Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
Cell ; 186(19): 4117-4133.e22, 2023 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-37591239

RESUMEN

Aging is the key risk factor for cognitive decline, yet the molecular changes underlying brain aging remain poorly understood. Here, we conducted spatiotemporal RNA sequencing of the mouse brain, profiling 1,076 samples from 15 regions across 7 ages and 2 rejuvenation interventions. Our analysis identified a brain-wide gene signature of aging in glial cells, which exhibited spatially defined changes in magnitude. By integrating spatial and single-nucleus transcriptomics, we found that glial aging was particularly accelerated in white matter compared with cortical regions, whereas specialized neuronal populations showed region-specific expression changes. Rejuvenation interventions, including young plasma injection and dietary restriction, exhibited distinct effects on gene expression in specific brain regions. Furthermore, we discovered differential gene expression patterns associated with three human neurodegenerative diseases, highlighting the importance of regional aging as a potential modulator of disease. Our findings identify molecular foci of brain aging, providing a foundation to target age-related cognitive decline.


Asunto(s)
Envejecimiento , Disfunción Cognitiva , Sustancia Blanca , Animales , Humanos , Ratones , Disfunción Cognitiva/genética , Perfilación de la Expresión Génica , Núcleo Solitario , Sustancia Blanca/patología , Análisis de Expresión Génica de una Sola Célula , Encéfalo/patología
2.
Nature ; 616(7958): 814-821, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-37046086

RESUMEN

Physiological homeostasis becomes compromised during ageing, as a result of impairment of cellular processes, including transcription and RNA splicing1-4. However, the molecular mechanisms leading to the loss of transcriptional fidelity are so far elusive, as are ways of preventing it. Here we profiled and analysed genome-wide, ageing-related changes in transcriptional processes across different organisms: nematodes, fruitflies, mice, rats and humans. The average transcriptional elongation speed (RNA polymerase II speed) increased with age in all five species. Along with these changes in elongation speed, we observed changes in splicing, including a reduction of unspliced transcripts and the formation of more circular RNAs. Two lifespan-extending interventions, dietary restriction and lowered insulin-IGF signalling, both reversed most of these ageing-related changes. Genetic variants in RNA polymerase II that reduced its speed in worms5 and flies6 increased their lifespan. Similarly, reducing the speed of RNA polymerase II by overexpressing histone components, to counter age-associated changes in nucleosome positioning, also extended lifespan in flies and the division potential of human cells. Our findings uncover fundamental molecular mechanisms underlying animal ageing and lifespan-extending interventions, and point to possible preventive measures.


Asunto(s)
Envejecimiento , Longevidad , Elongación de la Transcripción Genética , Animales , Humanos , Ratones , Ratas , Envejecimiento/genética , Insulina/metabolismo , Longevidad/genética , ARN Polimerasa II/genética , ARN Polimerasa II/metabolismo , Transducción de Señal , Drosophila melanogaster/genética , Caenorhabditis elegans/genética , ARN Circular , Somatomedinas , Nucleosomas , Histonas , División Celular , Restricción Calórica
3.
Mol Cell ; 79(2): 268-279.e5, 2020 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-32592682

RESUMEN

Circular RNAs (circRNAs) are abundant and accumulate with age in neurons of diverse species. However, only few circRNAs have been functionally characterized, and their role during aging has not been addressed. Here, we use transcriptome profiling during aging and find that accumulation of circRNAs is slowed down in long-lived insulin mutant flies. Next, we characterize the in vivo function of a circRNA generated by the sulfateless gene (circSfl), which is consistently upregulated, particularly in the brain and muscle, of diverse long-lived insulin mutants. Strikingly, lifespan extension of insulin mutants is dependent on circSfl, and overexpression of circSfl alone is sufficient to extend the lifespan. Moreover, circSfl is translated into a protein that shares the N terminus and potentially some functions with the full-length Sfl protein encoded by the host gene. Our study demonstrates that insulin signaling affects global circRNA accumulation and reveals an important role of circSfl during aging in vivo.


Asunto(s)
Drosophila/fisiología , Insulina/fisiología , Longevidad/genética , ARN Circular/fisiología , Envejecimiento , Animales , Animales Modificados Genéticamente , Drosophila/genética , Proteínas de Drosophila/genética , Femenino , Masculino , Mutación , Neuronas/fisiología , Sulfotransferasas/genética , Transcriptoma
4.
Proc Natl Acad Sci U S A ; 116(42): 20817-20819, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31570569

RESUMEN

Increasing life expectancy is causing the prevalence of age-related diseases to rise, and there is an urgent need for new strategies to improve health at older ages. Reduced activity of insulin/insulin-like growth factor signaling (IIS) and mechanistic target of rapamycin (mTOR) nutrient-sensing signaling network can extend lifespan and improve health during aging in diverse organisms. However, the extensive feedback in this network and adverse side effects of inhibition imply that simultaneous targeting of specific effectors in the network may most effectively combat the effects of aging. We show that the mitogen-activated protein kinase kinase (MEK) inhibitor trametinib, the mTOR complex 1 (mTORC1) inhibitor rapamycin, and the glycogen synthase kinase-3 (GSK-3) inhibitor lithium act additively to increase longevity in Drosophila Remarkably, the triple drug combination increased lifespan by 48%. Furthermore, the combination of lithium with rapamycin cancelled the latter's effects on lipid metabolism. In conclusion, a polypharmacology approach of combining established, prolongevity drug inhibitors of specific nodes may be the most effective way to target the nutrient-sensing network to improve late-life health.


Asunto(s)
Envejecimiento/efectos de los fármacos , Drosophila/efectos de los fármacos , Litio/farmacología , Longevidad/efectos de los fármacos , Nutrientes/metabolismo , Piridonas/farmacología , Pirimidinonas/farmacología , Sirolimus/farmacología , Anciano , Envejecimiento/metabolismo , Animales , Drosophila/genética , Drosophila/crecimiento & desarrollo , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Combinación de Medicamentos , Femenino , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3/metabolismo , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina/antagonistas & inhibidores , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Persona de Mediana Edad , Transducción de Señal/efectos de los fármacos
5.
PLoS Genet ; 14(11): e1007766, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30462643

RESUMEN

Dietary, pharmacological and genetic interventions can extend health- and lifespan in diverse mammalian species. DNA methylation has been implicated in mediating the beneficial effects of these interventions; methylation patterns deteriorate during ageing, and this is prevented by lifespan-extending interventions. However, whether these interventions also actively shape the epigenome, and whether such epigenetic reprogramming contributes to improved health at old age, remains underexplored. We analysed published, whole-genome, BS-seq data sets from mouse liver to explore DNA methylation patterns in aged mice in response to three lifespan-extending interventions: dietary restriction (DR), reduced TOR signaling (rapamycin), and reduced growth (Ames dwarf mice). Dwarf mice show enhanced DNA hypermethylation in the body of key genes in lipid biosynthesis, cell proliferation and somatotropic signaling, which strongly correlates with the pattern of transcriptional repression. Remarkably, DR causes a similar hypermethylation in lipid biosynthesis genes, while rapamycin treatment increases methylation signatures in genes coding for growth factor and growth hormone receptors. Shared changes of DNA methylation were restricted to hypermethylated regions, and they were not merely a consequence of slowed ageing, thus suggesting an active mechanism driving their formation. By comparing the overlap in ageing-independent hypermethylated patterns between all three interventions, we identified four regions, which, independent of genetic background or gender, may serve as novel biomarkers for longevity-extending interventions. In summary, we identified gene body hypermethylation as a novel and partly conserved signature of lifespan-extending interventions in mouse, highlighting epigenetic reprogramming as a possible intervention to improve health at old age.


Asunto(s)
Metilación de ADN , Epigénesis Genética , Hígado/metabolismo , Longevidad/genética , Envejecimiento/genética , Envejecimiento/metabolismo , Animales , Restricción Calórica , Metilación de ADN/efectos de los fármacos , Bases de Datos Genéticas , Femenino , Sustancias de Crecimiento/metabolismo , Metabolismo de los Lípidos/genética , Longevidad/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Mutantes , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/genética , Transducción de Señal , Sirolimus/farmacología
6.
Proc Natl Acad Sci U S A ; 115(41): E9620-E9629, 2018 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-30249665

RESUMEN

Mammals develop age-associated clonal expansion of somatic mtDNA mutations resulting in severe respiratory chain deficiency in a subset of cells in a variety of tissues. Both mathematical modeling based on descriptive data from humans and experimental data from mtDNA mutator mice suggest that the somatic mutations are formed early in life and then undergo mitotic segregation during adult life to reach very high levels in certain cells. To address whether mtDNA mutations have a universal effect on aging metazoans, we investigated their role in physiology and aging of fruit flies. To this end, we utilized genetically engineered flies expressing mutant versions of the catalytic subunit of mitochondrial DNA polymerase (DmPOLγA) as a means to introduce mtDNA mutations. We report here that lifespan and health in fruit flies are remarkably tolerant to mtDNA mutations. Our results show that the short lifespan and wide genetic bottleneck of fruit flies are limiting the extent of clonal expansion of mtDNA mutations both in individuals and between generations. However, an increase of mtDNA mutations to very high levels caused sensitivity to mechanical and starvation stress, intestinal stem cell dysfunction, and reduced lifespan under standard conditions. In addition, the effects of dietary restriction, widely considered beneficial for organismal health, were attenuated in flies with very high levels of mtDNA mutations.


Asunto(s)
ADN Mitocondrial , Longevidad/genética , Mutación , Animales , ADN Mitocondrial/genética , ADN Mitocondrial/metabolismo , Drosophila melanogaster
7.
Mol Syst Biol ; 13(9): 939, 2017 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-28916541

RESUMEN

Lowered activity of the insulin/IGF signalling (IIS) network can ameliorate the effects of ageing in laboratory animals and, possibly, humans. Although transcriptome remodelling in long-lived IIS mutants has been extensively documented, the causal mechanisms contributing to extended lifespan, particularly in specific tissues, remain unclear. We have characterized the proteomes of four key insulin-sensitive tissues in a long-lived Drosophila IIS mutant and control, and detected 44% of the predicted proteome (6,085 proteins). Expression of ribosome-associated proteins in the fat body was reduced in the mutant, with a corresponding, tissue-specific reduction in translation. Expression of mitochondrial electron transport chain proteins in fat body was increased, leading to increased respiration, which was necessary for IIS-mediated lifespan extension, and alone sufficient to mediate it. Proteasomal subunits showed altered expression in IIS mutant gut, and gut-specific over-expression of the RPN6 proteasomal subunit, was sufficient to increase proteasomal activity and extend lifespan, whilst inhibition of proteasome activity abolished IIS-mediated longevity. Our study thus uncovered strikingly tissue-specific responses of cellular processes to lowered IIS acting in concert to ameliorate ageing.


Asunto(s)
Envejecimiento/metabolismo , Drosophila/metabolismo , Redes Reguladoras de Genes , Proteómica/métodos , Animales , Proteínas de Drosophila , Cuerpo Adiposo/metabolismo , Insulina/metabolismo , Mucosa Intestinal/metabolismo , Modelos Animales , Mutación , Especificidad de Órganos , Proteínas Ribosómicas/metabolismo
8.
Acta Neuropathol ; 135(3): 445-457, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29380049

RESUMEN

A GGGGCC hexanucleotide repeat expansion in the C9orf72 gene is the most common genetic cause of amyotrophic lateral sclerosis and frontotemporal dementia. Neurodegeneration may occur via transcription of the repeats into inherently toxic repetitive sense and antisense RNA species, or via repeat-associated non-ATG initiated translation (RANT) of sense and antisense RNA into toxic dipeptide repeat proteins. We have previously demonstrated that regular interspersion of repeat RNA with stop codons prevents RANT (RNA-only models), allowing us to study the role of repeat RNA in isolation. Here we have created novel RNA-only Drosophila models, including the first models of antisense repeat toxicity, and flies expressing extremely large repeats, within the range observed in patients. We generated flies expressing ~ 100 repeat sense or antisense RNA either as part of a processed polyadenylated transcript or intronic sequence. We additionally created Drosophila expressing > 1000 RNA-only repeats in the sense direction. When expressed in adult Drosophila neurons polyadenylated repeat RNA is largely cytoplasmic in localisation, whilst intronic repeat RNA forms intranuclear RNA foci, as does > 1000 repeat RNA, thus allowing us to investigate both nuclear and cytoplasmic RNA toxicity. We confirmed that these RNA foci are capable of sequestering endogenous Drosophila RNA-binding proteins, and that the production of dipeptide proteins (poly-glycine-proline, and poly-glycine-arginine) is suppressed in our models. We find that neither cytoplasmic nor nuclear sense or antisense RNA are toxic when expressed in adult Drosophila neurons, suggesting they have a limited role in disease pathogenesis.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Proteína C9orf72/metabolismo , Demencia Frontotemporal/metabolismo , ARN/metabolismo , Animales , Animales Modificados Genéticamente , Proteína C9orf72/genética , Núcleo Celular/metabolismo , Núcleo Celular/patología , Citoplasma/metabolismo , Citoplasma/patología , Expansión de las Repeticiones de ADN , Modelos Animales de Enfermedad , Drosophila , Femenino , Demencia Frontotemporal/patología , Intrones , Masculino , Neuronas/metabolismo , Neuronas/patología
9.
J Neurosci ; 36(46): 11654-11670, 2016 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-27852774

RESUMEN

Glucocerebrosidase (GBA1) mutations are associated with Gaucher disease (GD), an autosomal recessive disorder caused by functional deficiency of glucocerebrosidase (GBA), a lysosomal enzyme that hydrolyzes glucosylceramide to ceramide and glucose. Neuronopathic forms of GD can be associated with rapid neurological decline (Type II) or manifest as a chronic form (Type III) with a wide spectrum of neurological signs. Furthermore, there is now a well-established link between GBA1 mutations and Parkinson's disease (PD), with heterozygote mutations in GBA1 considered the commonest genetic defect in PD. Here we describe a novel Drosophila model of GD that lacks the two fly GBA1 orthologs. This knock-out model recapitulates the main features of GD at the cellular level with severe lysosomal defects and accumulation of glucosylceramide in the fly brain. We also demonstrate a block in autophagy flux in association with reduced lifespan, age-dependent locomotor deficits and accumulation of autophagy substrates in dGBA-deficient fly brains. Furthermore, mechanistic target of rapamycin (mTOR) signaling is downregulated in dGBA knock-out flies, with a concomitant upregulation of Mitf gene expression, the fly ortholog of mammalian TFEB, likely as a compensatory response to the autophagy block. Moreover, the mTOR inhibitor rapamycin is able to partially ameliorate the lifespan, locomotor, and oxidative stress phenotypes. Together, our results demonstrate that this dGBA1-deficient fly model is a useful platform for the further study of the role of lysosomal-autophagic impairment and the potential therapeutic benefits of rapamycin in neuronopathic GD. These results also have important implications for the role of autophagy and mTOR signaling in GBA1-associated PD SIGNIFICANCE STATEMENT: We developed a Drosophila model of neuronopathic GD by knocking-out the fly orthologs of the GBA1 gene, demonstrating abnormal lysosomal pathology in the fly brain. Functioning lysosomes are required for autophagosome-lysosomal fusion in the autophagy pathway. We show in vivo that autophagy is impaired in dGBA-deficient fly brains. In response, mechanistic target of rapamycin (mTOR) activity is downregulated in dGBA-deficient flies and rapamycin ameliorates the lifespan, locomotor, and oxidative stress phenotypes. dGBA knock-out flies also display an upregulation of the Drosophila ortholog of mammalian TFEB, Mitf, a response that is unable to overcome the autophagy block. Together, our results suggest that rapamycin may have potential benefits in the treatment of GD, as well as PD linked to GBA1 mutations.


Asunto(s)
Modelos Animales de Enfermedad , Enfermedad de Gaucher/metabolismo , Enfermedad de Gaucher/prevención & control , Glucosilceramidasa/genética , Lisosomas/metabolismo , Sirolimus/administración & dosificación , Serina-Treonina Quinasas TOR/metabolismo , Animales , Animales Modificados Genéticamente , Autofagia/efectos de los fármacos , Drosophila , Enfermedad de Gaucher/patología , Técnicas de Inactivación de Genes , Transducción de Señal/efectos de los fármacos
10.
PLoS Biol ; 12(4): e1001824, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24690889

RESUMEN

Sleep fragmentation, particularly reduced and interrupted night sleep, impairs the quality of life of older people. Strikingly similar declines in sleep quality are seen during ageing in laboratory animals, including the fruit fly Drosophila. We investigated whether reduced activity of the nutrient- and stress-sensing insulin/insulin-like growth factor (IIS)/TOR signalling network, which ameliorates ageing in diverse organisms, could rescue the sleep fragmentation of ageing Drosophila. Lowered IIS/TOR network activity improved sleep quality, with increased night sleep and day activity and reduced sleep fragmentation. Reduced TOR activity, even when started for the first time late in life, improved sleep quality. The effects of reduced IIS/TOR network activity on day and night phenotypes were mediated through distinct mechanisms: Day activity was induced by adipokinetic hormone, dFOXO, and enhanced octopaminergic signalling. In contrast, night sleep duration and consolidation were dependent on reduced S6K and dopaminergic signalling. Our findings highlight the importance of different IIS/TOR components as potential therapeutic targets for pharmacological treatment of age-related sleep fragmentation in humans.


Asunto(s)
Drosophila/metabolismo , Privación de Sueño/metabolismo , Sueño/fisiología , Somatomedinas/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Envejecimiento , Animales , Ritmo Circadiano/genética , Ritmo Circadiano/fisiología , Dopamina/biosíntesis , Dopamina/metabolismo , Drosophila/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Femenino , Factores de Transcripción Forkhead/metabolismo , Proteínas Inhibidoras de la Apoptosis/genética , Hormonas de Insectos/metabolismo , Insulina/metabolismo , Octopamina/metabolismo , Oligopéptidos/metabolismo , Ácido Pirrolidona Carboxílico/análogos & derivados , Ácido Pirrolidona Carboxílico/metabolismo , Receptor de Insulina/genética , Receptores Dopaminérgicos/biosíntesis , Receptores de Glucagón/genética , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Transducción de Señal , Sirolimus/farmacología , Somatomedinas/biosíntesis , Somatomedinas/genética , Serina-Treonina Quinasas TOR/antagonistas & inhibidores
11.
PLoS Genet ; 9(1): e1003178, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23300484

RESUMEN

Regulation of mitochondrial DNA (mtDNA) expression is critical for the control of oxidative phosphorylation in response to physiological demand, and this regulation is often impaired in disease and aging. We have previously shown that mitochondrial transcription termination factor 3 (MTERF3) is a key regulator that represses mtDNA transcription in the mouse, but its molecular mode of action has remained elusive. Based on the hypothesis that key regulatory mechanisms for mtDNA expression are conserved in metazoans, we analyzed Mterf3 knockout and knockdown flies. We demonstrate here that decreased expression of MTERF3 not only leads to activation of mtDNA transcription, but also impairs assembly of the large mitochondrial ribosomal subunit. This novel function of MTERF3 in mitochondrial ribosomal biogenesis is conserved in the mouse, thus we identify a novel and unexpected role for MTERF3 in coordinating the crosstalk between transcription and translation for the regulation of mammalian mtDNA gene expression.


Asunto(s)
Proteínas de Drosophila , Drosophila melanogaster/genética , Mitocondrias , Proteínas Mitocondriales , Ribosomas , Animales , ADN Mitocondrial/genética , Proteínas de Drosophila/genética , Regulación de la Expresión Génica , Invertebrados/genética , Invertebrados/metabolismo , Ratones , Mitocondrias/genética , Mitocondrias/metabolismo , Proteínas Mitocondriales/genética , Fosforilación Oxidativa , Ribosomas/genética , Ribosomas/metabolismo , Transcripción Genética
12.
Acta Neuropathol ; 130(1): 35-47, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25862636

RESUMEN

The involvement of Amyloid-ß (Aß) in the pathogenesis of Alzheimer's disease (AD) is well established. However, it is becoming clear that the amyloid load in AD brains consists of a heterogeneous mixture of Aß peptides, implying that a thorough understanding of their respective role and toxicity is crucial for the development of efficient treatments. Besides the well-studied Aß40 and Aß42 species, recent data have raised the possibility that Aß43 peptides might be instrumental in AD pathogenesis, because they are frequently observed in both dense and diffuse amyloid plaques from human AD brains and are highly amyloidogenic in vitro. However, whether Aß43 is toxic in vivo is currently unclear. Using Drosophila transgenic models of amyloid pathology, we show that Aß43 peptides are mainly insoluble and highly toxic in vivo, leading to the progressive loss of photoreceptor neurons, altered locomotion and decreased lifespan when expressed in the adult fly nervous system. In addition, we demonstrate that Aß43 species are able to trigger the aggregation of the typically soluble and non-toxic Aß40, leading to synergistic toxic effects on fly lifespan and climbing ability, further suggesting that Aß43 peptides could act as a nucleating factor in AD brains. Altogether, our study demonstrates high pathogenicity of Aß43 species in vivo and supports the idea that Aß43 contributes to the pathological events leading to neurodegeneration in AD.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Péptidos beta-Amiloides/toxicidad , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/toxicidad , Péptidos beta-Amiloides/genética , Animales , Animales Modificados Genéticamente , Modelos Animales de Enfermedad , Drosophila , Femenino , Cabeza/patología , Cabeza/fisiología , Actividad Motora/fisiología , Fragmentos de Péptidos/genética , Células Fotorreceptoras de Invertebrados/patología , Células Fotorreceptoras de Invertebrados/fisiología , Agregado de Proteínas , ARN Mensajero/metabolismo , Solubilidad , Análisis de Supervivencia
13.
J Cell Sci ; 125(Pt 15): 3568-77, 2012 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-22505614

RESUMEN

Lipid droplets are the main lipid storage sites in cells. Lipid droplet homeostasis is regulated by the surface accessibility of lipases. Mammalian adipose triglyceride lipase (ATGL) and hormone-sensitive lipase (HSL) are two key lipases for basal and stimulated lipolysis, respectively. Perilipins, the best known lipid droplet surface proteins, can either recruit lipases or prevent the access of lipases to lipid droplets. Mammals have five perilipin proteins, which often exhibit redundant functions, precluding the analysis of the exact role of individual perilipins in vivo. Drosophila have only two perilipins, PLIN1/LSD-1 and PLIN2/LSD-2. Previous studies revealed that PLIN2 is important for protecting lipid droplets from lipolysis mediated by Brummer (BMM), the Drosophila homolog of ATGL. In this study, we report the functional analysis of PLIN1 and Drosophila HSL. Loss-of-function and overexpression studies reveal that unlike PLIN2, PLIN1 probably facilitates lipid mobilization. HSL is recruited from the cytosol to the surface of lipid droplets under starved conditions and PLIN1 is necessary for the starved induced lipid droplet localization of HSL. Moreover, phenotypic analysis of plin1;plin2 double mutants revealed that PLIN1 and PLIN2 might have redundant functions in protecting lipid droplets from lipolysis. Therefore, the two Drosophila perilipins have both opposite and redundant roles. Domain swapping and deletion analyses indicate that the C-terminal region of PLIN1 confers functional specificity to PLIN1. Our study highlights the complex roles of Drosophila perilipin proteins and the evolutionarily conserved regulation of HSL translocation by perilipins.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Metabolismo de los Lípidos/fisiología , Oxidorreductasas N-Desmetilantes/metabolismo , Animales , Drosophila/genética , Proteínas de Drosophila/genética , Oxidorreductasas N-Desmetilantes/genética , Transporte de Proteínas , Esterol Esterasa/genética , Esterol Esterasa/metabolismo
14.
PLoS Genet ; 7(10): e1002324, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22022283

RESUMEN

The bicoid stability factor (BSF) of Drosophila melanogaster has been reported to be present in the cytoplasm, where it stabilizes the maternally contributed bicoid mRNA and binds mRNAs expressed from early zygotic genes. BSF may also have other roles, as it is ubiquitously expressed and essential for survival of adult flies. We have performed immunofluorescence and cell fractionation analyses and show here that BSF is mainly a mitochondrial protein. We studied two independent RNAi knockdown fly lines and report that reduced BSF protein levels lead to a severe respiratory deficiency and delayed development at the late larvae stage. Ubiquitous knockdown of BSF results in a severe reduction of the polyadenylation tail lengths of specific mitochondrial mRNAs, accompanied by an enrichment of unprocessed polycistronic RNA intermediates. Furthermore, we observed a significant reduction in mRNA steady state levels, despite increased de novo transcription. Surprisingly, mitochondrial de novo translation is increased and abnormal mitochondrial translation products are present in knockdown flies, suggesting that BSF also has a role in coordinating the mitochondrial translation in addition to its role in mRNA maturation and stability. We thus report a novel function of BSF in flies and demonstrate that it has an important intra-mitochondrial role, which is essential for maintaining mtDNA gene expression and oxidative phosphorylation.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila melanogaster/crecimiento & desarrollo , Drosophila melanogaster/genética , Mitocondrias/genética , Poliadenilación/genética , ARN Mensajero/genética , Proteínas de Unión al ARN/genética , Animales , Peso Corporal/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/fisiología , Fertilidad/genética , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Mitocondrias/fisiología , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Fosforilación Oxidativa , Filogenia , Biosíntesis de Proteínas , Interferencia de ARN , Proteínas de Unión al ARN/metabolismo
15.
Nat Aging ; 4(4): 491-509, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38413780

RESUMEN

Suppression of target of rapamycin complex 1 (TORC1) by rapamycin ameliorates aging in diverse species. S6 kinase (S6K) is an essential mediator, but the mechanisms involved are unclear. Here we show that activation of S6K specifically in Drosophila fat-body blocked extension of lifespan by rapamycin, induced accumulation of multilamellar lysosomes and blocked age-associated hyperactivation of the NF-κB-like immune deficiency (IMD) pathway, indicative of reduced inflammaging. Syntaxin 13 mediated the effects of TORC1-S6K signaling on lysosome morphology and inflammaging, suggesting they may be linked. Inflammaging depended on the IMD receptor regulatory isoform PGRP-LC, and repression of the IMD pathway from midlife extended lifespan. Age-related inflammaging was higher in females than in males and was not lowered in males by rapamycin treatment or lowered S6K. Rapamycin treatment also elevated Syntaxin 12/13 levels in mouse liver and prevented age-related increase in noncanonical NF-κB signaling, suggesting that the effect of TORC1 on inflammaging is conserved from flies to mammals.


Asunto(s)
Longevidad , FN-kappa B , Animales , Femenino , Masculino , Ratones , Drosophila , Inflamación/tratamiento farmacológico , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , FN-kappa B/genética , Proteínas Quinasas S6 Ribosómicas/metabolismo , Sirolimus/farmacología
16.
Mol Metab ; 81: 101902, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38360109

RESUMEN

OBJECTIVE: Rapamycin, a powerful geroprotective drug, can have detrimental effects when administered chronically. We determined whether intermittent treatment of mice can reduce negative effects while maintaining benefits of chronic treatment. METHODS: From 6 months of age, male and female C3B6F1 hybrid mice were either continuously fed with 42 mg/kg rapamycin, or intermittently fed by alternating weekly feeding of 42 mg/kg rapamycin food with weekly control feeding. Survival of these mice compared to control animals was measured. Furthermore, longitudinal phenotyping including metabolic (body composition, GTT, ITT, indirect calorimetry) and fitness phenotypes (treadmil, rotarod, electrocardiography and open field) was performed. Organ specific pathology was assessed at 24 months of age. RESULTS: Chronic rapamycin treatment induced glucose intolerance, which was partially ameliorated by intermittent treatment. Chronic and intermittent rapamycin treatments increased lifespan equally in males, while in females chronic treatment resulted in slightly higher survival. The two treatments had equivalent effects on testicular degeneration, heart fibrosis and liver lipidosis. In males, the two treatment regimes led to a similar increase in motor coordination, heart rate and Q-T interval, and reduction in spleen weight, while in females, they equally reduced BAT inflammation and spleen weight and maintained heart rate and Q-T interval. However, other health parameters, including age related pathologies, were better prevented by continuous treatment. CONCLUSIONS: Intermittent rapamycin treatment is effective in prolonging lifespan and reduces some side-effects of chronic treatment, but chronic treatment is more beneficial to healthspan.


Asunto(s)
Hígado Graso , Intolerancia a la Glucosa , Masculino , Femenino , Ratones , Animales , Longevidad/fisiología , Sirolimus/farmacología , Inflamación
17.
PLoS Genet ; 6(2): e1000857, 2010 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-20195512

RESUMEN

Multicellular animals match costly activities, such as growth and reproduction, to the environment through nutrient-sensing pathways. The insulin/IGF signaling (IIS) pathway plays key roles in growth, metabolism, stress resistance, reproduction, and longevity in diverse organisms including mammals. Invertebrate genomes often contain multiple genes encoding insulin-like ligands, including seven Drosophila insulin-like peptides (DILPs). We investigated the evolution, diversification, redundancy, and functions of the DILPs, combining evolutionary analysis, based on the completed genome sequences of 12 Drosophila species, and functional analysis, based on newly-generated knock-out mutations for all 7 dilp genes in D. melanogaster. Diversification of the 7 DILPs preceded diversification of Drosophila species, with stable gene diversification and family membership, suggesting stabilising selection for gene function. Gene knock-outs demonstrated both synergy and compensation of expression between different DILPs, notably with DILP3 required for normal expression of DILPs 2 and 5 in brain neurosecretory cells and expression of DILP6 in the fat body compensating for loss of brain DILPs. Loss of DILP2 increased lifespan and loss of DILP6 reduced growth, while loss of DILP7 did not affect fertility, contrary to its proposed role as a Drosophila relaxin. Importantly, loss of DILPs produced in the brain greatly extended lifespan but only in the presence of the endosymbiontic bacterium Wolbachia, demonstrating a specific interaction between IIS and Wolbachia in lifespan regulation. Furthermore, loss of brain DILPs blocked the responses of lifespan and fecundity to dietary restriction (DR) and the DR response of these mutants suggests that IIS extends lifespan through mechanisms that both overlap with those of DR and through additional mechanisms that are independent of those at work in DR. Evolutionary conservation has thus been accompanied by synergy, redundancy, and functional differentiation between DILPs, and these features may themselves be of evolutionary advantage.


Asunto(s)
Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila/genética , Evolución Molecular , Animales , Peso Corporal/efectos de los fármacos , Dieta , Drosophila/efectos de los fármacos , Drosophila/crecimiento & desarrollo , Drosophila/microbiología , Resistencia a Medicamentos/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Metabolismo Energético/genética , Fertilidad/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Sitios Genéticos/genética , Longevidad/efectos de los fármacos , Mutación/genética , Óvulo/citología , Óvulo/efectos de los fármacos , Filogenia , Estrés Fisiológico/efectos de los fármacos , Estrés Fisiológico/genética , Análisis de Supervivencia , Factores de Tiempo , Wolbachia/metabolismo , Xenobióticos/farmacología
18.
Sci Adv ; 9(8): eade8137, 2023 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-36812323

RESUMEN

Reduced activity of insulin/insulin-like growth factor signaling (IIS) extends health and life span in mammals. Loss of the insulin receptor substrate 1 (Irs1) gene increases survival in mice and causes tissue-specific changes in gene expression. However, the tissues underlying IIS-mediated longevity are currently unknown. Here, we measured survival and health span in mice lacking IRS1 specifically in liver, muscle, fat, and brain. Tissue-specific loss of IRS1 did not increase survival, suggesting that lack of IRS1 in more than one tissue is required for life-span extension. Loss of IRS1 in liver, muscle, and fat did not improve health. In contrast, loss of neuronal IRS1 increased energy expenditure, locomotion, and insulin sensitivity, specifically in old males. Neuronal loss of IRS1 also caused male-specific mitochondrial dysfunction, activation of Atf4, and metabolic adaptations consistent with an activated integrated stress response at old age. Thus, we identified a male-specific brain signature of aging in response to reduced IIS associated with improved health at old age.


Asunto(s)
Resistencia a la Insulina , Insulina , Femenino , Masculino , Ratones , Animales , Insulina/metabolismo , Transducción de Señal/genética , Longevidad/genética , Neuronas/metabolismo , Mamíferos/metabolismo
19.
Cell Rep ; 42(7): 112722, 2023 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-37384530

RESUMEN

Aging impairs the capacity to respond to novel antigens, reducing immune protection against pathogens and vaccine efficacy. Dietary restriction (DR) extends life- and health span in diverse animals. However, little is known about the capacity of DR to combat the decline in immune function. Here, we study the changes in B cell receptor (BCR) repertoire during aging in DR and control mice. By sequencing the variable region of the BCR heavy chain in the spleen, we show that DR preserves diversity and attenuates the increase in clonal expansions throughout aging. Remarkably, mice starting DR in mid-life have repertoire diversity and clonal expansion rates indistinguishable from chronic DR mice. In contrast, in the intestine, these traits are unaffected by either age or DR. Reduced within-individual B cell repertoire diversity and increased clonal expansions are correlated with higher morbidity, suggesting a potential contribution of B cell repertoire dynamics to health during aging.


Asunto(s)
Envejecimiento , Linfocitos B , Ratones , Animales , Receptores de Antígenos de Linfocitos B
20.
Life Sci Alliance ; 6(9)2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37308278

RESUMEN

Hexanucleotide repeat expansions in the C9orf72 gene are the most prevalent genetic cause of amyotrophic lateral sclerosis and frontotemporal dementia. Transcripts of the expansions are translated into toxic dipeptide repeat (DPR) proteins. Most preclinical studies in cell and animal models have used protein-tagged polyDPR constructs to investigate DPR toxicity but the effects of tags on DPR toxicity have not been systematically explored. Here, we used Drosophila to assess the influence of protein tags on DPR toxicity. Tagging of 36 but not 100 arginine-rich DPRs with mCherry increased toxicity, whereas adding mCherry or GFP to GA100 completely abolished toxicity. FLAG tagging also reduced GA100 toxicity but less than the longer fluorescent tags. Expression of untagged but not GFP- or mCherry-tagged GA100 caused DNA damage and increased p62 levels. Fluorescent tags also affected GA100 stability and degradation. In summary, protein tags affect DPR toxicity in a tag- and DPR-dependent manner, and GA toxicity might be underestimated in studies using tagged GA proteins. Thus, including untagged DPRs as controls is important when assessing DPR toxicity in preclinical models.


Asunto(s)
Esclerosis Amiotrófica Lateral , Neoplasias Cutáneas , Animales , Dipéptidos , Proteína C9orf72 , Péptidos , Genes Reguladores , Drosophila
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA