Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
PLoS Genet ; 19(8): e1010875, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37578970

RESUMEN

Lipid droplets (LDs) are ubiquitous organelles that facilitate neutral lipid storage in cells, including energy-dense triglycerides. They are found in all investigated metazoan embryos where they are thought to provide energy for development. Intriguingly, early embryos of diverse metazoan species asymmetrically allocate LDs amongst cellular lineages, a process which can involve massive intracellular redistribution of LDs. However, the biological reason for asymmetric lineage allocation is unknown. To address this issue, we utilize the Drosophila embryo where the cytoskeletal mechanisms that drive allocation are well characterized. We disrupt allocation by two different means: Loss of the LD protein Jabba results in LDs adhering inappropriately to glycogen granules; loss of Klar alters the activities of the microtubule motors that move LDs. Both mutants cause the same dramatic change in LD tissue inheritance, shifting allocation of the majority of LDs to the yolk cell instead of the incipient epithelium. Embryos with such mislocalized LDs do not fully consume their LDs and are delayed in hatching. Through use of a dPLIN2 mutant, which appropriately localizes a smaller pool of LDs, we find that failed LD transport and a smaller LD pool affect embryogenesis in a similar manner. Embryos of all three mutants display overlapping changes in their transcriptome and proteome, suggesting that lipid deprivation results in a shared embryonic response and a widespread change in metabolism. Excitingly, we find abundant changes related to redox homeostasis, with many proteins related to glutathione metabolism upregulated. LD deprived embryos have an increase in peroxidized lipids and rely on increased utilization of glutathione-related proteins for survival. Thus, embryos are apparently able to mount a beneficial response upon lipid stress, rewiring their metabolism to survive. In summary, we demonstrate that early embryos allocate LDs into specific lineages for subsequent optimal utilization, thus protecting against oxidative stress and ensuring punctual development.


Asunto(s)
Drosophila , Metabolismo de los Lípidos , Animales , Drosophila/genética , Metabolismo de los Lípidos/genética , Gotas Lipídicas/metabolismo , Triglicéridos/metabolismo , Homeostasis/genética , Oxidación-Reducción
2.
Development ; 150(20)2023 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-37306387

RESUMEN

Lipid droplets (LDs), crucial regulators of lipid metabolism, accumulate during oocyte development. However, their roles in fertility remain largely unknown. During Drosophila oogenesis, LD accumulation coincides with the actin remodeling necessary for follicle development. Loss of the LD-associated Adipose Triglyceride Lipase (ATGL) disrupts both actin bundle formation and cortical actin integrity, an unusual phenotype also seen when the prostaglandin (PG) synthase Pxt is missing. Dominant genetic interactions and PG treatment of follicles indicate that ATGL acts upstream of Pxt to regulate actin remodeling. Our data suggest that ATGL releases arachidonic acid (AA) from LDs to serve as the substrate for PG synthesis. Lipidomic analysis detects AA-containing triglycerides in ovaries, and these are increased when ATGL is lost. High levels of exogenous AA block follicle development; this is enhanced by impairing LD formation and suppressed by reducing ATGL. Together, these data support the model that AA stored in LD triglycerides is released by ATGL to drive the production of PGs, which promote the actin remodeling necessary for follicle development. We speculate that this pathway is conserved across organisms to regulate oocyte development and promote fertility.


Asunto(s)
Proteínas de Drosophila , Prostaglandinas , Animales , Gotas Lipídicas , Actinas , Adipogénesis , Drosophila , Lipasa , Peroxidasas , Proteínas de Drosophila/genética
3.
Development ; 150(20)2023 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-36805634

RESUMEN

Animal embryos are provided by their mothers with a diverse nutrient supply that is crucial for development. In Drosophila, the three most abundant nutrients (triglycerides, proteins and glycogen) are sequestered in distinct storage structures: lipid droplets (LDs), yolk vesicles (YVs) and glycogen granules (GGs). Using transmission electron microscopy as well as live and fixed sample fluorescence imaging, we find that all three storage structures are dispersed throughout the egg but are then spatially allocated to distinct tissues by gastrulation: LDs largely to the peripheral epithelium, YVs and GGs to the central yolk cell. To confound the embryo's ability to sort its nutrients, we employ Jabba and mauve mutants to generate LD-GG and LD-YV compound structures. In these mutants, LDs are mis-sorted to the yolk cell and their turnover is delayed. Our observations demonstrate dramatic spatial nutrient sorting in early embryos and provide the first evidence for its functional importance.


Asunto(s)
Proteínas de Drosophila , Drosophila , Animales , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , Transporte de Proteínas , Nutrientes , Gotas Lipídicas/metabolismo , Glucógeno/metabolismo , Metabolismo de los Lípidos , Proteínas Portadoras/metabolismo
4.
Methods Mol Biol ; 2626: 233-251, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36715908

RESUMEN

Lipid droplets (LDs) are fat storage organelles highly abundant in oocytes and eggs of many vertebrates and invertebrates. They have roles both during oogenesis and in provisioning the developing embryo. In Drosophila, large numbers of LDs are generated in nurse cells during mid-oogenesis and then transferred to oocytes. Their number and spatial distribution changes developmentally and in response to various experimental manipulations. This chapter demonstrates how to visualize LDs in Drosophila follicles, both in fixed tissues and living samples. For fixed samples, the protocol explains how to prepare female flies, dissect ovaries, isolate follicles, fix, apply stains, mount the tissue, and perform imaging. For live samples, the protocol shows how to dissect ovaries, apply a fluorescent LD dye, and culture follicles such that they remain alive and healthy during imaging. Finally, a method is provided that employs in vivo centrifugation to assess colocalization of markers with LDs.


Asunto(s)
Proteínas de Drosophila , Drosophila , Animales , Femenino , Masculino , Drosophila/metabolismo , Gotas Lipídicas/metabolismo , Oogénesis , Proteínas de Drosophila/metabolismo , Oocitos/metabolismo , Metabolismo de los Lípidos/fisiología
5.
J Vis Exp ; (178)2021 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-34958089

RESUMEN

Early Drosophila embryos are large cells containing a vast array of conventional and embryo-specific organelles. During the first three hours of embryogenesis, these organelles undergo dramatic movements powered by actin-based cytoplasmic streaming and motor-driven trafficking along microtubules. The development of a multitude of small, organelle-specific fluorescent probes (FPs) makes it possible to visualize a wide range of different lipid-containing structures in any genotype, allowing live imaging without requiring a genetically encoded fluorophore. This protocol shows how to inject vital dyes and molecular probes into Drosophila embryos to monitor the trafficking of specific organelles by live imaging. This approach is demonstrated by labeling lipid droplets (LDs) and following their bulk movement by particle image velocimetry (PIV). This protocol provides a strategy amenable to the study of other organelles, including lysosomes, mitochondria, yolk vesicles, and the ER, and for tracking the motion of individual LDs along microtubules. Using commercially available dyes brings the benefits of separation into the violet/blue and far-red regions of the spectrum. By multiplex co-labeling of organelles and/or cytoskeletal elements via microinjection, all the genetic resources in Drosophila are available for trafficking studies without the need to introduce fluorescently tagged proteins. Unlike genetically encoded fluorophores, which have low quantum yields and bleach easily, many of the available dyes allow for rapid and simultaneous capture of several channels with high photon yields.


Asunto(s)
Drosophila , Orgánulos , Animales , Colorantes Fluorescentes/química , Gotas Lipídicas , Lípidos/análisis , Lisosomas/metabolismo , Microtúbulos/metabolismo , Orgánulos/metabolismo
6.
Development ; 148(15)2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-34355743

RESUMEN

Because both dearth and overabundance of histones result in cellular defects, histone synthesis and demand are typically tightly coupled. In Drosophila embryos, histones H2B, H2A and H2Av accumulate on lipid droplets (LDs), which are cytoplasmic fat storage organelles. Without LD binding, maternally provided H2B, H2A and H2Av are absent; however, how LDs ensure histone storage is unclear. Using quantitative imaging, we uncover when during oogenesis these histones accumulate, and which step of accumulation is LD dependent. LDs originate in nurse cells (NCs) and are transported to the oocyte. Although H2Av accumulates on LDs in NCs, the majority of the final H2Av pool is synthesized in oocytes. LDs promote intercellular transport of the histone anchor Jabba and thus its presence in the ooplasm. Ooplasmic Jabba then prevents H2Av degradation, safeguarding the H2Av stockpile. Our findings provide insight into the mechanism for establishing histone stores during Drosophila oogenesis and shed light on the function of LDs as protein-sequestration sites.


Asunto(s)
Histonas/metabolismo , Gotas Lipídicas/metabolismo , Animales , Proteínas Portadoras/metabolismo , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , Femenino , Oocitos/metabolismo , Oogénesis/fisiología
7.
PLoS Biol ; 18(1): e3000595, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31961851

RESUMEN

Triglycerides are the major form of stored fat in all animals. One important determinant of whole-body fat storage is whether an animal is male or female. Here, we use Drosophila, an established model for studies on triglyceride metabolism, to gain insight into the genes and physiological mechanisms that contribute to sex differences in fat storage. Our analysis of triglyceride storage and breakdown in both sexes identified a role for triglyceride lipase brummer (bmm) in the regulation of sex differences in triglyceride homeostasis. Normally, male flies have higher levels of bmm mRNA both under normal culture conditions and in response to starvation, a lipolytic stimulus. We find that loss of bmm largely eliminates the sex difference in triglyceride storage and abolishes the sex difference in triglyceride breakdown via strongly male-biased effects. Although we show that bmm function in the fat body affects whole-body triglyceride levels in both sexes, in males, we identify an additional role for bmm function in the somatic cells of the gonad and in neurons in the regulation of whole-body triglyceride homeostasis. Furthermore, we demonstrate that lipid droplets are normally present in both the somatic cells of the male gonad and in neurons, revealing a previously unrecognized role for bmm function, and possibly lipid droplets, in these cell types in the regulation of whole-body triglyceride homeostasis. Taken together, our data reveal a role for bmm function in the somatic cells of the gonad and in neurons in the regulation of male-female differences in fat storage and breakdown and identify bmm as a link between the regulation of triglyceride homeostasis and biological sex.


Asunto(s)
Proteínas de Drosophila/fisiología , Drosophila/genética , Drosophila/metabolismo , Lipasa/fisiología , Metabolismo de los Lípidos/genética , Lipólisis/genética , Caracteres Sexuales , Animales , Animales Modificados Genéticamente , Metabolismo Energético/genética , Femenino , Lipasa/genética , Lipasa/metabolismo , Masculino , Micronutrientes/metabolismo , Triglicéridos/metabolismo
8.
Lab Invest ; 99(12): 1822-1834, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31409893

RESUMEN

Lipid droplets (LDs) utilize microtubules (MTs) to participate in intracellular trafficking of cargo proteins. Cancer cells accumulate LDs and acidify their tumor microenvironment (TME) by increasing the proton pump V-ATPase. However, it is not known whether these two metabolic changes are mechanistically related or influence LD movement. We postulated that LD density and velocity are progressively increased with tumor aggressiveness and are dependent on V-ATPase and the lipolysis regulator pigment epithelium-derived factor (PEDF). LD density was assessed in human prostate cancer (PCa) specimens across Gleason scores (GS) 6-8. LD distribution and velocity were analyzed in low and highly aggressive tumors using live-cell imaging and in cells exposed to low pH and/or treated with V-ATPase inhibitors. The MT network was disrupted and analyzed by α-tubulin staining. LD density positively correlated with advancing GS in human tumors. Acidification promoted peripheral localization and clustering of LDs. Highly aggressive prostate, breast, and pancreatic cell lines had significantly higher maximum LD velocity (LDVmax) than less aggressive and benign cells. LDVmax was MT-dependent and suppressed by blocking V-ATPase directly or indirectly with PEDF. Upon lowering pH, LDs moved to the cell periphery and carried metalloproteinases. These results suggest that acidification of the TME can alter intracellular LD movement and augment velocity in cancer. Restoration of PEDF or blockade of V-ATPase can normalize LD distribution and decrease velocity. This study identifies V-ATPase and PEDF as new modulators of LD trafficking in the cancer microenvironment.


Asunto(s)
Proteínas del Ojo/metabolismo , Gotas Lipídicas/fisiología , Factores de Crecimiento Nervioso/metabolismo , Neoplasias de la Próstata/metabolismo , Serpinas/metabolismo , Microambiente Tumoral , ATPasas de Translocación de Protón Vacuolares/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Células MCF-7 , Masculino , Clasificación del Tumor , Células PC-3 , Próstata/patología , Neoplasias de la Próstata/patología
9.
Mol Cell ; 73(5): 1001-1014.e8, 2019 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-30527540

RESUMEN

In Parkinson's disease (PD), α-synuclein (αS) pathologically impacts the brain, a highly lipid-rich organ. We investigated how alterations in αS or lipid/fatty acid homeostasis affect each other. Lipidomic profiling of human αS-expressing yeast revealed increases in oleic acid (OA, 18:1), diglycerides, and triglycerides. These findings were recapitulated in rodent and human neuronal models of αS dyshomeostasis (overexpression; patient-derived triplication or E46K mutation; E46K mice). Preventing lipid droplet formation or augmenting OA increased αS yeast toxicity; suppressing the OA-generating enzyme stearoyl-CoA-desaturase (SCD) was protective. Genetic or pharmacological SCD inhibition ameliorated toxicity in αS-overexpressing rat neurons. In a C. elegans model, SCD knockout prevented αS-induced dopaminergic degeneration. Conversely, we observed detrimental effects of OA on αS homeostasis: in human neural cells, excess OA caused αS inclusion formation, which was reversed by SCD inhibition. Thus, monounsaturated fatty acid metabolism is pivotal for αS-induced neurotoxicity, and inhibiting SCD represents a novel PD therapeutic approach.


Asunto(s)
Antiparkinsonianos/farmacología , Descubrimiento de Drogas/métodos , Inhibidores Enzimáticos/farmacología , Metabolismo de los Lípidos/efectos de los fármacos , Metabolómica/métodos , Neuronas/efectos de los fármacos , Enfermedad de Parkinson/tratamiento farmacológico , Estearoil-CoA Desaturasa/antagonistas & inhibidores , alfa-Sinucleína/toxicidad , Animales , Caenorhabditis elegans/efectos de los fármacos , Caenorhabditis elegans/enzimología , Caenorhabditis elegans/genética , Línea Celular , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/enzimología , Corteza Cerebral/patología , Diglicéridos/metabolismo , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/enzimología , Neuronas Dopaminérgicas/patología , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/enzimología , Células Madre Pluripotentes Inducidas/patología , Gotas Lipídicas/efectos de los fármacos , Gotas Lipídicas/enzimología , Ratones Endogámicos C57BL , Ratones Transgénicos , Terapia Molecular Dirigida , Degeneración Nerviosa , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/enzimología , Células-Madre Neurales/patología , Neuronas/enzimología , Neuronas/patología , Ácido Oléico/metabolismo , Enfermedad de Parkinson/enzimología , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Ratas Sprague-Dawley , Saccharomyces cerevisiae/efectos de los fármacos , Saccharomyces cerevisiae/enzimología , Saccharomyces cerevisiae/genética , Estearoil-CoA Desaturasa/metabolismo , Triglicéridos/metabolismo , alfa-Sinucleína/genética
10.
J Cell Sci ; 131(13)2018 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-29792311

RESUMEN

Prostate tumors make metabolic adaptations to ensure adequate energy and amplify cell cycle regulators, such as centrosomes, to sustain their proliferative capacity. It is not known whether cancer-associated fibroblasts (CAFs) undergo metabolic re-programming. We postulated that CAFs augment lipid storage and amplify centrosomal or non-centrosomal microtubule-organizing centers (MTOCs) through a pigment epithelium-derived factor (PEDF)-dependent lipid-MTOC signaling axis. Primary human normal prostate fibroblasts (NFs) and CAFs were evaluated for lipid content, triacylglycerol-regulating proteins, MTOC number and distribution. CAFs were found to store more neutral lipids than NFs. Adipose triglyceride lipase (ATGL) and PEDF were strongly expressed in NFs, whereas CAFs had minimal to undetectable levels of PEDF or ATGL protein. At baseline, CAFs demonstrated MTOC amplification when compared to 1-2 perinuclear MTOCs consistently observed in NFs. Treatment with PEDF or blockade of lipogenesis suppressed lipid content and MTOC number. In summary, our data support that CAFs have acquired a tumor-like phenotype by re-programming lipid metabolism and amplifying MTOCs. Normalization of MTOCs by restoring PEDF or by blocking lipogenesis highlights a previously unrecognized plasticity in centrosomes, which is regulated through a new lipid-MTOC axis.This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Fibroblastos Asociados al Cáncer/metabolismo , Proteínas del Ojo/metabolismo , Metabolismo de los Lípidos , Centro Organizador de los Microtúbulos/metabolismo , Factores de Crecimiento Nervioso/metabolismo , Neoplasias de la Próstata/metabolismo , Serpinas/metabolismo , Proteínas del Ojo/genética , Fibroblastos/metabolismo , Humanos , Lipasa/genética , Lipasa/metabolismo , Lipogénesis , Masculino , Factores de Crecimiento Nervioso/genética , Próstata/metabolismo , Neoplasias de la Próstata/genética , Serpinas/genética , Triglicéridos/metabolismo
11.
Dev Cell ; 45(4): 427-432, 2018 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-29787708

RESUMEN

Lipid droplets (LDs) are ubiquitous fat storage organelles and play key roles in lipid metabolism and energy homeostasis; in addition, they contribute to protein storage, folding, and degradation. However, a role for LDs in the nervous system remains largely unexplored. We discuss evidence supporting an intimate functional connection between LDs and motor neuron disease (MND) pathophysiology, examining how LD functions in systemic energy homeostasis, in neuron-glia metabolic coupling, and in protein folding and clearance may affect or contribute to disease pathology. An integrated understanding of LD biology and neurodegeneration may open the way for new therapeutic interventions.


Asunto(s)
Gotas Lipídicas/fisiología , Lípidos/química , Enfermedad de la Neurona Motora/fisiopatología , Orgánulos/metabolismo , Homeostasis , Humanos , Metabolismo de los Lípidos , Lipólisis
12.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1862(10 Pt B): 1260-1272, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28735096

RESUMEN

Lipid droplets are cytoplasmic organelles that store neutral lipids and are critically important for energy metabolism. Their function in energy storage is firmly established and increasingly well characterized. However, emerging evidence indicates that lipid droplets also play important and diverse roles in the cellular handling of lipids and proteins that may not be directly related to energy homeostasis. Lipid handling roles of droplets include the storage of hydrophobic vitamin and signaling precursors, and the management of endoplasmic reticulum and oxidative stress. Roles of lipid droplets in protein handling encompass functions in the maturation, storage, and turnover of cellular and viral polypeptides. Other potential roles of lipid droplets may be connected with their intracellular motility and, in some cases, their nuclear localization. This diversity highlights that lipid droplets are very adaptable organelles, performing different functions in different biological contexts. This article is part of a Special Issue entitled: Recent Advances in Lipid Droplet Biology edited by Rosalind Coleman and Matthijs Hesselink.


Asunto(s)
Estrés del Retículo Endoplásmico , Metabolismo Energético , Gotas Lipídicas/metabolismo , Metabolismo de los Lípidos , Estrés Oxidativo , Animales , Humanos
13.
J Cell Sci ; 129(7): 1416-28, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26906417

RESUMEN

During bidirectional transport, individual cargoes move continuously back and forth along microtubule tracks, yet the cargo population overall displays directed net transport. How such transport is controlled temporally is not well understood. We analyzed this issue for bidirectionally moving lipid droplets in Drosophila embryos, a system in which net transport direction is developmentally controlled. By quantifying how the droplet distribution changes as embryos develop, we characterize temporal transitions in net droplet transport and identify the crucial contribution of the previously identified, but poorly characterized, transacting regulator Halo. In particular, we find that Halo is transiently expressed; rising and falling Halo levels control the switches in global distribution. Rising Halo levels have to pass a threshold before net plus-end transport is initiated. This threshold level depends on the amount of the motor kinesin-1: the more kinesin-1 is present, the more Halo is needed before net plus-end transport commences. Because Halo and kinesin-1 are present in common protein complexes, we propose that Halo acts as a rate-limiting co-factor of kinesin-1.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/embriología , Cinesinas/metabolismo , Gotas Lipídicas/metabolismo , Animales , Animales Modificados Genéticamente , Transporte Biológico , Drosophila melanogaster/metabolismo
14.
Cell ; 163(2): 269-70, 2015 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-26451474

RESUMEN

A new paper by Bailey et al. reveals that lipid droplets, crucial organelles for energy storage, can also protect against oxidative stress. In Drosophila larvae, lipid droplets in glia allow neuronal stem cells to keep proliferating under hypoxic conditions. Protection likely involves sequestering vulnerable membrane lipids away from reactive oxygen species.


Asunto(s)
Drosophila/citología , Drosophila/metabolismo , Gotas Lipídicas/metabolismo , Nicho de Células Madre/efectos de los fármacos , Animales
15.
Curr Biol ; 25(11): R470-81, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-26035793

RESUMEN

Lipid droplets are the intracellular sites for neutral lipid storage. They are critical for lipid metabolism and energy homeostasis, and their dysfunction has been linked to many diseases. Accumulating evidence suggests that the roles lipid droplets play in biology are significantly broader than previously anticipated. Lipid droplets are the source of molecules important in the nucleus: they can sequester transcription factors and chromatin components and generate the lipid ligands for certain nuclear receptors. Lipid droplets have also emerged as important nodes for fatty acid trafficking, both inside the cell and between cells. In immunity, new roles for droplets, not directly linked to lipid metabolism, have been uncovered, with evidence that they act as assembly platforms for specific viruses and as reservoirs for proteins that fight intracellular pathogens. Until recently, knowledge about droplets in the nervous system has been minimal, but now there are multiple links between lipid droplets and neurodegeneration: many candidate genes for hereditary spastic paraplegia also have central roles in lipid-droplet formation and maintenance, and mitochondrial dysfunction in neurons can lead to transient accumulation of lipid droplets in neighboring glial cells, an event that may, in turn, contribute to neuronal damage. As the cell biology and biochemistry of lipid droplets become increasingly well understood, the next few years should yield many new mechanistic insights into these novel functions of lipid droplets.


Asunto(s)
Gotas Lipídicas/fisiología , Animales , Núcleo Celular/metabolismo , Ácidos Grasos/metabolismo , Humanos , Mitocondrias/metabolismo , Neuroglía/metabolismo , Proteínas/metabolismo , Paraplejía Espástica Hereditaria/metabolismo , Ensamble de Virus
16.
Biochim Biophys Acta ; 1851(9): 1156-85, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25882628

RESUMEN

Research into lipid droplets is rapidly expanding, and new cellular and organismal roles for these lipid-storage organelles are continually being discovered. The early Drosophila embryo is particularly well suited for addressing certain questions in lipid-droplet biology and combines technical advantages with unique biological phenomena. This review summarizes key features of this experimental system and the techniques available to study it, in order to make it accessible to researchers outside this field. It then describes the two topics most heavily studied in this system, lipid-droplet motility and protein sequestration on droplets, discusses what is known about the molecular players involved, points to open questions, and compares the results from Drosophila embryo studies to what it is known about lipid droplets in other systems.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Embrión no Mamífero/metabolismo , Gotas Lipídicas/metabolismo , Microtúbulos/metabolismo , Proteínas Motoras Moleculares/metabolismo , Animales , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/crecimiento & desarrollo , Embrión no Mamífero/citología , Regulación del Desarrollo de la Expresión Génica , Gotas Lipídicas/química , Metabolismo de los Lípidos , Microtúbulos/química , Microtúbulos/genética , Proteínas Motoras Moleculares/genética , Transducción de Señal , Grabación en Video
17.
J Cell Biol ; 206(2): 199-215, 2014 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-25049271

RESUMEN

Communication usually applies feedback loop-based filters and amplifiers to ensure undistorted delivery of messages. Such an amplifier acts during Drosophila melanogaster midoogenesis, when oskar messenger ribonucleic acid (mRNA) anchoring depends on its own locally translated protein product. We find that the motor regulator Klar ß mediates a gain-control process that prevents saturation-based distortions in this positive feedback loop. We demonstrate that, like oskar mRNA, Klar ß localizes to the posterior pole of oocytes in a kinesin-1-dependent manner. By live imaging and semiquantitative fluorescent in situ hybridization, we show that Klar ß restrains oskar ribonucleoprotein motility and decreases the posterior-ward translocation of oskar mRNA, thereby adapting the rate of oskar delivery to the output of the anchoring machinery. This negative regulatory effect of Klar is particularly important for overriding temperature-induced changes in motility. We conclude that by preventing defects in oskar anchoring, this mechanism contributes to the developmental robustness of a poikilothermic organism living in a variable temperature environment.


Asunto(s)
Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/fisiología , Drosophila melanogaster/metabolismo , Proteínas de Transporte de Membrana/fisiología , Temperatura , Animales , Polaridad Celular , Proteínas de Drosophila/análisis , Proteínas de Drosophila/genética , Drosophila melanogaster/embriología , Retroalimentación Fisiológica , Hibridación Fluorescente in Situ , Cinesinas/metabolismo , Cinesinas/fisiología , Proteínas de Transporte de Membrana/análisis , Proteínas de Transporte de Membrana/metabolismo , Oocitos/metabolismo , Transporte de Proteínas , ARN Mensajero/análisis , ARN Mensajero/metabolismo , Ribonucleoproteínas/metabolismo
18.
Curr Biol ; 24(13): 1485-91, 2014 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-24930966

RESUMEN

Assembly of DNA into chromatin requires a delicate balancing act, as both dearth and excess of histones severely disrupt chromatin function [1-3]. In particular, cells need to carefully control histone stoichiometry: if different types of histones are incorporated into chromatin in an imbalanced manner, it can lead to altered gene expression, mitotic errors, and death [4-6]. Both the balance between individual core histones and the balance between core histones and histone variants are critical [5, 7]. Here, we find that in early Drosophila embryos, histone balance in the nuclei is regulated by lipid droplets, cytoplasmic fat-storage organelles [8]. Lipid droplets were previously known to function in long-term histone storage: newly laid embryos contain large amounts of excess histones generated during oogenesis [9], and the maternal supplies of core histone H2A and the histone variant H2Av are anchored to lipid droplets via the novel protein Jabba [3]. We find that in these embryos, synthesis of new H2A and H2Av is imbalanced, and that newly produced H2Av can be recruited to lipid droplets. When droplet sequestration is disrupted by mutating Jabba, embryos display an elevated H2Av/H2A ratio in nuclei as well as mitotic defects, reduced viability, and hypersensitivity to H2Av overexpression. We propose that in Drosophila embryos, lipid droplets serve as a histone buffer, not only storing maternal histones to support the early cell cycles but also transiently sequestering H2Av produced in excess and thus ensuring proper histone balance in the nucleus.


Asunto(s)
Ensamble y Desensamble de Cromatina/fisiología , Drosophila/embriología , Histonas/metabolismo , Gotas Lipídicas/metabolismo , Animales , Western Blotting , Proteínas Portadoras/metabolismo , Proteínas de Drosophila/metabolismo , Modelos Biológicos
19.
Genes Dev ; 28(2): 167-81, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24402316

RESUMEN

The transcription factors of the Snail family are key regulators of epithelial-mesenchymal transitions, cell morphogenesis, and tumor metastasis. Since its discovery in Drosophila ∼25 years ago, Snail has been extensively studied for its role as a transcriptional repressor. Here we demonstrate that Drosophila Snail can positively modulate transcriptional activation. By combining information on in vivo occupancy with expression profiling of hand-selected, staged snail mutant embryos, we identified 106 genes that are potentially directly regulated by Snail during mesoderm development. In addition to the expected Snail-repressed genes, almost 50% of Snail targets showed an unanticipated activation. The majority of "Snail-activated" genes have enhancer elements cobound by Twist and are expressed in the mesoderm at the stages of Snail occupancy. Snail can potentiate Twist-mediated enhancer activation in vitro and is essential for enhancer activity in vivo. Using a machine learning approach, we show that differentially enriched motifs are sufficient to predict Snail's regulatory response. In silico mutagenesis revealed a likely causative motif, which we demonstrate is essential for enhancer activation. Taken together, these data indicate that Snail can potentiate enhancer activation by collaborating with different activators, providing a new mechanism by which Snail regulates development.


Asunto(s)
Drosophila/genética , Drosophila/metabolismo , Factores de Transcripción/metabolismo , Secuencias de Aminoácidos , Animales , Drosophila/embriología , Proteínas de Drosophila/metabolismo , Embrión no Mamífero , Elementos de Facilitación Genéticos/genética , Regulación del Desarrollo de la Expresión Génica , Mesodermo/metabolismo , Unión Proteica , Factores de Transcripción de la Familia Snail , Factores de Transcripción/genética , Proteína 1 Relacionada con Twist/metabolismo
20.
Elife ; 1: e00003, 2012 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-23150794

RESUMEN

We previously discovered histones bound to cytosolic lipid droplets (LDs); here we show that this forms a cellular antibacterial defense system. Sequestered on droplets under normal conditions, in the presence of bacterial lipopolysaccharide (LPS) or lipoteichoic acid (LTA), histones are released from the droplets and kill bacteria efficiently in vitro. Droplet-bound histones also function in vivo: when injected into Drosophila embryos lacking droplet-bound histones, bacteria grow rapidly. In contrast, bacteria injected into embryos with droplet-bound histones die. Embryos with droplet-bound histones displayed more than a fourfold survival advantage when challenged with four different bacterial species. Our data suggests that this intracellular antibacterial defense system may function in adult flies, and also potentially in mice.DOI:http://dx.doi.org/10.7554/eLife.00003.001.


Asunto(s)
Drosophila melanogaster/inmunología , Embrión no Mamífero/inmunología , Histonas/inmunología , Gotas Lipídicas/inmunología , Hígado/inmunología , Animales , Bacillus subtilis/efectos de los fármacos , Bacillus subtilis/crecimiento & desarrollo , Drosophila melanogaster/metabolismo , Drosophila melanogaster/microbiología , Embrión no Mamífero/efectos de los fármacos , Embrión no Mamífero/metabolismo , Embrión no Mamífero/microbiología , Escherichia coli/efectos de los fármacos , Escherichia coli/crecimiento & desarrollo , Histonas/metabolismo , Histonas/farmacología , Gotas Lipídicas/química , Gotas Lipídicas/metabolismo , Lipopolisacáridos/farmacología , Listeria monocytogenes/efectos de los fármacos , Listeria monocytogenes/crecimiento & desarrollo , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/microbiología , Ratones , Ratones Endogámicos C57BL , Staphylococcus epidermidis/efectos de los fármacos , Staphylococcus epidermidis/crecimiento & desarrollo , Ácidos Teicoicos/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...