Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
G3 (Bethesda) ; 2024 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-38409337

RESUMEN

Robust genetic systems to control the expression of transgenes in a spatial and temporal manner are a valuable asset for researchers. The GeneSwitch system induced by the drug RU486 has gained widespread use in the Drosophila community. However, some concerns were raised as negative effects were seen depending on the stock, transgene, stage and tissue under study. Here, we characterized the adverse effects triggered by activating the GeneSwitch system in adult muscles using the MHC-GS-GAL4 driver. When a control, mock UAS-RNAi transgene was induced by feeding adult flies with RU486, we found that the overall muscle structure, including myofibrils and mitochondrial shape, was significantly disrupted and led to a significant reduction in the lifespan. Remarkably, lifespan was even shorter when two copies of the driver were used even without the mock UAS-RNAi transgene. Thus, researchers should be cautious when interpreting the results given the adverse effects we found when inducing RU486-dependent MHC-GS-GAL4 in adult muscles. To account for the impact of these effects we recommend adjusting the dose of RU486, setting up additional control groups, such as a mock UAS-RNAi transgene, to validate the findings when using this inducible genetic system, as comparing the phenotypes between RU486-treated and untreated animals could be insufficient.

2.
Front Immunol ; 14: 1266359, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37799716

RESUMEN

Introduction: Inflammatory epidermolysis bullosa acquisita (EBA) is characterized by a neutrophilic response to anti-type VII collagen (COL7) antibodies resulting in the development of skin inflammation and blistering. The antibody transfer model of EBA closely mirrors this EBA phenotype. Methods: To better understand the changes induced in neutrophils upon recruitment from peripheral blood into lesional skin in EBA, we performed single-cell RNA-sequencing of whole blood and skin dissociate to capture minimally perturbed neutrophils and characterize their transcriptome. Results: Through this approach, we identified clear distinctions between circulating activated neutrophils and intradermal neutrophils. Most strikingly, the gene expression of multiple C-type lectin receptors, which have previously been reported to orchestrate host defense against fungi and select bacteria, were markedly dysregulated. After confirming the upregulation of Clec4n, Clec4d, and Clec4e in experimental EBA as well as in lesional skin from patients with inflammatory EBA, we performed functional studies in globally deficient Clec4e-/- and Clec4d-/- mice as well as in neutrophil-specific Clec4n-/- mice. Deficiency in these genes did not reduce disease in the EBA model. Discussion: Collectively, our results suggest that while the upregulation of Clec4n, Clec4d, and Clec4e is a hallmark of activated dermal neutrophil populations, their individual contribution to the pathogenesis of EBA is dispensable.


Asunto(s)
Epidermólisis Ampollosa Adquirida , Humanos , Animales , Ratones , Neutrófilos , Autoanticuerpos , Piel , Vesícula
3.
Proc Natl Acad Sci U S A ; 120(15): e2220770120, 2023 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-37011211

RESUMEN

The canonical role of the transcription factor E2F is to control the expression of cell cycle genes by binding to the E2F sites in their promoters. However, the list of putative E2F target genes is extensive and includes many metabolic genes, yet the significance of E2F in controlling the expression of these genes remains largely unknown. Here, we used the CRISPR/Cas9 technology to introduce point mutations in the E2F sites upstream of five endogenous metabolic genes in Drosophila melanogaster. We found that the impact of these mutations on both the recruitment of E2F and the expression of the target genes varied, with the glycolytic gene, Phosphoglycerate kinase (Pgk), being mostly affected. The loss of E2F regulation on the Pgk gene led to a decrease in glycolytic flux, tricarboxylic acid cycle intermediates levels, adenosine triphosphate (ATP) content, and an abnormal mitochondrial morphology. Remarkably, chromatin accessibility was significantly reduced at multiple genomic regions in PgkΔE2F mutants. These regions contained hundreds of genes, including metabolic genes that were downregulated in PgkΔE2F mutants. Moreover, PgkΔE2F animals had shortened life span and exhibited defects in high-energy consuming organs, such as ovaries and muscles. Collectively, our results illustrate how the pleiotropic effects on metabolism, gene expression, and development in the PgkΔE2F animals underscore the importance of E2F regulation on a single E2F target, Pgk.


Asunto(s)
Proteínas de Drosophila , Drosophila , Factores de Transcripción E2F , Fosfoglicerato Quinasa , Animales , Cromatina , Drosophila/genética , Factores de Transcripción E2F/genética , Fosfoglicerato Quinasa/genética , Fosfoglicerato Quinasa/metabolismo , Regiones Promotoras Genéticas , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo
4.
Elife ; 102021 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-34251339

RESUMEN

The E2F transcription factors play a critical role in controlling cell fate. In Drosophila, the inactivation of E2F in either muscle or fat body results in lethality, suggesting an essential function for E2F in these tissues. However, the cellular and organismal consequences of inactivating E2F in these tissues are not fully understood. Here, we show that the E2F loss exerts both tissue-intrinsic and systemic effects. The proteomic profiling of E2F-deficient muscle and fat body revealed that E2F regulates carbohydrate metabolism, a conclusion further supported by metabolomic profiling. Intriguingly, animals with E2F-deficient fat body had a lower level of circulating trehalose and reduced storage of fat. Strikingly, a sugar supplement was sufficient to restore both trehalose and fat levels, and subsequently rescued animal lethality. Collectively, our data highlight the unexpected complexity of E2F mutant phenotype, which is a result of combining both tissue-specific and systemic changes that contribute to animal development.


Asunto(s)
Proteínas de Drosophila/metabolismo , Factores de Transcripción E2F/metabolismo , Cuerpo Adiposo/metabolismo , Factores de Transcripción/metabolismo , Animales , Metabolismo de los Hidratos de Carbono , Ciclo Celular , Drosophila , Proteínas de Drosophila/genética , Factores de Transcripción E2F/genética , Regulación del Desarrollo de la Expresión Génica , Metabolómica/métodos , Músculos/metabolismo , Fenotipo , Proteómica/métodos , Factores de Transcripción/genética , Transcripción Genética , Trehalosa/metabolismo
5.
EMBO Rep ; 21(10): e49555, 2020 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-32815271

RESUMEN

In Drosophila, the wing disc-associated muscle precursor cells give rise to the fibrillar indirect flight muscles (IFM) and the tubular direct flight muscles (DFM). To understand early transcriptional events underlying this muscle diversification, we performed single-cell RNA-sequencing experiments and built a cell atlas of myoblasts associated with third instar larval wing disc. Our analysis identified distinct transcriptional signatures for IFM and DFM myoblasts that underlie the molecular basis of their divergence. The atlas further revealed various states of differentiation of myoblasts, thus illustrating previously unappreciated spatial and temporal heterogeneity among them. We identified and validated novel markers for both IFM and DFM myoblasts at various states of differentiation by immunofluorescence and genetic cell-tracing experiments. Finally, we performed a systematic genetic screen using a panel of markers from the reference cell atlas as an entry point and found a novel gene, Amalgam which is functionally important in muscle development. Our work provides a framework for leveraging scRNA-seq for gene discovery and details a strategy that can be applied to other scRNA-seq datasets.


Asunto(s)
Proteínas de Drosophila , Drosophila , Animales , Drosophila/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Desarrollo de Músculos/genética , Alas de Animales
6.
Cell Rep ; 26(3): 702-719.e6, 2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30650361

RESUMEN

The importance of the retinoblastoma tumor suppressor protein pRB in cell cycle control is well established. However, less is known about its role in differentiation during animal development. Here, we investigated the role of Rbf, the Drosophila pRB homolog, in adult skeletal muscles. We found that the depletion of Rbf severely reduced muscle growth and altered myofibrillogenesis but only minimally affected myoblast proliferation. We identified an Rbf-dependent transcriptional program in late muscle development that is distinct from the canonical role of Rbf in cell cycle control. Unexpectedly, Rbf acts as a transcriptional activator of the myogenic and metabolic genes in the growing muscles. The genomic regions bound by Rbf contained the binding sites of several factors that genetically interacted with Rbf by modulating Rbf-dependent phenotype. Thus, our results reveal a distinctive role for Rbf as a direct activator of the myogenic transcriptional program that drives late muscle differentiation.


Asunto(s)
Proteínas de Drosophila/genética , Desarrollo de Músculos/genética , Músculo Esquelético/metabolismo , Proteína de Retinoblastoma/genética , Factores de Transcripción/metabolismo , Animales , Diferenciación Celular , Drosophila , Proteína de Retinoblastoma/metabolismo , Factores de Transcripción/genética
7.
Nat Commun ; 9(1): 5024, 2018 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-30479347

RESUMEN

The function of Retinoblastoma tumor suppressor (pRB) is greatly influenced by the cellular context, therefore the consequences of pRB inactivation are cell-type-specific. Here we employ single cell RNA-sequencing (scRNA-seq) to profile the impact of an Rbf mutation during Drosophila eye development. First, we build a catalogue of 11,500 wild type eye disc cells containing major known cell types. We find a transcriptional switch occurring in differentiating photoreceptors at the time of axonogenesis. Next, we map a cell landscape of Rbf mutant and identify a mutant-specific cell population that shows intracellular acidification due to increase in glycolytic activity. Genetic experiments demonstrate that such metabolic changes, restricted to this unique Rbf mutant population, sensitize cells to apoptosis and define the pattern of cell death in Rbf mutant eye disc. Thus, these results illustrate how scRNA-seq can be applied to dissect mutant phenotypes.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Mutación/genética , Proteína de Retinoblastoma/genética , Análisis de Secuencia de ARN/métodos , Análisis de la Célula Individual/métodos , Factores de Transcripción/genética , Animales , Apoptosis , Axones/metabolismo , Ojo/citología , Larva/citología , Modelos Biológicos , Neurogénesis , Células Fotorreceptoras de Invertebrados/metabolismo , Transcripción Genética
8.
Dev Cell ; 43(6): 689-703.e5, 2017 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-29233476

RESUMEN

To understand the consequences of the complete elimination of E2F regulation, we profiled the proteome of Drosophila dDP mutants that lack functional E2F/DP complexes. The results uncovered changes in the larval fat body, a differentiated tissue that grows via endocycles. We report an unexpected mechanism of E2F/DP action that promotes quiescence in this tissue. In the fat body, dE2F/dDP limits cell-cycle progression by suppressing DNA damage responses. Loss of dDP upregulates dATM, allowing cells to sense and repair DNA damage and increasing replication of loci that are normally under-replicated in wild-type tissues. Genetic experiments show that ectopic dATM is sufficient to promote DNA synthesis in wild-type fat body cells. Strikingly, reducing dATM levels in dDP-deficient fat bodies restores cell-cycle control, improves tissue morphology, and extends animal development. These results show that, in some cellular contexts, dE2F/dDP-dependent suppression of DNA damage signaling is key for cell-cycle control and needed for normal development.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/biosíntesis , Proteínas de Drosophila/metabolismo , Factores de Transcripción E2F/genética , Factores de Transcripción E2F/metabolismo , Cuerpo Adiposo/fisiología , Transactivadores/metabolismo , Animales , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , División Celular/fisiología , Replicación del ADN , Proteínas de Unión al ADN/metabolismo , Drosophila , Proteínas de Drosophila/biosíntesis , Proteínas de Drosophila/genética , Cuerpo Adiposo/citología , Proteínas Serina-Treonina Quinasas , Transactivadores/genética , Transcriptoma
9.
Nat Commun ; 7: 10509, 2016 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-26823289

RESUMEN

The E2F transcription factor is a key cell cycle regulator. However, the inactivation of the entire E2F family in Drosophila is permissive throughout most of animal development until pupation when lethality occurs. Here we show that E2F function in the adult skeletal muscle is essential for animal viability since providing E2F function in muscles rescues the lethality of the whole-body E2F-deficient animals. Muscle-specific loss of E2F results in a significant reduction in muscle mass and thinner myofibrils. We demonstrate that E2F is dispensable for proliferation of muscle progenitor cells, but is required during late myogenesis to directly control the expression of a set of muscle-specific genes. Interestingly, E2f1 provides a major contribution to the regulation of myogenic function, while E2f2 appears to be less important. These findings identify a key function of E2F in skeletal muscle required for animal viability, and illustrate how the cell cycle regulator is repurposed in post-mitotic cells.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Factor de Transcripción E2F1/metabolismo , Factor de Transcripción E2F2/metabolismo , Desarrollo de Músculos/genética , Músculos/embriología , Transactivadores/metabolismo , Animales , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Factor de Transcripción E2F1/genética , Factor de Transcripción E2F2/genética , Femenino , Regulación del Desarrollo de la Expresión Génica/fisiología , Técnicas de Silenciamiento del Gen , Larva , Masculino , Pupa , Transactivadores/genética
10.
BMC Neurosci ; 13: 78, 2012 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-22762289

RESUMEN

BACKGROUND: Members of the proteolipid protein family, including the four-transmembrane glycoprotein M6a, are involved in neuronal plasticity in mammals. Results from our group previously demonstrated that M6, the only proteolipid protein expressed in Drosophila, localizes to the cell membrane in follicle cells. M6 loss triggers female sterility, which suggests a role for M6 in follicular cell remodeling. These results were the basis of the present study, which focused on the function and requirements of M6 in the fly nervous system. RESULTS: The present study identified two novel, tissue-regulated M6 isoforms with variable N- and C- termini, and showed that M6 is the functional fly ortholog of Gpm6a. In the adult brain, the protein was localized to several neuropils, such as the optic lobe, the central complex, and the mushroom bodies. Interestingly, although reduced M6 levels triggered a mild rough-eye phenotype, hypomorphic M6 mutants exhibited a defective response to light. CONCLUSIONS: Based on its ability to induce filopodium formation we propose that M6 is key in cell remodeling processes underlying visual system function. These results bring further insight into the role of M6/M6a in biological processes involving neuronal plasticity and behavior in flies and mammals.


Asunto(s)
Conducta Animal/fisiología , Ojo/metabolismo , Regulación de la Expresión Génica/fisiología , Glicoproteínas de Membrana/fisiología , Vías Visuales/metabolismo , Empalme Alternativo/genética , Animales , Animales Modificados Genéticamente , Línea Celular Tumoral , Clonación Molecular , Secuencia Conservada/genética , Drosophila , Proteínas de Drosophila/genética , Ojo/ultraestructura , Regulación de la Expresión Génica/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Esperanza de Vida , Glicoproteínas de Membrana/genética , Microscopía Electrónica de Rastreo , Actividad Motora/genética , Mutación/genética , Neuroblastoma/patología , Neurópilo/metabolismo , Neurópilo/ultraestructura , Lóbulo Óptico de Animales no Mamíferos/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Seudópodos/metabolismo , ARN Mensajero/metabolismo , Transfección , Vías Visuales/ultraestructura
11.
PLoS One ; 6(5): e19715, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21603606

RESUMEN

We had previously shown that the transmembrane glycoprotein M6a, a member of the proteolipid protein (PLP) family, regulates neurite/filopodium outgrowth, hence, M6a might be involved in neuronal remodeling and differentiation. In this work we focused on M6, the only PLP family member present in Drosophila, and ortholog to M6a. Unexpectedly, we found that decreased expression of M6 leads to female sterility. M6 is expressed in the membrane of the follicular epithelium in ovarioles throughout oogenesis. Phenotypes triggered by M6 downregulation in hypomorphic mutants included egg collapse and egg permeability, thus suggesting M6 involvement in eggshell biosynthesis. In addition, RNAi-mediated M6 knockdown targeted specifically to follicle cells induced an arrest of egg chamber development, revealing that M6 is essential in oogenesis. Interestingly, M6-associated phenotypes evidenced abnormal changes of the follicle cell shape and disrupted follicular epithelium in mid- and late-stage egg chambers. Therefore, we propose that M6 plays a role in follicular epithelium maintenance involving membrane cell remodeling during oogenesis in Drosophila.


Asunto(s)
Proteínas de Drosophila/fisiología , Proteínas de la Membrana/fisiología , Oogénesis , Animales , Drosophila , Epitelio , Femenino , Folículo Ovárico
12.
Biochem Pharmacol ; 75(9): 1817-26, 2008 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-18342838

RESUMEN

Fluoxetine, a selective serotonin reuptake inhibitor, is widely used for the treatment of depressive symptoms of cancer patients. However, there are contradictory evidences about its effects on immunity and cancer. Thus, we studied the effects of fluoxetine on tumor growth and on antitumoral T-cell-mediated immunity. In vivo chronic fluoxetine treatment inhibited tumor growth, and increased latency of appearance of solid tumors and survival of mice. Fluoxetine administration also increased mitogen-induced T-cell proliferation and Tumor Necrosis Factor-alpha (TNF-alpha) and Interferon-gamma (IFN-gamma) expression, without altering CD4(+)/CD8(+) ratio. In vitro, fluoxetine did not affect tumor cells proliferation, but it exerted a direct effect on T lymphocytes. Both fluoxetine and serotonin stimulated proliferation induced by a suboptimal mitogen concentration but inhibited proliferation at the optimal one. When both drugs were combined the results indicated that the effects of fluoxetine are in part independent of its ability to elevate serotonin extracellular levels. Finally, continue fluoxetine administration in nude mice - devoid of T lymphocytes - did not modify tumor progression, thus supporting the hypothesis of an immuno-modulatory effect of this drug on T cells that drives tumor growth control. These findings indicate, for the first time, that fluoxetine inhibits tumor growth through modulation of T-cell-mediated immunity by the already known serotonin-dependent pathway and by a novel independent mechanism.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Fluoxetina/uso terapéutico , Linfoma de Células T/tratamiento farmacológico , Inhibidores Selectivos de la Recaptación de Serotonina/uso terapéutico , Serotonina/metabolismo , Linfocitos T/efectos de los fármacos , Animales , Relación CD4-CD8 , Línea Celular Tumoral , Citocinas/biosíntesis , Femenino , Fluoxetina/administración & dosificación , Fluoxetina/farmacología , Inmunidad Celular/efectos de los fármacos , Ganglios Linfáticos/efectos de los fármacos , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Linfoma de Células T/inmunología , Linfoma de Células T/metabolismo , Linfoma de Células T/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Trasplante de Neoplasias , Serotonina/farmacología , Inhibidores Selectivos de la Recaptación de Serotonina/administración & dosificación , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Linfocitos T/inmunología , Linfocitos T/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...