Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52.203
Filtrar
1.
J Mol Biol ; 436(17): 168520, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-39237197

RESUMEN

The red flour beetle Tribolium castaneum has emerged as a powerful model in insect functional genomics. However, a major limitation in the field is the lack of a detailed spatio-temporal view of the genetic signatures underpinning the function of distinct tissues and life stages. Here, we present an ontogenetic and tissue-specific web-based resource for Tribolium transcriptomics: BeetleAtlas (https://www.beetleatlas.org). This web application provides access to a database populated with quantitative expression data for nine adult and seven larval tissues, as well as for four embryonic stages of Tribolium. BeetleAtlas allows one to search for individual Tribolium genes to obtain values of both total gene expression and enrichment in different tissues, together with data for individual isoforms. To facilitate cross-species studies, one can also use Drosophila melanogaster gene identifiers to search for related Tribolium genes. For retrieved genes there are options to identify and display the tissue expression of related Tribolium genes or homologous Drosophila genes. Five additional search modes are available to find genes conforming to any of the following criteria: exhibiting high expression in a particular tissue; showing significant differences in expression between larva and adult; having a peak of expression at a specific stage of embryonic development; belonging to a particular functional category; and displaying a pattern of tissue expression similar to that of a query gene. We illustrate how the different feaures of BeetleAtlas can be used to illuminate our understanding of the genetic mechanisms underpinning the biology of what is the largest animal group on earth.


Asunto(s)
Transcriptoma , Tribolium , Animales , Tribolium/genética , Tribolium/embriología , Regulación del Desarrollo de la Expresión Génica , Perfilación de la Expresión Génica , Larva/genética , Larva/crecimiento & desarrollo , Larva/metabolismo , Bases de Datos Genéticas , Especificidad de Órganos , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo
2.
Food Res Int ; 194: 114885, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39232523

RESUMEN

The Chinese bayberry pomace wine (CPW) was prepared with the assisted fermentation of lactic acid bacteria and acetic acid bacteria, and its antioxidant effect on Drosophila melanogaster was researched. After mixed fermentation, CPW had a better color, which means there was more retention of anthocyanins, and the functional activity of anthocyanins could enhance the antioxidant capacity of flies. We found that the lifespan of flies exposed to CPW was prolonged, and the reproductive capacity of these flies was decreased. The food intake of flies was also influenced by CPW with gender differences. Furthermore, CPW alleviated the excessive proliferation of the intestinal precursor cells of H2O2-induced flies and activated the transcription level of antibacterial peptide genes. CPW had a protective effect on H2O2-induced acute injury flies, with an increased survival rate, enhanced SOD and CAT activities, and decreased malondialdehyde (MDA) content in flies. The expression of oxidative stress-related genes including CuZn-SOD, Mn-SOD, and CAT was also significantly upregulated by CPW, but the downregulation effect of CPW on age-related gene expression such as methuselah (MTH), the target of rapamycin (TOR) and ribosomaiprotein S6 kinase (S6K) was sex-specific. These results suggested that CPW played an important role in anti-oxidative stress injury, which was beneficial to promoting the reuse of by-products from Chinese bayberry processing.


Asunto(s)
Antioxidantes , Drosophila melanogaster , Fermentación , Myrica , Estrés Oxidativo , Vino , Animales , Drosophila melanogaster/efectos de los fármacos , Drosophila melanogaster/metabolismo , Estrés Oxidativo/efectos de los fármacos , Vino/análisis , Antioxidantes/farmacología , Antioxidantes/metabolismo , Masculino , Femenino , Myrica/química , Longevidad/efectos de los fármacos , Antocianinas/farmacología , Peróxido de Hidrógeno/metabolismo , Frutas/química , Pueblos del Este de Asia
3.
Elife ; 122024 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-39240259

RESUMEN

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.


Asunto(s)
Proteínas de Drosophila , Drosophila melanogaster , Hormonas de Insectos , Neuronas , Receptores de Esteroides , Conducta Sexual Animal , Animales , Drosophila melanogaster/fisiología , Drosophila melanogaster/crecimiento & desarrollo , Femenino , Conducta Sexual Animal/fisiología , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Neuronas/fisiología , Neuronas/metabolismo , Hormonas de Insectos/metabolismo , Receptores de Esteroides/metabolismo , Receptores de Esteroides/genética , Ecdisona/metabolismo , Metamorfosis Biológica/fisiología , Masculino , Larva/crecimiento & desarrollo , Larva/fisiología , Proteínas de Insectos
4.
Life Sci Alliance ; 7(11)2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39237365

RESUMEN

Sleep and circadian rhythm dysfunctions are common clinical features of Alzheimer's disease (AD). Increasing evidence suggests that in addition to being a symptom, sleep disturbances can also drive the progression of neurodegeneration. Protein aggregation is a pathological hallmark of AD; however, the molecular pathways behind how sleep affects protein homeostasis remain elusive. Here we demonstrate that sleep modulation influences proteostasis and the progression of neurodegeneration in Drosophila models of tauopathy. We show that sleep deprivation enhanced Tau aggregational toxicity resulting in exacerbated synaptic degeneration. In contrast, sleep induction using gaboxadol led to reduced toxic Tau accumulation in neurons as a result of modulated autophagic flux and enhanced clearance of ubiquitinated Tau, suggesting altered protein processing and clearance that resulted in improved synaptic integrity and function. These findings highlight the complex relationship between sleep and regulation of protein homeostasis and the neuroprotective potential of sleep-enhancing therapeutics to slow the progression or delay the onset of neurodegeneration.


Asunto(s)
Enfermedad de Alzheimer , Autofagia , Modelos Animales de Enfermedad , Proteostasis , Sueño , Proteínas tau , Animales , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Sueño/fisiología , Proteínas tau/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Neuronas/metabolismo , Drosophila melanogaster/metabolismo , Humanos , Drosophila , Privación de Sueño/metabolismo , Privación de Sueño/fisiopatología , Tauopatías/metabolismo , Tauopatías/patología , Tauopatías/etiología
5.
Development ; 151(18)2024 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-39284714

RESUMEN

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.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Diferenciación Celular , Proteínas de Drosophila , Células-Madre Neurales , Células Neuroepiteliales , Receptores Notch , Transducción de Señal , Animales , Células-Madre Neurales/metabolismo , Células-Madre Neurales/citología , Receptores Notch/metabolismo , Receptores Notch/genética , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Células Neuroepiteliales/metabolismo , Células Neuroepiteliales/citología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Diferenciación Celular/genética , Regulación del Desarrollo de la Expresión Génica , Drosophila melanogaster/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/citología , Factores de Tiempo , Drosophila/metabolismo
6.
Cell Mol Life Sci ; 81(1): 396, 2024 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-39261338

RESUMEN

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.


Asunto(s)
Progresión de la Enfermedad , Proteínas de Drosophila , Regulación hacia Arriba , Animales , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Humanos , Neoplasias/patología , Neoplasias/metabolismo , Neoplasias/genética , Apoptosis/genética , Transducción de Señal , Proteína Wnt1/metabolismo , Proteína Wnt1/genética , Fosfoenolpiruvato Carboxiquinasa (ATP)/metabolismo , Fosfoenolpiruvato Carboxiquinasa (ATP)/genética , Glucosa/metabolismo , Inestabilidad Genómica , Fosfoenolpiruvato Carboxiquinasa (GTP)/metabolismo , Fosfoenolpiruvato Carboxiquinasa (GTP)/genética , Línea Celular Tumoral , Drosophila melanogaster/metabolismo , Drosophila melanogaster/genética , Regulación Neoplásica de la Expresión Génica , Trehalosa/metabolismo , Carbohidratos de la Dieta/efectos adversos , Drosophila/metabolismo
7.
Sci Rep ; 14(1): 21182, 2024 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-39261567

RESUMEN

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.


Asunto(s)
Acrilamida , Quitosano , Drosophila melanogaster , Limosilactobacillus fermentum , Nanopartículas , Probióticos , Animales , Quitosano/química , Quitosano/farmacología , Probióticos/administración & dosificación , Nanopartículas/química , Acrilamida/química , Acrilamida/toxicidad , Drosophila melanogaster/efectos de los fármacos , Femenino , Microbioma Gastrointestinal/efectos de los fármacos
8.
Pestic Biochem Physiol ; 204: 106102, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39277424

RESUMEN

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.


Asunto(s)
Sistema Enzimático del Citocromo P-450 , Drosophila melanogaster , Butóxido de Piperonilo , Animales , Butóxido de Piperonilo/farmacología , Drosophila melanogaster/efectos de los fármacos , Drosophila melanogaster/genética , Sistema Enzimático del Citocromo P-450/genética , Sistema Enzimático del Citocromo P-450/metabolismo , Insecticidas/toxicidad , Insecticidas/farmacología , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Transcripción Genética/efectos de los fármacos
9.
Nat Commun ; 15(1): 8053, 2024 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-39277611

RESUMEN

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.


Asunto(s)
Animales Modificados Genéticamente , Proteínas de Drosophila , Drosophila melanogaster , Tecnología de Genética Dirigida , Animales , Femenino , Masculino , Drosophila melanogaster/genética , Tecnología de Genética Dirigida/métodos , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Alelos , Sistemas CRISPR-Cas , Genes Dominantes , Control Biológico de Vectores/métodos , Infertilidad/genética , Infertilidad/terapia , ARN Guía de Sistemas CRISPR-Cas/genética , Proteínas de Unión al ADN
10.
Narra J ; 4(2): e898, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39280279

RESUMEN

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.


Asunto(s)
Antiinflamatorios no Esteroideos , Drosophila melanogaster , Indometacina , Enfermedades Intestinales , Animales , Drosophila melanogaster/efectos de los fármacos , Indometacina/efectos adversos , Indometacina/farmacología , Antiinflamatorios no Esteroideos/farmacología , Antiinflamatorios no Esteroideos/efectos adversos , Enfermedades Intestinales/inducido químicamente , Enfermedades Intestinales/patología , Enfermedades Intestinales/genética , Enfermedades Intestinales/tratamiento farmacológico , Modelos Animales de Enfermedad , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo
11.
Narra J ; 4(2): e818, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39280322

RESUMEN

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.


Asunto(s)
Envejecimiento , Cafeína , Larva , Animales , Larva/efectos de los fármacos , Larva/inmunología , Cafeína/farmacología , Envejecimiento/efectos de los fármacos , Envejecimiento/inmunología , Drosophila/efectos de los fármacos , Drosophila melanogaster/efectos de los fármacos , Drosophila melanogaster/inmunología , Drosophila melanogaster/microbiología
12.
Sci Adv ; 10(36): eadi9101, 2024 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-39231215

RESUMEN

A-to-I RNA editing is a cellular mechanism that generates transcriptomic and proteomic diversity, which is essential for neuronal and immune functions. It involves the conversion of specific adenosines in RNA molecules to inosines, which are recognized as guanosines by cellular machinery. Despite the vast number of editing sites observed across the animal kingdom, pinpointing critical sites and understanding their in vivo functions remains challenging. Here, we study the function of an evolutionary conserved editing site in Drosophila, located in glutamate-gated chloride channel (GluClα). Our findings reveal that flies lacking editing at this site exhibit reduced olfactory responses to odors and impaired pheromone-dependent social interactions. Moreover, we demonstrate that editing of this site is crucial for the proper processing of olfactory information in projection neurons. Our results highlight the value of using evolutionary conservation as a criterion for identifying editing events with potential functional significance and paves the way for elucidating the intricate link between RNA modification, neuronal physiology, and behavior.


Asunto(s)
Canales de Cloruro , Edición de ARN , Animales , Canales de Cloruro/metabolismo , Canales de Cloruro/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Olfato/fisiología , Olfato/genética , Conducta Animal , Drosophila melanogaster/genética , Drosophila melanogaster/fisiología , Inosina/metabolismo , Inosina/genética , Odorantes , Adenosina/metabolismo , Drosophila/genética
13.
Genes Brain Behav ; 23(5): e70000, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39231190

RESUMEN

Mutations in voltage-gated sodium (Nav) channels, which are essential for generating and propagating action potentials, can lead to serious neurological disorders, such as epilepsy. However, disease-causing Nav channel mutations do not always result in severe symptoms, suggesting that the disease conditions are significantly affected by other genetic factors and various environmental exposures, collectively known as the "exposome". Notably, recent research emphasizes the pivotal role of commensal bacteria in neural development and function. Although these bacteria typically benefit the nervous system under normal conditions, their impact during pathological states remains largely unknown. Here, we investigated the influence of commensal microbes on seizure-like phenotypes exhibited by paraShu-a gain-of-function mutant of the Drosophila Nav channel gene, paralytic. Remarkably, the elimination of endogenous bacteria considerably ameliorated neurological impairments in paraShu. Consistently, reintroducing bacteria, specifically from the Lactobacillus or Acetobacter genera, heightened the phenotypic severity in the bacteria-deprived mutants. These findings posit that particular native bacteria contribute to the severity of seizure-like phenotypes in paraShu. We further uncovered that treating paraShu with antibiotics boosted Nrf2 signaling in the gut, and that global Nrf2 activation mirrored the effects of removing bacteria from paraShu. This raises the possibility that the removal of commensal bacteria suppresses the seizure-like manifestations through augmented antioxidant responses. Since bacterial removal during development was critical for suppression of adult paraShu phenotypes, our research sets the stage for subsequent studies, aiming to elucidate the interplay between commensal bacteria and the developing nervous system in conditions predisposed to the hyperexcitable nervous system.


Asunto(s)
Convulsiones , Canales de Sodio Activados por Voltaje , Animales , Convulsiones/genética , Canales de Sodio Activados por Voltaje/genética , Canales de Sodio Activados por Voltaje/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/microbiología , Fenotipo , Mutación , Acetobacter/genética , Acetobacter/metabolismo , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Simbiosis/genética , Microbioma Gastrointestinal
14.
Nat Commun ; 15(1): 7733, 2024 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-39231950

RESUMEN

Strains of the Bacillus cereus (Bc) group are sporulating bacteria commonly associated with foodborne outbreaks. Spores are dormant cells highly resistant to extreme conditions. Nevertheless, the pathological processes associated with the ingestion of either vegetative cells or spores remain poorly understood. Here, we demonstrate that while ingestion of vegetative bacteria leads to their rapid elimination from the intestine of Drosophila melanogaster, a single ingestion of spores leads to the persistence of bacteria for at least 10 days. We show that spores do not germinate in the anterior part of the intestine which bears the innate immune defenses. Consequently, spores reach the posterior intestine where they germinate and activate both the Imd and Toll immune pathways. Unexpectedly, this leads to the induction of amidases, which are negative regulators of the immune response, but not to antimicrobial peptides. Thereby, the local germination of spores in the posterior intestine dampens the immune signaling that in turn fosters the persistence of Bc bacteria. This study provides evidence for how Bc spores hijack the intestinal immune defenses allowing the localized birth of vegetative bacteria responsible for the digestive symptoms associated with foodborne illness outbreaks.


Asunto(s)
Bacillus cereus , Drosophila melanogaster , Esporas Bacterianas , Bacillus cereus/inmunología , Esporas Bacterianas/inmunología , Animales , Drosophila melanogaster/inmunología , Drosophila melanogaster/microbiología , Intestinos/microbiología , Intestinos/inmunología , Inmunidad Innata , Proteínas de Drosophila/metabolismo , Transducción de Señal/inmunología , Receptores Toll-Like/metabolismo , Receptores Toll-Like/inmunología , Femenino
15.
Sci Rep ; 14(1): 20867, 2024 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-39242711

RESUMEN

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.


Asunto(s)
Proteínas de Drosophila , Proteínas del Choque Térmico HSP40 , Proteína Huntingtina , Enfermedad de Huntington , Animales , Humanos , Animales Modificados Genéticamente , Modelos Animales de Enfermedad , Drosophila , Drosophila melanogaster , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas del Choque Térmico HSP40/genética , Proteínas del Choque Térmico HSP40/metabolismo , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/patología , Agregado de Proteínas , Agregación Patológica de Proteínas/genética , Saccharomyces cerevisiae
16.
Nat Commun ; 15(1): 8020, 2024 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-39271704

RESUMEN

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.


Asunto(s)
Gránulos Citoplasmáticos , Proteínas de Drosophila , Drosophila melanogaster , Biosíntesis de Proteínas , ARN Mensajero , Animales , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Gránulos Citoplasmáticos/metabolismo , Drosophila melanogaster/metabolismo , Drosophila melanogaster/genética , ARN Mensajero/metabolismo , ARN Mensajero/genética , Células Germinativas/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , Imagen Individual de Molécula , Drosophila/metabolismo , Drosophila/genética , Condensados Biomoleculares/metabolismo
17.
Cells ; 13(17)2024 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-39273016

RESUMEN

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.


Asunto(s)
Drosophila melanogaster , Colorantes Fluorescentes , Animales , Drosophila melanogaster/metabolismo , Colorantes Fluorescentes/química , Procesos Estocásticos , Proteínas de Drosophila/metabolismo , Microscopía Fluorescente/métodos , Rodaminas/química
18.
Molecules ; 29(17)2024 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-39275038

RESUMEN

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.


Asunto(s)
Modelos Animales de Enfermedad , Drosophila melanogaster , Juglans , Enfermedad de Parkinson , Aceites de Plantas , Animales , Drosophila melanogaster/efectos de los fármacos , Juglans/química , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/metabolismo , Aceites de Plantas/farmacología , Aceites de Plantas/química , Dopamina/metabolismo , Rotenona , Encéfalo/metabolismo , Encéfalo/efectos de los fármacos , Fármacos Neuroprotectores/farmacología
19.
PLoS Biol ; 22(9): e3002783, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39226305

RESUMEN

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.


Asunto(s)
Forma de la Célula , Proteínas de Drosophila , Drosophila melanogaster , Distroglicanos , Matriz Extracelular , Integrinas , Laminina , Retina , Animales , Distroglicanos/metabolismo , Laminina/metabolismo , Integrinas/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Matriz Extracelular/metabolismo , Retina/metabolismo , Retina/crecimiento & desarrollo , Retina/citología , Retina/embriología , Drosophila melanogaster/metabolismo , Drosophila melanogaster/crecimiento & desarrollo , Drosophila melanogaster/genética , Morfogénesis , Adhesión Celular , Drosophila/metabolismo , Drosophila/crecimiento & desarrollo
20.
PLoS Genet ; 20(9): e1011387, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39226333

RESUMEN

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.


Asunto(s)
Daño del ADN , Drosophila melanogaster , Alas de Animales , Animales , Alas de Animales/crecimiento & desarrollo , Alas de Animales/metabolismo , Drosophila melanogaster/crecimiento & desarrollo , Drosophila melanogaster/genética , Proliferación Celular , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Apoptosis , Discos Imaginales/crecimiento & desarrollo , Discos Imaginales/metabolismo , Cicatrización de Heridas/genética , Senescencia Celular , Sistema de Señalización de MAP Quinasas , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Ciclo Celular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA