Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Cell ; 163(2): 340-53, 2015 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-26451484

RESUMEN

Stem cells reside in specialized microenvironments known as niches. During Drosophila development, glial cells provide a niche that sustains the proliferation of neural stem cells (neuroblasts) during starvation. We now find that the glial cell niche also preserves neuroblast proliferation under conditions of hypoxia and oxidative stress. Lipid droplets that form in niche glia during oxidative stress limit the levels of reactive oxygen species (ROS) and inhibit the oxidation of polyunsaturated fatty acids (PUFAs). These droplets protect glia and also neuroblasts from peroxidation chain reactions that can damage many types of macromolecules. The underlying antioxidant mechanism involves diverting PUFAs, including diet-derived linoleic acid, away from membranes to the core of lipid droplets, where they are less vulnerable to peroxidation. This study reveals an antioxidant role for lipid droplets that could be relevant in many different biological contexts.


Asunto(s)
Drosophila/citología , Drosophila/metabolismo , Gotas Lipídicas/metabolismo , Nicho de Células Madre/efectos de los fármacos , Animales , Antioxidantes/farmacología , Proliferación Celular , Drosophila/crecimiento & desarrollo , Ácidos Grasos Insaturados/farmacología , Larva/citología , Larva/crecimiento & desarrollo , Larva/metabolismo , Neuroglía/metabolismo , Estrés Oxidativo , Oxígeno/metabolismo , Células Madre/citología , Células Madre/efectos de los fármacos
2.
Nature ; 615(7953): 705-711, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36922598

RESUMEN

Artificial sweeteners are used as calorie-free sugar substitutes in many food products and their consumption has increased substantially over the past years1. Although generally regarded as safe, some concerns have been raised about the long-term safety of the consumption of certain sweeteners2-5. In this study, we show that the intake of high doses of sucralose in mice results in immunomodulatory effects by limiting T cell proliferation and T cell differentiation. Mechanistically, sucralose affects the membrane order of T cells, accompanied by a reduced efficiency of T cell receptor signalling and intracellular calcium mobilization. Mice given sucralose show decreased CD8+ T cell antigen-specific responses in subcutaneous cancer models and bacterial infection models, and reduced T cell function in models of T cell-mediated autoimmunity. Overall, these findings suggest that a high intake of sucralose can dampen T cell-mediated responses, an effect that could be used in therapy to mitigate T cell-dependent autoimmune disorders.


Asunto(s)
Sacarosa , Edulcorantes , Linfocitos T , Animales , Ratones , Sacarosa/análogos & derivados , Edulcorantes/administración & dosificación , Edulcorantes/efectos adversos , Edulcorantes/farmacología , Edulcorantes/uso terapéutico , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/patología , Inocuidad de los Alimentos , Señalización del Calcio/efectos de los fármacos , Receptores de Antígenos de Linfocitos T/efectos de los fármacos , Receptores de Antígenos de Linfocitos T/inmunología , Infecciones Bacterianas/inmunología , Neoplasias/inmunología , Autoinmunidad/efectos de los fármacos , Autoinmunidad/inmunología , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología
3.
Cell ; 146(3): 435-47, 2011 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-21816278

RESUMEN

Developing animals survive periods of starvation by protecting the growth of critical organs at the expense of other tissues. Here, we use Drosophila to explore the as yet unknown mechanisms regulating this privileged tissue growth. As in mammals, we observe in Drosophila that the CNS is more highly spared than other tissues during nutrient restriction (NR). We demonstrate that anaplastic lymphoma kinase (Alk) efficiently protects neural progenitor (neuroblast) growth against reductions in amino acids and insulin-like peptides during NR via two mechanisms. First, Alk suppresses the growth requirement for amino acid sensing via Slimfast/Rheb/TOR complex 1. And second, Alk, rather than insulin-like receptor, primarily activates PI3-kinase. Alk maintains PI3-kinase signaling during NR as its ligand, Jelly belly (Jeb), is constitutively expressed from a glial cell niche surrounding neuroblasts. Together, these findings identify a brain-sparing mechanism that shares some regulatory features with the starvation-resistant growth programs of mammalian tumors.


Asunto(s)
Drosophila melanogaster/crecimiento & desarrollo , Drosophila melanogaster/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Quinasa de Linfoma Anaplásico , Animales , Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Sistema Nervioso Central/crecimiento & desarrollo , Sistema Nervioso Central/metabolismo , Privación de Alimentos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Larva/crecimiento & desarrollo , Larva/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Poliploidía
4.
PLoS Biol ; 19(5): e3001230, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33945525

RESUMEN

Obesity-related renal lipotoxicity and chronic kidney disease (CKD) are prevalent pathologies with complex aetiologies. One hallmark of renal lipotoxicity is the ectopic accumulation of lipid droplets in kidney podocytes and in proximal tubule cells. Renal lipid droplets are observed in human CKD patients and in high-fat diet (HFD) rodent models, but their precise role remains unclear. Here, we establish a HFD model in Drosophila that recapitulates renal lipid droplets and several other aspects of mammalian CKD. Cell type-specific genetic manipulations show that lipid can overflow from adipose tissue and is taken up by renal cells called nephrocytes. A HFD drives nephrocyte lipid uptake via the multiligand receptor Cubilin (Cubn), leading to the ectopic accumulation of lipid droplets. These nephrocyte lipid droplets correlate with endoplasmic reticulum (ER) and mitochondrial deficits, as well as with impaired macromolecular endocytosis, a key conserved function of renal cells. Nephrocyte knockdown of diglyceride acyltransferase 1 (DGAT1), overexpression of adipose triglyceride lipase (ATGL), and epistasis tests together reveal that fatty acid flux through the lipid droplet triglyceride compartment protects the ER, mitochondria, and endocytosis of renal cells. Strikingly, boosting nephrocyte expression of the lipid droplet resident enzyme ATGL is sufficient to rescue HFD-induced defects in renal endocytosis. Moreover, endocytic rescue requires a conserved mitochondrial regulator, peroxisome proliferator-activated receptor-gamma coactivator 1α (PGC1α). This study demonstrates that lipid droplet lipolysis counteracts the harmful effects of a HFD via a mitochondrial pathway that protects renal endocytosis. It also provides a genetic strategy for determining whether lipid droplets in different biological contexts function primarily to release beneficial or to sequester toxic lipids.


Asunto(s)
Lipasa/metabolismo , Gotas Lipídicas/metabolismo , Insuficiencia Renal Crónica/metabolismo , Tejido Adiposo/metabolismo , Animales , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Endocitosis/fisiología , Células Epiteliales/metabolismo , Ácidos Grasos/metabolismo , Humanos , Riñón/patología , Lipasa/fisiología , Gotas Lipídicas/fisiología , Metabolismo de los Lípidos/fisiología , Lípidos/fisiología , Mitocondrias/metabolismo , Obesidad/complicaciones , Insuficiencia Renal Crónica/fisiopatología , Triglicéridos/metabolismo
5.
EMBO J ; 38(7)2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30804004

RESUMEN

Rewired metabolism of glutamine in cancer has been well documented, but less is known about other amino acids such as histidine. Here, we use Drosophila cancer models to show that decreasing the concentration of histidine in the diet strongly inhibits the growth of mutant clones induced by loss of Nerfin-1 or gain of Notch activity. In contrast, changes in dietary histidine have much less effect on the growth of wildtype neural stem cells and Prospero neural tumours. The reliance of tumours on dietary histidine and also on histidine decarboxylase (Hdc) depends upon their growth requirement for Myc. We demonstrate that Myc overexpression in nerfin-1 tumours is sufficient to switch their mode of growth from histidine/Hdc sensitive to resistant. This study suggests that perturbations in histidine metabolism selectively target neural tumours that grow via a dedifferentiation process involving large cell size increases driven by Myc.


Asunto(s)
Desdiferenciación Celular , Neoplasias del Sistema Nervioso Central/patología , Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Histidina/administración & dosificación , Células-Madre Neurales/patología , Factores de Transcripción/metabolismo , Animales , Neoplasias del Sistema Nervioso Central/genética , Neoplasias del Sistema Nervioso Central/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/efectos de los fármacos , Drosophila melanogaster/genética , Drosophila melanogaster/crecimiento & desarrollo , Femenino , Histidina Descarboxilasa/genética , Histidina Descarboxilasa/metabolismo , Masculino , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/metabolismo , Factores de Transcripción/genética
6.
Cell ; 133(5): 891-902, 2008 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-18510932

RESUMEN

The timing mechanisms responsible for terminating cell proliferation toward the end of development remain unclear. In the Drosophila CNS, individual progenitors called neuroblasts are known to express a series of transcription factors endowing daughter neurons with different temporal identities. Here we show that Castor and Seven-Up, members of this temporal series, regulate key events in many different neuroblast lineages during late neurogenesis. First, they schedule a switch in the cell size and identity of neurons involving the targets Chinmo and Broad Complex. Second, they regulate the time at which neuroblasts undergo Prospero-dependent cell-cycle exit or Reaper/Hid/Grim-dependent apoptosis. Both types of progenitor termination require the combined action of a late phase of the temporal series and indirect feedforward via Castor targets such as Grainyhead and Dichaete. These studies identify the timing mechanism ending CNS proliferation and reveal how aging progenitors transduce bursts of transcription factors into long-lasting changes in cell proliferation and cell identity.


Asunto(s)
Proliferación Celular , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citología , Neuronas/citología , Células Madre/citología , Factores de Transcripción/metabolismo , Animales , Apoptosis , Ciclo Celular , Diferenciación Celular , Tamaño de la Célula , Proteínas de Unión al ADN/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas del Grupo de Alta Movilidad/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptores de Esteroides/metabolismo , Factores de Transcripción SOX
7.
PLoS Biol ; 15(10): e2002252, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28976974

RESUMEN

Sexual dimorphisms in body size are widespread throughout the animal kingdom but their underlying mechanisms are not well characterized. Most models for how sex chromosome genes specify size dimorphism have emphasized the importance of gonadal hormones and cell-autonomous influences in mammals versus strictly cell-autonomous mechanisms in Drosophila melanogaster. Here, we use tissue-specific genetics to investigate how sexual size dimorphism (SSD) is established in Drosophila. We find that the larger body size characteristic of Drosophila females is established very early in larval development via an increase in the growth rate per unit of body mass. We demonstrate that the female sex determination gene, Sex-lethal (Sxl), functions in central nervous system (CNS) neurons as part of a relay that specifies the early sex-specific growth trajectories of larval but not imaginal tissues. Neuronal Sxl acts additively in 2 neuronal subpopulations, one of which corresponds to 7 median neurosecretory cells: the insulin-producing cells (IPCs). Surprisingly, however, male-female differences in the production of insulin-like peptides (Ilps) from the IPCs do not appear to be involved in establishing SSD in early larvae, although they may play a later role. These findings support a relay model in which Sxl in neurons and Sxl in local tissues act together to specify the female-specific growth of the larval body. They also reveal that, even though the sex determination pathways in Drosophila and mammals are different, they both modulate body growth via a combination of tissue-autonomous and nonautonomous inputs.


Asunto(s)
Drosophila/crecimiento & desarrollo , Neuronas/fisiología , Procesos de Determinación del Sexo/genética , Animales , Tamaño Corporal/genética , Drosophila/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/fisiología , Ingestión de Alimentos , Femenino , Neuronas GABAérgicas/fisiología , Células Secretoras de Insulina/metabolismo , Larva , Masculino , Neuropéptidos , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/fisiología , Caracteres Sexuales
8.
Angew Chem Int Ed Engl ; 59(41): 18194-18200, 2020 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-32603009

RESUMEN

OrbiSIMS is a recently developed instrument for label-free imaging of chemicals with micron spatial resolution and high mass resolution. We report a cryogenic workflow for OrbiSIMS (Cryo-OrbiSIMS) that improves chemical detection of lipids and other biomolecules in tissues. Cryo-OrbiSIMS boosts ionization yield and decreases ion-beam induced fragmentation, greatly improving the detection of biomolecules such as triacylglycerides. It also increases chemical coverage to include molecules with intermediate or high vapor pressures, such as free fatty acids and semi-volatile organic compounds (SVOCs). We find that Cryo-OrbiSIMS reveals the hitherto unknown localization patterns of SVOCs with high spatial and chemical resolution in diverse plant, animal, and human tissues. We also show that Cryo-OrbiSIMS can be combined with genetic analysis to identify enzymes regulating SVOC metabolism. Cryo-OrbiSIMS is applicable to high resolution imaging of a wide variety of non-volatile and semi-volatile molecules across many areas of biomedicine.


Asunto(s)
Espectrometría de Masas/métodos , Compuestos Orgánicos Volátiles/análisis , Frío , Historia del Siglo XV
9.
J Proteome Res ; 18(4): 1503-1512, 2019 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-30757904

RESUMEN

The measurement of absolute metabolite concentrations in small samples remains a significant analytical challenge. This is particularly the case when the sample volume is only a few microliters or less and cannot be determined accurately via direct measurement. We previously developed volume determination with two standards (VDTS) as a method to address this challenge for biofluids. As a proof-of-principle, we applied VDTS to NMR spectra of polar metabolites in the hemolymph (blood) of the tiny yet powerful genetic model Drosophila melanogaster. This showed that VDTS calculation of absolute metabolite concentrations in fed versus starved Drosophila larvae is more accurate than methods utilizing normalization to total spectral signal. Here, we introduce paired VDTS (pVDTS), an improved VDTS method for biofluids and solid tissues that implements the statistical power of paired control and experimental replicates. pVDTS utilizes new equations that also include a correction for dilution errors introduced by the variable surface wetness of solid samples. We then show that metabolite concentrations in Drosophila larvae are more precisely determined and logically consistent using pVDTS than using the original VDTS method. The refined pVDTS workflow described in this study is applicable to a wide range of different tissues and biofluids.


Asunto(s)
Metaboloma/fisiología , Metabolómica/métodos , Aminoácidos/análisis , Animales , Carbohidratos/análisis , Ácidos Carboxílicos/análisis , Drosophila melanogaster/química , Drosophila melanogaster/metabolismo , Femenino , Hemolinfa/química , Hemolinfa/metabolismo , Larva/química , Larva/metabolismo , Espectroscopía de Resonancia Magnética , Masculino
10.
PLoS Genet ; 12(8): e1006154, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27500738

RESUMEN

Cell growth and proliferation depend upon many different aspects of lipid metabolism. One key signaling pathway that is utilized in many different anabolic contexts involves Phosphatidylinositide 3-kinase (PI3K) and its membrane lipid products, the Phosphatidylinositol (3,4,5)-trisphosphates. It remains unclear, however, which other branches of lipid metabolism interact with the PI3K signaling pathway. Here, we focus on specialized fat metabolizing cells in Drosophila called larval oenocytes. In the presence of dietary nutrients, oenocytes undergo PI3K-dependent cell growth and contain very few lipid droplets. In contrast, during starvation, oenocytes decrease PI3K signaling, shut down cell growth and accumulate abundant lipid droplets. We now show that PI3K in larval oenocytes, but not in fat body cells, functions to suppress lipid droplet accumulation. Several enzymes of fatty acid, triglyceride and hydrocarbon metabolism are required in oenocytes primarily for lipid droplet induction rather than for cell growth. In contrast, a very long chain fatty-acyl-CoA reductase (FarO) and a putative lipid dehydrogenase/reductase (Spidey, also known as Kar) not only promote lipid droplet induction but also inhibit oenocyte growth. In the case of Spidey/Kar, we show that the growth suppression mechanism involves inhibition of the PI3K signaling pathway upstream of Akt activity. Together, the findings in this study show how Spidey/Kar and FarO regulate the balance between the cell growth and lipid storage of larval oenocytes.


Asunto(s)
Acil-CoA Deshidrogenasa/genética , Proteínas de Drosophila/genética , Metabolismo de los Lípidos/genética , Oxidorreductasas/genética , Fosfatidilinositol 3-Quinasas/genética , Acil-CoA Deshidrogenasa/metabolismo , Animales , Proliferación Celular/genética , Drosophila/genética , Drosophila/crecimiento & desarrollo , Drosophila/metabolismo , Cuerpo Adiposo/crecimiento & desarrollo , Cuerpo Adiposo/metabolismo , Larva/genética , Larva/metabolismo , Gotas Lipídicas/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Triglicéridos/metabolismo
11.
Nature ; 481(7382): 516-9, 2012 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-22246326

RESUMEN

Mass spectrometry with stable isotope labels has been seminal in discovering the dynamic state of living matter, but is limited to bulk tissues or cells. We developed multi-isotope imaging mass spectrometry (MIMS) that allowed us to view and measure stable isotope incorporation with submicrometre resolution. Here we apply MIMS to diverse organisms, including Drosophila, mice and humans. We test the 'immortal strand hypothesis', which predicts that during asymmetric stem cell division chromosomes containing older template DNA are segregated to the daughter destined to remain a stem cell, thus insuring lifetime genetic stability. After labelling mice with (15)N-thymidine from gestation until post-natal week 8, we find no (15)N label retention by dividing small intestinal crypt cells after a four-week chase. In adult mice administered (15)N-thymidine pulse-chase, we find that proliferating crypt cells dilute the (15)N label, consistent with random strand segregation. We demonstrate the broad utility of MIMS with proof-of-principle studies of lipid turnover in Drosophila and translation to the human haematopoietic system. These studies show that MIMS provides high-resolution quantification of stable isotope labels that cannot be obtained using other techniques and that is broadly applicable to biological and medical research.


Asunto(s)
División Celular , Espectrometría de Masas/métodos , Células Madre/citología , Células Madre/metabolismo , Animales , Animales Recién Nacidos , ADN/biosíntesis , ADN/genética , ADN/metabolismo , Drosophila melanogaster/citología , Enterocitos/citología , Fibroblastos/citología , Humanos , Intestino Delgado/citología , Marcaje Isotópico , Isótopos , Leucocitos/citología , Metabolismo de los Lípidos , Linfopoyesis , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Células Madre/patología , Moldes Genéticos , Timidina/metabolismo
12.
Development ; 141(7): 1492-502, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24574009

RESUMEN

Hox genes encode a conserved family of homeodomain transcription factors regulating development along the major body axis. During embryogenesis, Hox proteins are expressed in segment-specific patterns and control numerous different segment-specific cell fates. It has been unclear, however, whether Hox proteins drive the epithelial cell segregation mechanism that is thought to initiate the segmentation process. Here, we investigate the role of vertebrate Hox proteins during the partitioning of the developing hindbrain into lineage-restricted units called rhombomeres. Loss-of-function mutants and ectopic expression assays reveal that Hoxb4 and its paralogue Hoxd4 are necessary and sufficient for cell segregation, and for the most caudal rhombomere boundary (r6/r7). Hox4 proteins regulate Eph/ephrins and other cell-surface proteins, and can function in a non-cell-autonomous manner to induce apical cell enlargement on both sides of their expression border. Similarly, other Hox proteins expressed at more rostral rhombomere interfaces can also regulate Eph/ephrins, induce apical remodelling and drive cell segregation in ectopic expression assays. However, Krox20, a key segmentation factor expressed in odd rhombomeres (r3 and r5), can largely override Hox proteins at the level of regulation of a cell surface target, Epha4. This study suggests that most, if not all, Hox proteins share a common potential to induce cell segregation but in some contexts this is masked or modulated by other transcription factors.


Asunto(s)
Tipificación del Cuerpo/genética , Movimiento Celular/genética , Proteínas de Homeodominio/fisiología , Rombencéfalo/embriología , Animales , Animales Modificados Genéticamente , Embrión de Pollo , Embrión de Mamíferos , Femenino , Regulación del Desarrollo de la Expresión Génica , Genes Homeobox/fisiología , Proteínas de la Membrana/genética , Ratones , Rombencéfalo/metabolismo , Factores de Transcripción/fisiología
13.
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
14.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1862(8): 792-796, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28302590

RESUMEN

Metabolic pathway flux is a fundamental element of biological activity, which can be quantified using a variety of mass spectrometric techniques to monitor incorporation of stable isotope-labelled substrates into metabolic products. This article contrasts developments in electrospray ionisation mass spectrometry (ESI-MS) for the measurement of lipid metabolism with more established gas chromatography mass spectrometry and isotope ratio mass spectrometry methodologies. ESI-MS combined with diagnostic tandem MS/MS scans permits the sensitive and specific analysis of stable isotope-labelled substrates into intact lipid molecular species without the requirement for lipid hydrolysis and derivatisation. Such dynamic lipidomic methodologies using non-toxic stable isotopes can be readily applied to quantify lipid metabolic fluxes in clinical and metabolic studies in vivo. However, a significant current limitation is the absence of appropriate software to generate kinetic models of substrate incorporation into multiple products in the time domain. Finally, we discuss the future potential of stable isotope-mass spectrometry imaging to quantify the location as well as the extent of lipid synthesis. This article is part of a Special Issue entitled: BBALIP_Lipidomics Opinion Articles edited by Sepp Kohlwein.


Asunto(s)
Isótopos/química , Metabolismo de los Lípidos/fisiología , Lípidos/química , Animales , Humanos , Marcaje Isotópico/métodos , Redes y Vías Metabólicas/fisiología , Espectrometría de Masa por Ionización de Electrospray/métodos , Espectrometría de Masas en Tándem/métodos
15.
Nature ; 471(7339): 508-12, 2011 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-21346761

RESUMEN

Many stem, progenitor and cancer cells undergo periods of mitotic quiescence from which they can be reactivated. The signals triggering entry into and exit from this reversible dormant state are not well understood. In the developing Drosophila central nervous system, multipotent self-renewing progenitors called neuroblasts undergo quiescence in a stereotypical spatiotemporal pattern. Entry into quiescence is regulated by Hox proteins and an internal neuroblast timer. Exit from quiescence (reactivation) is subject to a nutritional checkpoint requiring dietary amino acids. Organ co-cultures also implicate an unidentified signal from an adipose/hepatic-like tissue called the fat body. Here we provide in vivo evidence that Slimfast amino-acid sensing and Target of rapamycin (TOR) signalling activate a fat-body-derived signal (FDS) required for neuroblast reactivation. Downstream of this signal, Insulin-like receptor signalling and the Phosphatidylinositol 3-kinase (PI3K)/TOR network are required in neuroblasts for exit from quiescence. We demonstrate that nutritionally regulated glial cells provide the source of Insulin-like peptides (ILPs) relevant for timely neuroblast reactivation but not for overall larval growth. Conversely, ILPs secreted into the haemolymph by median neurosecretory cells systemically control organismal size but do not reactivate neuroblasts. Drosophila thus contains two segregated ILP pools, one regulating proliferation within the central nervous system and the other controlling tissue growth systemically. Our findings support a model in which amino acids trigger the cell cycle re-entry of neural progenitors via a fat-body-glia-neuroblasts relay. This mechanism indicates that dietary nutrients and remote organs, as well as local niches, are key regulators of transitions in stem-cell behaviour.


Asunto(s)
Adipocitos/metabolismo , Drosophila melanogaster/metabolismo , Insulina/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neuroglía/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Adipocitos/efectos de los fármacos , Aminoácidos/farmacología , Animales , Sistema Nervioso Central/citología , Sistema Nervioso Central/efectos de los fármacos , Sistema Nervioso Central/metabolismo , Dieta , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citología , Drosophila melanogaster/efectos de los fármacos , Drosophila melanogaster/crecimiento & desarrollo , Cuerpo Adiposo/citología , Cuerpo Adiposo/efectos de los fármacos , Cuerpo Adiposo/metabolismo , Larva/citología , Larva/efectos de los fármacos , Larva/metabolismo , Células-Madre Neurales/efectos de los fármacos , Neuroglía/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal/efectos de los fármacos
16.
PLoS Biol ; 11(9): e1001666, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24086110

RESUMEN

Cardiomyocytes are vulnerable to hypoxia in the adult, but adapted to hypoxia in utero. Current understanding of endogenous cardiac oxygen sensing pathways is limited. Myocardial oxygen consumption is determined by regulation of energy metabolism, which shifts from glycolysis to lipid oxidation soon after birth, and is reversed in failing adult hearts, accompanying re-expression of several "fetal" genes whose role in disease phenotypes remains unknown. Here we show that hypoxia-controlled expression of the transcription factor Hand1 determines oxygen consumption by inhibition of lipid metabolism in the fetal and adult cardiomyocyte, leading to downregulation of mitochondrial energy generation. Hand1 is under direct transcriptional control by HIF1α. Transgenic mice prolonging cardiac Hand1 expression die immediately following birth, failing to activate the neonatal lipid metabolising gene expression programme. Deletion of Hand1 in embryonic cardiomyocytes results in premature expression of these genes. Using metabolic flux analysis, we show that Hand1 expression controls cardiomyocyte oxygen consumption by direct transcriptional repression of lipid metabolising genes. This leads, in turn, to increased production of lactate from glucose, decreased lipid oxidation, reduced inner mitochondrial membrane potential, and mitochondrial ATP generation. We found that this pathway is active in adult cardiomyocytes. Up-regulation of Hand1 is protective in a mouse model of myocardial ischaemia. We propose that Hand1 is part of a novel regulatory pathway linking cardiac oxygen levels with oxygen consumption. Understanding hypoxia adaptation in the fetal heart may allow development of strategies to protect cardiomyocytes vulnerable to ischaemia, for example during cardiac ischaemia or surgery.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Metabolismo Energético/genética , Metabolismo de los Lípidos/genética , Miocardio/metabolismo , Consumo de Oxígeno/genética , Adenosina Trifosfato/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Hipoxia de la Célula/genética , Línea Celular , Regulación del Desarrollo de la Expresión Génica , Corazón/embriología , Corazón/fisiología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Potencial de la Membrana Mitocondrial/genética , Ratones , Ratones Transgénicos , Mitocondrias/metabolismo , Isquemia Miocárdica/genética , Isquemia Miocárdica/metabolismo , Miocitos Cardíacos/metabolismo , Oxígeno/metabolismo , Activación Transcripcional
17.
Annu Rev Entomol ; 59: 405-25, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24397521

RESUMEN

Oenocytes have intrigued insect physiologists since the nineteenth century. Many years of careful but mostly descriptive research on these cells highlights their diverse sizes, numbers, and anatomical distributions across Insecta. Contemporary molecular genetic studies in Drosophila melanogaster and Tribolium castaneum support the hypothesis that oenocytes are of ectodermal origin. They also suggest that, in both short and long germ-band species, oenocytes are induced from a Spalt major/Engrailed ectodermal zone by MAPK signaling. Recent glimpses into some of the physiological functions of oenocytes indicate that they involve fatty acid and hydrocarbon metabolism. Genetic studies in D. melanogaster have shown that larval oenocytes synthesize very-long-chain fatty acids required for tracheal waterproofing and that adult oenocytes produce cuticular hydrocarbons required for desiccation resistance and pheromonal communication. Exciting areas of future research include the evolution of oenocytes and their cross talk with other tissues involved in lipid metabolism such as the fat body.


Asunto(s)
Insectos/citología , Insectos/crecimiento & desarrollo , Animales , Drosophila melanogaster/citología , Drosophila melanogaster/embriología , Drosophila melanogaster/genética , Drosophila melanogaster/crecimiento & desarrollo , Ectodermo/citología , Ectodermo/embriología , Ectodermo/crecimiento & desarrollo , Embrión no Mamífero/citología , Embrión no Mamífero/embriología , Insectos/embriología , Insectos/genética , Larva/citología , Larva/genética , Larva/crecimiento & desarrollo , Tribolium/citología , Tribolium/embriología , Tribolium/genética , Tribolium/crecimiento & desarrollo
18.
Anal Chem ; 85(24): 12046-54, 2013 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-24251761

RESUMEN

The accurate measurement of metabolite concentrations in miniscule biological sample volumes is often desirable, yet it remains challenging. In many cases, the starting analyte volumes are imprecisely known, or not directly measurable, and hence absolute metabolite concentrations are difficult to calculate. Here, we introduce volume determination using two standards (VDTS) as a general quantitative method for the analysis of polar metabolites in submicrolitre samples using (1)H NMR spectroscopy. This approach permits the back calculation of absolute metabolite concentrations from small biological samples of unknown volume. Where small sample volumes are also variable, VDTS can improve multivariate chemometric analysis. In this context, principal component analysis (PCA) yielded more logically consistent and biologically insightful outputs when we used volume-corrected spectra, calculated using VDTS, rather than probabilistic quotient normalization (PQN) of raw spectra. As proof-of-principle, the VDTS-based method and PCA were used to analyze polar metabolites in the hemolymph (blood) extracted from larvae of the very small but widely used genetic model organism Drosophila. This analysis showed that the hemolymph metabolomes of males and females are markedly different when larvae are well fed. However, gender-specific metabolomes tend to converge when larval dietary nutrients are restricted. We discuss the biological implications of these surprising results and compare and contrast them to previous analyses of Drosophila hemolymph and mammalian blood plasma. Together, these findings reveal an interesting and hitherto unknown sexual dimorphism in systemic Drosophila metabolites, clearly warranting further biological investigation. Importantly, the VDTS approach should be adaptable to many different analytical platforms, including mass spectrometry.


Asunto(s)
Límite de Detección , Espectroscopía de Resonancia Magnética/normas , Metabolómica/normas , Estadística como Asunto/normas , Animales , Drosophila melanogaster/metabolismo , Femenino , Hemolinfa/metabolismo , Larva/metabolismo , Masculino , Estándares de Referencia
19.
Nature ; 445(7125): 275-80, 2007 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-17136098

RESUMEN

Lipid metabolism is essential for growth and generates much of the energy needed during periods of starvation. In Drosophila, fasting larvae release large quantities of lipid from the fat body but it is unclear how and where this is processed. Here we identify the oenocyte as the principal cell type accumulating lipid droplets during starvation. Tissue-specific manipulations of the Slimfast amino-acid channel, the Lsd2 fat-storage regulator and the Brummer lipase indicate that oenocytes act downstream of the fat body. In turn, oenocytes are required for depleting stored lipid from the fat body during fasting. Hence, lipid-metabolic coupling between the fat body and oenocytes is bidirectional. When food is plentiful, oenocytes have critical roles in regulating growth, development and feeding behaviour. In addition, they specifically express many different lipid-metabolizing proteins, including Cyp4g1, an omega-hydroxylase regulating triacylglycerol composition. These findings provide evidence that some lipid-processing functions of the mammalian liver are performed in insects by oenocytes.


Asunto(s)
Drosophila melanogaster/citología , Drosophila melanogaster/metabolismo , Hepatocitos/metabolismo , Metabolismo de los Lípidos , Animales , Drosophila melanogaster/enzimología , Drosophila melanogaster/crecimiento & desarrollo , Ayuno , Cuerpo Adiposo/metabolismo , Hepatocitos/enzimología , Larva/crecimiento & desarrollo , Larva/metabolismo , Lipasa/metabolismo , Oxigenasas de Función Mixta/metabolismo , Pupa/crecimiento & desarrollo , Pupa/metabolismo , Inanición
20.
Dev Cell ; 58(10): 836-846.e6, 2023 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-37084728

RESUMEN

Glioblastoma is thought to originate from neural stem cells (NSCs) of the subventricular zone that acquire genetic alterations. In the adult brain, NSCs are largely quiescent, suggesting that deregulation of quiescence maintenance may be a prerequisite for tumor initiation. Although inactivation of the tumor suppressor p53 is a frequent event in gliomagenesis, whether or how it affects quiescent NSCs (qNSCs) remains unclear. Here, we show that p53 maintains quiescence by inducing fatty-acid oxidation (FAO) and that acute p53 deletion in qNSCs results in their premature activation to a proliferative state. Mechanistically, this occurs through direct transcriptional induction of PPARGC1a, which in turn activates PPARα to upregulate FAO genes. Dietary supplementation with fish oil containing omega-3 fatty acids, natural PPARα ligands, fully restores quiescence of p53-deficient NSCs and delays tumor initiation in a glioblastoma mouse model. Thus, diet can silence glioblastoma driver mutations, with important implications for cancer prevention.


Asunto(s)
Glioblastoma , Células-Madre Neurales , Ratones , Animales , Proteína p53 Supresora de Tumor , PPAR alfa , Dieta , Mutación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA