.


Sujet(s)
Vieillissement , Caféine , Larve , Animaux , Larve/effets des médicaments et des substances chimiques , Larve/immunologie , Caféine/pharmacologie , Vieillissement/effets des médicaments et des substances chimiques , Vieillissement/immunologie , Drosophila/effets des médicaments et des substances chimiques , Drosophila melanogaster/effets des médicaments et des substances chimiques , Drosophila melanogaster/immunologie , Drosophila melanogaster/microbiologie
7.
Elife ; 132024 Sep 18.
Article de Anglais | MEDLINE | ID: mdl-39291827

RÉSUMÉ

Differentiation of female germline stem cells into a mature oocyte includes the expression of RNAs and proteins that drive early embryonic development in Drosophila. We have little insight into what activates the expression of these maternal factors. One candidate is the zinc-finger protein OVO. OVO is required for female germline viability and has been shown to positively regulate its own expression, as well as a downstream target, ovarian tumor, by binding to the transcriptional start site (TSS). To find additional OVO targets in the female germline and further elucidate OVO's role in oocyte development, we performed ChIP-seq to determine genome-wide OVO occupancy, as well as RNA-seq comparing hypomorphic and wild type rescue ovo alleles. OVO preferentially binds in close proximity to target TSSs genome-wide, is associated with open chromatin, transcriptionally active histone marks, and OVO-dependent expression. Motif enrichment analysis on OVO ChIP peaks identified a 5'-TAACNGT-3' OVO DNA binding motif spatially enriched near TSSs. However, the OVO DNA binding motif does not exhibit precise motif spacing relative to the TSS characteristic of RNA polymerase II complex binding core promoter elements. Integrated genomics analysis showed that 525 genes that are bound and increase in expression downstream of OVO are known to be essential maternally expressed genes. These include genes involved in anterior/posterior/germ plasm specification (bcd, exu, swa, osk, nos, aub, pgc, gcl), egg activation (png, plu, gnu, wisp, C(3)g, mtrm), translational regulation (cup, orb, bru1, me31B), and vitelline membrane formation (fs(1)N, fs(1)M3, clos). This suggests that OVO is a master transcriptional regulator of oocyte development and is responsible for the expression of structural components of the egg as well as maternally provided RNAs that are required for early embryonic development.


Sujet(s)
Protéines de Drosophila , Drosophila melanogaster , Site d'initiation de la transcription , Animaux , Femelle , Protéines de Drosophila/métabolisme , Protéines de Drosophila/génétique , Drosophila melanogaster/génétique , Drosophila melanogaster/métabolisme , Drosophila melanogaster/embryologie , Régulation de l'expression des gènes au cours du développement , Ovocytes/métabolisme , Protéines de liaison à l'ADN , Facteurs de transcription
8.
Elife ; 122024 Sep 18.
Article de Anglais | MEDLINE | ID: mdl-39291956

RÉSUMÉ

Synaptic heterogeneity is a hallmark of nervous systems that enables complex and adaptable communication in neural circuits. To understand circuit function, it is thus critical to determine the factors that contribute to the functional diversity of synapses. We investigated the contributions of voltage-gated calcium channel (VGCC) abundance, spatial organization, and subunit composition to synapse diversity among and between synapses formed by two closely related Drosophila glutamatergic motor neurons with distinct neurotransmitter release probabilities (Pr). Surprisingly, VGCC levels are highly predictive of heterogeneous Pr among individual synapses of either low- or high-Pr inputs, but not between inputs. We find that the same number of VGCCs are more densely organized at high-Pr synapses, consistent with tighter VGCC-synaptic vesicle coupling. We generated endogenously tagged lines to investigate VGCC subunits in vivo and found that the α2δ-3 subunit Straightjacket along with the CAST/ELKS active zone (AZ) protein Bruchpilot, both key regulators of VGCCs, are less abundant at high-Pr inputs, yet positively correlate with Pr among synapses formed by either input. Consistently, both Straightjacket and Bruchpilot levels are dynamically increased across AZs of both inputs when neurotransmitter release is potentiated to maintain stable communication following glutamate receptor inhibition. Together, these findings suggest a model in which VGCC and AZ protein abundance intersects with input-specific spatial and molecular organization to shape the functional diversity of synapses.


Sujet(s)
Canaux calciques , Protéines de Drosophila , Synapses , Animaux , Synapses/métabolisme , Synapses/physiologie , Protéines de Drosophila/métabolisme , Protéines de Drosophila/génétique , Canaux calciques/métabolisme , Motoneurones/métabolisme , Motoneurones/physiologie , Drosophila/physiologie , Drosophila melanogaster/métabolisme , Transmission synaptique/physiologie
9.
Proc Natl Acad Sci U S A ; 121(39): e2319666121, 2024 Sep 24.
Article de Anglais | MEDLINE | ID: mdl-39288176

RÉSUMÉ

Mammalian Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) and Drosophila Yorkie (Yki) are transcription cofactors of the highly conserved Hippo signaling pathway. It has been long assumed that the YAP/TAZ/Yki signaling drives cell proliferation during organ growth. However, its instructive role in regulating developmentally programmed organ growth, if any, remains elusive. Out-of-context gain of YAP/TAZ/Yki signaling often turns oncogenic. Paradoxically, mechanically strained, and differentiated squamous epithelia display developmentally programmed constitutive nuclear YAP/TAZ/Yki signaling. The unknown, therefore, is how a growth-promoting YAP/TAZ/Yki signaling restricts proliferation in differentiated squamous epithelia. Here, we show that reminiscent of a tumor suppressor, Yki negatively regulates the cell growth-promoting PI3K/Akt/TOR signaling in the squamous epithelia of Drosophila tubular organs. Thus, downregulation of Yki signaling in the squamous epithelium of the adult male accessory gland (MAG) up-regulates PI3K/Akt/TOR signaling, inducing cell hypertrophy, exit from their cell cycle arrest, and, finally, culminating in squamous cell carcinoma (SCC). Thus, blocking PI3K/Akt/TOR signaling arrests Yki loss-induced MAG-SCC. Further, MAG-SCCs, like other lethal carcinomas, secrete a cachectin, Impl2-the Drosophila homolog of mammalian IGFBP7-inducing cachexia and shortening the lifespan of adult males. Moreover, in the squamous epithelium of other tubular organs, like the dorsal trunk of larval tracheal airways or adult Malpighian tubules, downregulation of Yki signaling triggers PI3K/Akt/TOR-induced cell hypertrophy. Our results reveal that Yki signaling plays an instructive, antiproliferative role in the squamous epithelia of tubular organs.


Sujet(s)
Protéines de Drosophila , Drosophila melanogaster , Protéines nucléaires , Protein-Serine-Threonine Kinases , Transduction du signal , Transactivateurs , Protéines de signalisation YAP , Animaux , Protéines de Drosophila/métabolisme , Protéines de Drosophila/génétique , Protéines de signalisation YAP/métabolisme , Protéines de signalisation YAP/génétique , Transactivateurs/métabolisme , Transactivateurs/génétique , Mâle , Drosophila melanogaster/métabolisme , Protéines nucléaires/métabolisme , Protéines nucléaires/génétique , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique , Épithélium/métabolisme , Prolifération cellulaire , Phosphatidylinositol 3-kinases/métabolisme , Protéines et peptides de signalisation intracellulaire/métabolisme , Protéines et peptides de signalisation intracellulaire/génétique , Protéines proto-oncogènes c-akt/métabolisme , Protéines suppresseurs de tumeurs/métabolisme , Protéines suppresseurs de tumeurs/génétique
10.
Bioinformatics ; 40(Suppl 2): ii128-ii136, 2024 09 01.
Article de Anglais | MEDLINE | ID: mdl-39230697

RÉSUMÉ

SUMMARY: Single-neuron morphology, the study of the structure, form, and shape of a group of specialized cells in the nervous system, is of vital importance to define the type of neurons, assess changes in neuronal development and aging and determine the effects of brain disorders and treatments. Despite the recent surge in the amount of available neuron morphology reconstructions due to advancements in microscopy imaging, existing computational and deep learning methods for modeling neuron morphology have been limited in both scale and accuracy. In this paper, we propose MorphRep, a model for learning meaningful representation of neuron morphology pre-trained with over 250 000 existing neuron morphology data. By encoding the neuron morphology into graph-structured data, using graph transformers for feature encoding and enforcing the consistency between multiple augmented views of neuron morphology, MorphRep achieves the state of the art performance on widely used benchmarking datasets. Meanwhile, MorphRep can accurately characterize the neuron morphology space across neuron morphometrics, fine-grained cell types, brain regions and ages. Furthermore, MorphRep can be applied to distinguish neurons under a wide range of conditions, including genetic perturbation, drug injection, environment change and disease. In summary, MorphRep provides an effective strategy to embed and represent neuron morphology and can be a valuable tool in integrating cell morphology into single-cell multiomics analysis. AVAILABILITY AND IMPLEMENTATION: The codebase has been deposited in https://github.com/YaxuanLi-cn/MorphRep.


Sujet(s)
Apprentissage profond , Neurones , Neurones/cytologie , Humains , Animaux , Rats , Drosophila melanogaster , Forme de la cellule , Jeux de données comme sujet
11.
Molecules ; 29(17)2024 Sep 04.
Article de Anglais | MEDLINE | ID: mdl-39275038

RÉSUMÉ

A nutritional approach could be a promising strategy to prevent or decrease the progression of neurodegenerative disorders such as Parkinson's disease (PD). The neuroprotective role of walnut oil (WO) was investigated in Drosophila melanogaster treated with rotenone (Rot), as a PD model, WO, or their combination, and compared to controls. WO reduced mortality and improved locomotor activity impairment after 3 and 7 days, induced by Rot. LC-MS analyses of fatty acid levels in Drosophila heads showed a significant increase in linolenic (ALA) and linoleic acid (LA) both in flies fed with the WO-enriched diet and in those treated with the association of WO with Rot. Flies supplemented with the WO diet showed an increase in brain dopamine (DA) level, while Rot treatment significantly depleted dopamine content; conversely, the association of Rot with WO did not modify DA content compared to controls. The greater intake of ALA and LA in the enriched diet enhanced their levels in Drosophila brain, suggesting a neuroprotective role of polyunsaturated fatty acids against Rot-induced neurotoxicity. The involvement of the dopaminergic system in the improvement of behavioral and biochemical parameters in Drosophila fed with WO is also suggested.


Sujet(s)
Modèles animaux de maladie humaine , Drosophila melanogaster , Juglans , Maladie de Parkinson , Huiles végétales , Animaux , Drosophila melanogaster/effets des médicaments et des substances chimiques , Juglans/composition chimique , Maladie de Parkinson/traitement médicamenteux , Maladie de Parkinson/métabolisme , Huiles végétales/pharmacologie , Huiles végétales/composition chimique , Dopamine/métabolisme , Roténone , Encéphale/métabolisme , Encéphale/effets des médicaments et des substances chimiques , Neuroprotecteurs/pharmacologie
12.
Development ; 151(18)2024 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-39284714

RÉSUMÉ

The number of neural stem cells reflects the total number of neurons in the mature brain. As neural stem cells arise from neuroepithelial cells, the neuroepithelial cell population must be expanded to secure a sufficient number of neural stem cells. However, molecular mechanisms that regulate timely differentiation from neuroepithelial to neural stem cells are largely unclear. Here, we show that TCF4/Daughterless is a key factor that determines the timing of the differentiation in Drosophila. The neuroepithelial cells initiated but never completed the differentiation in the absence of TCF4/Daughterless. We also found that TCF4/Daughterless binds to the Notch locus, suggesting that Notch is one of its downstream candidate genes. Consistently, Notch expression was ectopically induced in the absence of TCF4/Daughterless. Furthermore, ectopic activation of Notch signaling phenocopied loss of TCF4/Daughterless. Our findings demonstrate that TCF4/Daughterless directly inactivates Notch signaling pathway, resulting in completion of the differentiation from neuroepithelial cells into neural stem cells with optimal timing. Thus, the present results suggest that TCF4/Daughterless is essential for determining whether to move to the next state or stay in the current state in differentiating neuroepithelial cells.


Sujet(s)
Facteurs de transcription à motif basique hélice-boucle-hélice , Différenciation cellulaire , Protéines de Drosophila , Cellules souches neurales , Cellules neuroépithéliales , Récepteurs Notch , Transduction du signal , Animaux , Cellules souches neurales/métabolisme , Cellules souches neurales/cytologie , Récepteurs Notch/métabolisme , Récepteurs Notch/génétique , Protéines de Drosophila/métabolisme , Protéines de Drosophila/génétique , Cellules neuroépithéliales/métabolisme , Cellules neuroépithéliales/cytologie , Facteurs de transcription à motif basique hélice-boucle-hélice/métabolisme , Facteurs de transcription à motif basique hélice-boucle-hélice/génétique , Différenciation cellulaire/génétique , Régulation de l'expression des gènes au cours du développement , Drosophila melanogaster/métabolisme , Drosophila melanogaster/génétique , Drosophila melanogaster/cytologie , Facteurs temps , Drosophila/métabolisme
13.
Sci Rep ; 14(1): 20867, 2024 09 06.
Article de Anglais | MEDLINE | ID: mdl-39242711

RÉSUMÉ

Huntington's disease (HD) is a rare neurodegenerative disease caused due to aggregation of Huntingtin (HTT) protein. This study involves the cloning of 40 DnaJ chaperones from Drosophila, and overexpressing them in yeasts and fly models of HD. Accordingly, DnaJ chaperones were catalogued as enhancers or suppressors based on their growth phenotypes and aggregation properties. 2 of the chaperones that came up as targets were CG5001 and P58IPK. Protein aggregation and slow growth phenotype was rescued in yeasts, S2 cells, and Drosophila transgenic lines of HTT103Q with these overexpressed chaperones. Since DnaJ chaperones have protein sequence similarity across species, they can be used as possible tools to combat the effects of neurodegenerative diseases.


Sujet(s)
Protéines de Drosophila , Protéines du choc thermique HSP40 , Protéine huntingtine , Maladie de Huntington , Animaux , Humains , Animal génétiquement modifié , Modèles animaux de maladie humaine , Drosophila , Drosophila melanogaster , Protéines de Drosophila/métabolisme , Protéines de Drosophila/génétique , Protéines du choc thermique HSP40/génétique , Protéines du choc thermique HSP40/métabolisme , Protéine huntingtine/génétique , Protéine huntingtine/métabolisme , Maladie de Huntington/génétique , Maladie de Huntington/métabolisme , Maladie de Huntington/anatomopathologie , Agrégats de protéines , Agrégation pathologique de protéines/génétique , Saccharomyces cerevisiae
14.
Pestic Biochem Physiol ; 204: 106102, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39277424

RÉSUMÉ

While much attention has been devoted to understanding the transcriptional changes underlying resistance to insecticides, comparatively little is known about the transcriptional response of naive insects to agrochemicals. In this study, we analyze the transcriptomic response of an insecticide susceptible strain of Drosophila melanogaster to nine agrochemicals using a robust method that goes beyond classical replication standards. Our findings demonstrate that exposure to piperonyl butoxide (PBO), but not to eight other compounds, elicits a robust transcriptional response in a wild-type strain of Drosophila melanogaster. PBO exposure leads to the upregulation of a subset of Cyps, GSTs, UGTs and EcKls. This response is both time and concentration-dependent, suggesting that the degree of inhibition of P450 activity correlates with the magnitude of the transcriptional response. Furthermore, the upregulation of these enzymes is excluded from reproductive organs. Additionally, different sets of genes are regulated in the digestive/secretory tract and the carcass. Our results suggest that P450s play a role in metabolizing yet unidentified endogenous compounds and are involved in an as-yet-unknown physiological regulatory feedback loop.


Sujet(s)
Cytochrome P-450 enzyme system , Drosophila melanogaster , Butoxyde de pipéronyle , Animaux , Butoxyde de pipéronyle/pharmacologie , Drosophila melanogaster/effets des médicaments et des substances chimiques , Drosophila melanogaster/génétique , Cytochrome P-450 enzyme system/génétique , Cytochrome P-450 enzyme system/métabolisme , Insecticides/toxicité , Insecticides/pharmacologie , Protéines de Drosophila/génétique , Protéines de Drosophila/métabolisme , Transcription génétique/effets des médicaments et des substances chimiques
15.
Elife ; 122024 Sep 06.
Article de Anglais | MEDLINE | ID: mdl-39240259

RÉSUMÉ

Female sexual receptivity is essential for reproduction of a species. Neuropeptides play the main role in regulating female receptivity. However, whether neuropeptides regulate female sexual receptivity during the neurodevelopment is unknown. Here, we found the peptide hormone prothoracicotropic hormone (PTTH), which belongs to the insect PG (prothoracic gland) axis, negatively regulated virgin female receptivity through ecdysone during neurodevelopment in Drosophila melanogaster. We identified PTTH neurons as doublesex-positive neurons, they regulated virgin female receptivity before the metamorphosis during the third-instar larval stage. PTTH deletion resulted in the increased EcR-A expression in the whole newly formed prepupae. Furthermore, the ecdysone receptor EcR-A in pC1 neurons positively regulated virgin female receptivity during metamorphosis. The decreased EcR-A in pC1 neurons induced abnormal morphological development of pC1 neurons without changing neural activity. Among all subtypes of pC1 neurons, the function of EcR-A in pC1b neurons was necessary for virgin female copulation rate. These suggested that the changes of synaptic connections between pC1b and other neurons decreased female copulation rate. Moreover, female receptivity significantly decreased when the expression of PTTH receptor Torso was reduced in pC1 neurons. This suggested that PTTH not only regulates female receptivity through ecdysone but also through affecting female receptivity associated neurons directly. The PG axis has similar functional strategy as the hypothalamic-pituitary-gonadal axis in mammals to trigger the juvenile-adult transition. Our work suggests a general mechanism underlying which the neurodevelopment during maturation regulates female sexual receptivity.


Sujet(s)
Protéines de Drosophila , Drosophila melanogaster , Hormones des insectes , Neurones , Récepteurs aux stéroïdes , Comportement sexuel chez les animaux , Animaux , Drosophila melanogaster/physiologie , Drosophila melanogaster/croissance et développement , Femelle , Comportement sexuel chez les animaux/physiologie , Protéines de Drosophila/métabolisme , Protéines de Drosophila/génétique , Neurones/physiologie , Neurones/métabolisme , Hormones des insectes/métabolisme , Récepteurs aux stéroïdes/métabolisme , Récepteurs aux stéroïdes/génétique , Ecdysone/métabolisme , Métamorphose biologique/physiologie , Mâle , Larve/croissance et développement , Larve/physiologie , Protéines d'insecte
16.
Nat Commun ; 15(1): 8020, 2024 Sep 13.
Article de Anglais | MEDLINE | ID: mdl-39271704

RÉSUMÉ

Most RNA-protein condensates are composed of heterogeneous immiscible phases. However, how this multiphase organization contributes to their biological functions remains largely unexplored. Drosophila germ granules, a class of RNA-protein condensates, are the site of mRNA storage and translational activation. Here, using super-resolution microscopy and single-molecule imaging approaches, we show that germ granules have a biphasic organization and that translation occurs in the outer phase and at the surface of the granules. The localization, directionality, and compaction of mRNAs within the granule depend on their translation status, translated mRNAs being enriched in the outer phase with their 5'end oriented towards the surface. Translation is strongly reduced when germ granule biphasic organization is lost. These findings reveal the intimate links between the architecture of RNA-protein condensates and the organization of their different functions, highlighting the functional compartmentalization of these condensates.


Sujet(s)
Granulations cytoplasmiques , Protéines de Drosophila , Drosophila melanogaster , Biosynthèse des protéines , ARN messager , Animaux , Protéines de Drosophila/métabolisme , Protéines de Drosophila/génétique , Granulations cytoplasmiques/métabolisme , Drosophila melanogaster/métabolisme , Drosophila melanogaster/génétique , ARN messager/métabolisme , ARN messager/génétique , Cellules germinales/métabolisme , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/génétique , Imagerie de molécules uniques , Drosophila/métabolisme , Drosophila/génétique , Condensats biomoléculaires/métabolisme
17.
Cells ; 13(17)2024 Aug 28.
Article de Anglais | MEDLINE | ID: mdl-39273016

RÉSUMÉ

Super-resolution single-molecule localization microscopy (SMLM) of presynaptic active zones (AZs) and postsynaptic densities contributed to the observation of protein nanoclusters that are involved in defining functional characteristics and in plasticity of synaptic connections. Among SMLM techniques, direct stochastic optical reconstruction microscopy (dSTORM) depends on organic fluorophores that exert high brightness and reliable photoswitching. While multicolor imaging is highly desirable, the requirements necessary for high-quality dSTORM make it challenging to identify combinations of equally performing, spectrally separated dyes. Red-excited carbocyanine dyes, e.g., Alexa Fluor 647 (AF647) or Cy5, are currently regarded as "gold standard" fluorophores for dSTORM imaging. However, a recent study introduced a set of chemically modified rhodamine dyes, including CF583R, that promise to display similar performance in dSTORM. In this study, we defined CF583R's performance compared to AF647 and CF568 based on a nanoscopic analysis of Bruchpilot (Brp), a nanotopologically well-characterized scaffold protein at Drosophila melanogaster AZs. We demonstrate equal suitability of AF647, CF568 and CF583R for basal AZ morphometry, while in Brp subcluster analysis CF583R outperforms CF568 and is on par with AF647. Thus, the AF647/CF583R combination will be useful in future dSTORM-based analyses of AZs and other subcellularly located marker molecules and their role in physiological and pathophysiological contexts.


Sujet(s)
Drosophila melanogaster , Colorants fluorescents , Animaux , Drosophila melanogaster/métabolisme , Colorants fluorescents/composition chimique , Processus stochastiques , Protéines de Drosophila/métabolisme , Microscopie de fluorescence/méthodes , Rhodamines/composition chimique
18.
Cell Mol Life Sci ; 81(1): 396, 2024 Sep 11.
Article de Anglais | MEDLINE | ID: mdl-39261338

RÉSUMÉ

High dietary sugar (HDS), a contemporary dietary concern due to excessive intake of added sugars and carbohydrates, escalates the risk of metabolic disorders and concomitant cancers. However, the molecular mechanisms underlying HDS-induced cancer progression are not completely understood. We found that phosphoenolpyruvate carboxykinase 1 (PEPCK1), a pivotal enzyme in gluconeogenesis, is paradoxically upregulated in tumors by HDS, but not by normal dietary sugar (NDS), during tumor progression. Targeted knockdown of pepck1, but not pepck2, specifically in tumor tissue in Drosophila in vivo, not only attenuates HDS-induced tumor growth but also significantly improves the survival of Ras/Src tumor-bearing animals fed HDS. Interestingly, HP1a-mediated heterochromatin interacts directly with the pepck1 gene and downregulates pepck1 gene expression in wild-type Drosophila. Mechanistically, we demonstrated that, under HDS conditions, pepck1 knockdown reduces both wingless and TOR signaling, decreases evasion of apoptosis, reduces genome instability, and suppresses glucose uptake and trehalose levels in tumor cells in vivo. Moreover, rational pharmacological inhibition of PEPCK1, using hydrazinium sulfate, greatly improves the survival of tumor-bearing animals with pepck1 knockdown under HDS. This study is the first to show that elevated levels of dietary sugar induce aberrant upregulation of PEPCK1, which promotes tumor progression through altered cell signaling, evasion of apoptosis, genome instability, and reprogramming of carbohydrate metabolism. These findings contribute to our understanding of the complex relationship between diet and cancer at the molecular, cellular, and organismal levels and reveal PEPCK1 as a potential target for the prevention and treatment of cancers associated with metabolic disorders.


Sujet(s)
Évolution de la maladie , Protéines de Drosophila , Régulation positive , Animaux , Protéines de Drosophila/métabolisme , Protéines de Drosophila/génétique , Humains , Tumeurs/anatomopathologie , Tumeurs/métabolisme , Tumeurs/génétique , Apoptose/génétique , Transduction du signal , Protéine Wnt1/métabolisme , Protéine Wnt1/génétique , Phosphoenolpyruvate carboxykinase (ATP)/métabolisme , Phosphoenolpyruvate carboxykinase (ATP)/génétique , Glucose/métabolisme , Instabilité du génome , Phosphoenolpyruvate carboxykinase (GTP)/métabolisme , Phosphoenolpyruvate carboxykinase (GTP)/génétique , Lignée cellulaire tumorale , Drosophila melanogaster/métabolisme , Drosophila melanogaster/génétique , Régulation de l'expression des gènes tumoraux , Tréhalose/métabolisme , Hydrates de carbone alimentaires/effets indésirables , Drosophila/métabolisme
19.
Sci Rep ; 14(1): 21182, 2024 09 11.
Article de Anglais | MEDLINE | ID: mdl-39261567

RÉSUMÉ

Acrylamide (ACR) with its extensive industrial applications is a classified occupational hazard toxin and carcinogenic compound. Its formation in fried potatoes, red meat and coffee during high-temperature cooking is a cause for consideration. The fabrication of chitosan-coated probiotic nanoparticles (CSP NPs) aims to enhance the bioavailability of probiotics in the gut, thereby improving their efficacy against ACR-induced toxicity in Drosophila melanogaster. Nanoencapsulation, a vital domain of the medical nanotechnology field plays a key role in targeted drug delivery, bioavailability, multi-drug load delivery systems and synergistic treatment options. Our study exploited the nanoencapsulation technology to coat Lactobacillus fermentum (probiotic) with chitosan (prebiotic), both with substantial immunomodulatory effects, to ensure the stability and sustained release of microbial load and its secondary metabolites in the gut. The combination of pre-and probiotic components, called synbiotic formulations establishes the correlation between the gut microbiota and the overall well-being of an organism. Our study aimed to develop a potent synbiotic to alleviate the impacts of heat-processed dietary toxins that significantly influence behaviour, development, and survival. Our synbiotic co-treatment with ACR in fruit flies normalised neuro-behavioural, survival, redox status, and restored ovarian mitochondrial activity, contrasting with several physiological deficits observed in the ACR-treated model.


Sujet(s)
Acrylamide , Chitosane , Drosophila melanogaster , Limosilactobacillus fermentum , Nanoparticules , Probiotiques , Animaux , Chitosane/composition chimique , Chitosane/pharmacologie , Probiotiques/administration et posologie , Nanoparticules/composition chimique , Acrylamide/composition chimique , Acrylamide/toxicité , Drosophila melanogaster/effets des médicaments et des substances chimiques , Femelle , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques
20.
Elife ; 132024 Sep 10.
Article de Anglais | MEDLINE | ID: mdl-39255019

RÉSUMÉ

Stem cell niche is critical for regulating the behavior of stem cells. Drosophila neural stem cells (Neuroblasts, NBs) are encased by glial niche cells closely, but it still remains unclear whether glial niche cells can regulate the self-renewal and differentiation of NBs. Here, we show that ferritin produced by glia, cooperates with Zip13 to transport iron into NBs for the energy production, which is essential to the self-renewal and proliferation of NBs. The knockdown of glial ferritin encoding genes causes energy shortage in NBs via downregulating aconitase activity and NAD+ level, which leads to the low proliferation and premature differentiation of NBs mediated by Prospero entering nuclei. More importantly, ferritin is a potential target for tumor suppression. In addition, the level of glial ferritin production is affected by the status of NBs, establishing a bicellular iron homeostasis. In this study, we demonstrate that glial cells are indispensable to maintain the self-renewal of NBs, unveiling a novel role of the NB glial niche during brain development.


Iron is an essential nutrient for almost all living organisms. For example, iron contributes to the replication of DNA, the generation of energy inside cells, and the transport of oxygen around the body. Iron deficiency is the most common of all nutrient deficiencies, affecting over 40% of children worldwide. This can lead to anemia and also impair how the brain and nervous system develop, potentially resulting in long-lasting cognitive damage, even after the deficiency has been treated. It is poorly understood how iron contributes to the development of the brain and nervous system. In particular, whether and how it supports nerve stem cells (or NSCs for short) which give rise to the various neural types in the mature brain. To investigate, Ma et al. experimentally reduced the levels of ferritin (a protein which stores iron) in the developing brains of fruit fly larvae. This reduction in ferritin led to lower numbers of NSCs and a smaller brain. Unexpectedly, this effect was largest when ferritin levels were reduced in glial cells which support and send signals to NSCs, rather than in the stem cells themselves. Ma et al. then used fluorescence microscopy to confirm that glial cells make and contain a lot of ferritin which can be transported to NSCs. Adding iron supplements to the diet of flies lacking ferritin did not lead to normal numbers of stem cells in the brains of the developing fruit flies, whereas adding compounds that reduce the amount of iron led to lower numbers of stem cells. Together, this suggests that ferritin transports iron from glial cells to the NSCs. Without ferritin and iron, the NSCs could not produce enough energy to divide and make new stem cells. This caused the NSCs to lose the characteristics of stem cells and prematurely turn into other types of neurons or glial cells. Together, these findings show that when iron cannot move from glial cells to NSCs this leads to defects in brain development. Future experiments will have to test whether a similar transport of iron from supporting cells to NSCs also occurs in the developing brains of mammals, and whether this mechanism applies to stem cells in other parts of the body.


Sujet(s)
Protéines de Drosophila , Ferritines , Fer , Cellules souches neurales , Névroglie , Animaux , Cellules souches neurales/métabolisme , Névroglie/métabolisme , Fer/métabolisme , Ferritines/métabolisme , Ferritines/génétique , Protéines de Drosophila/métabolisme , Protéines de Drosophila/génétique , Drosophila/métabolisme , Prolifération cellulaire , Différenciation cellulaire , Drosophila melanogaster/métabolisme , Drosophila melanogaster/génétique , Auto-renouvellement cellulaire
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE