Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Aging Cell ; : e14141, 2024 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-38465473

RESUMO

The genetic pathways that modulate ageing in multicellular organisms are typically highly conserved across wide evolutionary distances. Recently RNA polymerase III (Pol III) was shown to promote ageing in yeast, C. elegans and D. melanogaster. In this study we investigated the role of Pol III in mammalian ageing using C57BL/6N mice heterozygous for Pol III (Polr3b+/- ). We identified sexually dimorphic, organ-specific beneficial as well as detrimental effects of the Polr3b+/- mutation on health. Female Polr3b+/- mice displayed improved bone health during ageing, but their ability to maintain an effective gut barrier function was compromised and they were susceptible to idiopathic dermatitis (ID). In contrast, male Polr3b+/- mice were lighter than wild-type (WT) males and had a significantly improved gut barrier function in old age. Several metabolic parameters were affected by both age and sex, but no genotype differences were detected. Neither male nor female Polr3b+/- mice were long-lived compared to WT controls. Overall, we find no evidence that a reduced Pol III activity extends mouse lifespan but we do find some potential organ- and sex-specific benefits for old-age health.

2.
Proc Natl Acad Sci U S A ; 121(4): e2311313121, 2024 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-38241436

RESUMO

Pharmacological therapies are promising interventions to slow down aging and reduce multimorbidity in the elderly. Studies in animal models are the first step toward translation of candidate molecules into human therapies, as they aim to elucidate the molecular pathways, cellular mechanisms, and tissue pathologies involved in the anti-aging effects. Trametinib, an allosteric inhibitor of MEK within the Ras/MAPK (Ras/Mitogen-Activated Protein Kinase) pathway and currently used as an anti-cancer treatment, emerged as a geroprotector candidate because it extended lifespan in the fruit fly Drosophila melanogaster. Here, we confirm that trametinib consistently and robustly extends female lifespan, and reduces intestinal stem cell (ISC) proliferation, tumor formation, tissue dysplasia, and barrier disruption in guts in aged flies. In contrast, pro-longevity effects of trametinib are weak and inconsistent in males, and it does not influence gut homeostasis. Inhibition of the Ras/MAPK pathway specifically in ISCs is sufficient to partially recapitulate the effects of trametinib. Moreover, in ISCs, trametinib decreases the activity of the RNA polymerase III (Pol III), a conserved enzyme synthesizing transfer RNAs and other short, non-coding RNAs, and whose inhibition also extends lifespan and reduces gut pathology. Finally, we show that the pro-longevity effect of trametinib in ISCs is partially mediated by Maf1, a repressor of Pol III, suggesting a life-limiting Ras/MAPK-Maf1-Pol III axis in these cells. The mechanism of action described in this work paves the way for further studies on the anti-aging effects of trametinib in mammals and shows its potential for clinical application in humans.


Assuntos
Drosophila melanogaster , Drosophila , Piridonas , Pirimidinonas , Animais , Masculino , Humanos , Feminino , Idoso , Drosophila melanogaster/genética , Envelhecimento/fisiologia , Células-Tronco/metabolismo , Mamíferos
3.
Genome Res ; 32(2): 258-265, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35078808

RESUMO

Reduced provision of protein translation machinery promotes healthy aging in a number of animal models. In humans, however, inborn impairments in translation machinery are a known cause of several developmental disorders, collectively termed ribosomopathies. Here, we use casual inference approaches in genetic epidemiology to investigate whether adult, tissue-specific biogenesis of translation machinery drives human aging. We assess naturally occurring variation in the expression of genes encoding subunits specific to the two RNA polymerases (Pols) that transcribe ribosomal and transfer RNAs, namely Pol I and III, and the variation in expression of ribosomal protein (RP) genes, using Mendelian randomization. We find each causally associated with human longevity (ß = -0.15 ± 0.047, P = 9.6 × 10-4, q = 0.015; ß = -0.13 ± 0.040, P = 1.4 × 10-3, q = 0.023; ß = -0.048 ± 0.016, P = 3.5 × 10-3, q = 0.056, respectively), and this does not appear to be mediated by altered susceptibility to a single disease. We find that reduced expression of Pol III, RPs, or Pol I promotes longevity from different organs, namely visceral adipose, liver, and skeletal muscle, echoing the tissue specificity of ribosomopathies. Our study shows the utility of leveraging genetic variation in expression to elucidate how essential cellular processes impact human aging. The findings extend the evolutionary conservation of protein synthesis as a critical process that drives animal aging to include humans.


Assuntos
Envelhecimento , Biossíntese de Proteínas , RNA Polimerase I , Envelhecimento/genética , Animais , RNA Polimerases Dirigidas por DNA , Humanos , Análise da Randomização Mendeliana , RNA Polimerase I/metabolismo , Proteínas Ribossômicas/genética , Ribossomos/genética , Ribossomos/metabolismo
4.
Nat Aging ; 2(12): 1176-1190, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-37118537

RESUMO

A transient, homeostatic transcriptional response can result in transcriptional memory, programming subsequent transcriptional outputs. Transcriptional memory has great but unappreciated potential to alter animal aging as animals encounter a multitude of diverse stimuli throughout their lifespan. Here we show that activating an evolutionarily conserved, longevity-promoting transcription factor, dFOXO, solely in early adulthood of female fruit flies is sufficient to improve their subsequent health and survival in midlife and late life. This youth-restricted dFOXO activation causes persistent changes to chromatin landscape in the fat body and requires chromatin remodelers such as the SWI/SNF and ISWI complexes to program health and longevity. Chromatin remodeling is accompanied by a long-lasting transcriptional program that is distinct from that observed during acute dFOXO activation and includes induction of Xbp1. We show that this later-life induction of Xbp1 is sufficient to curtail later-life mortality. Our study demonstrates that transcriptional memory can profoundly alter how animals age.


Assuntos
Montagem e Desmontagem da Cromatina , Proteínas de Drosophila , Animais , Feminino , Montagem e Desmontagem da Cromatina/genética , Fatores de Transcrição/genética , Regulação da Expressão Gênica , Drosophila/metabolismo , Cromatina/genética , Proteínas de Ligação a DNA/genética
5.
Front Genet ; 12: 758135, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34539762

RESUMO

[This corrects the article DOI: 10.3389/fgene.2021.705122.].

6.
Front Genet ; 12: 705122, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34295356

RESUMO

Transcription in eukaryotic cells is performed by three RNA polymerases. RNA polymerase I synthesises most rRNAs, whilst RNA polymerase II transcribes all mRNAs and many non-coding RNAs. The largest of the three polymerases is RNA polymerase III (Pol III) which transcribes a variety of short non-coding RNAs including tRNAs and the 5S rRNA, in addition to other small RNAs such as snRNAs, snoRNAs, SINEs, 7SL RNA, Y RNA, and U6 spilceosomal RNA. Pol III-mediated transcription is highly dynamic and regulated in response to changes in cell growth, cell proliferation and stress. Pol III-generated transcripts are involved in a wide variety of cellular processes, including translation, genome and transcriptome regulation and RNA processing, with Pol III dys-regulation implicated in diseases including leukodystrophy, Alzheimer's, Fragile X-syndrome and various cancers. More recently, Pol III was identified as an evolutionarily conserved determinant of organismal lifespan acting downstream of mTORC1. Pol III inhibition extends lifespan in yeast, worms and flies, and in worms and flies acts from the intestine and intestinal stem cells respectively to achieve this. Intriguingly, Pol III activation achieved through impairment of its master repressor, Maf1, has also been shown to promote longevity in model organisms, including mice. In this review we introduce the Pol III transcription apparatus and review the current understanding of RNA Pol III's role in ageing and lifespan in different model organisms. We then discuss the potential of Pol III as a therapeutic target to improve age-related health in humans.

8.
Trends Genet ; 36(5): 373-382, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32294417

RESUMO

The increasing number of older people is resulting in an increased prevalence of age-related diseases. Research has shown that the ageing process itself is a potential point of intervention. Indeed, gene expression can be optimised for health in older ages through manipulation of transcription factor (TF) activity. This review is focused on the ever-growing number of TFs whose effects on ageing are evolutionarily conserved. These regulate a plethora of functions, including stress resistance, metabolism, and growth. They are engaged in complex interactions within and between different cell types, impacting the physiology of the entire organism. Since ageing is not programmed, the conservation of their effects on lifespan is most likely a reflection of the conservation of their functions in youth.


Assuntos
Sequência Conservada/genética , Evolução Molecular , Longevidade/genética , Fatores de Transcrição/genética , Envelhecimento/genética , Regulação da Expressão Gênica/genética , Humanos
9.
Aging Cell ; 19(5): e13137, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32291952

RESUMO

Inhibition of signalling through several receptor tyrosine kinases (RTKs), including the insulin-like growth factor receptor and its orthologues, extends healthy lifespan in organisms from diverse evolutionary taxa. This raises the possibility that other RTKs, including those already well studied for their roles in cancer and developmental biology, could be promising targets for extending healthy lifespan. Here, we focus on anaplastic lymphoma kinase (Alk), an RTK with established roles in nervous system development and in multiple cancers, but whose effects on aging remain unclear. We find that several means of reducing Alk signalling, including mutation of its ligand jelly belly (jeb), RNAi knock-down of Alk, or expression of dominant-negative Alk in adult neurons, can extend healthy lifespan in female, but not male, Drosophila. Moreover, reduced Alk signalling preserves neuromuscular function with age, promotes resistance to starvation and xenobiotic stress, and improves night sleep consolidation. We find further that inhibition of Alk signalling in adult neurons modulates the expression of several insulin-like peptides, providing a potential mechanistic link between neuronal Alk signalling and organism-wide insulin-like signalling. Finally, we show that TAE-684, a small molecule inhibitor of Alk, can extend healthy lifespan in Drosophila, suggesting that the repurposing of Alk inhibitors may be a promising direction for strategies to promote healthy aging.


Assuntos
Quinase do Linfoma Anaplásico/metabolismo , Longevidade , Quinase do Linfoma Anaplásico/antagonistas & inibidores , Animais , Senescência Celular/efeitos dos fármacos , Drosophila , Feminino , Longevidade/efeitos dos fármacos , Masculino , Inibidores de Proteínas Quinases/farmacologia , Transdução de Sinais/efeitos dos fármacos
10.
Cell Rep ; 30(6): 1661-1669.e4, 2020 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-32049000

RESUMO

Health and survival in old age can be improved by changes in gene expression. RNA polymerase (Pol) I is the essential, conserved enzyme whose task is to generate the pre-ribosomal RNA (rRNA). We find that reducing the levels of Pol I activity is sufficient to extend lifespan in the fruit fly. This effect can be recapitulated by partial, adult-restricted inhibition, with both enterocytes and stem cells of the adult midgut emerging as important cell types. In stem cells, Pol I appears to act in the same longevity pathway as Pol III, implicating rRNA synthesis in these cells as the key lifespan determinant. Importantly, reduction in Pol I activity delays broad, age-related impairment and pathology, improving the function of diverse organ systems. Hence, our study shows that Pol I activity in the adult drives systemic, age-related decline in animal health and anticipates mortality.


Assuntos
Proteínas de Drosophila/genética , Drosophila melanogaster/genética , RNA Polimerase I/antagonistas & inibidores , Animais , Longevidade
11.
Sci Rep ; 10(1): 3418, 2020 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-32099025

RESUMO

The Insulin/IGF-1 signalling (IIS) pathway plays an essential role in the regulation of glucose and lipid homeostasis. At the same time, a reduction in the IIS pathway activity can extend lifespan and healthspan in various model organisms. Amongst a number of body organs that sense and respond to insulin/IGF-1, the adipose tissue has a central role in both the metabolic and lifespan effects of IIS at the organismal level. Genetic inactivation of IIS components specifically in the adipose tissue has been shown before to improve metabolic profile and extend lifespan in various model organisms. We sought to identify conserved molecular mechanisms that may underlie the beneficial effects of IIS inhibition in the adipose tissue, specifically at the level of phosphoinositide 3-kinase (PI3K), a key IIS effector molecule. To this end, we inactivated PI3K by genetic means in the fly fat body and by pharmacological inhibition in mammalian adipocytes. Gene expression studies revealed changes to metabolism and upregulation of mitochondrial activity in mouse adipocytes and fly fat bodies with downregulated PI3K, which were confirmed by biochemical assays in mammalian adipocytes. These data suggest that PI3K inactivation has a conserved effect of upregulating mitochondrial metabolism in both fly and mammalian adipose tissue, which likely contributes to the health- and life-span extending effect of IIS pathway downregulation.


Assuntos
Tecido Adiposo/metabolismo , Proteínas de Drosophila/metabolismo , Insulina/metabolismo , Metabolismo dos Lipídeos , Mitocôndrias/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Células 3T3-L1 , Animais , Proteínas de Drosophila/genética , Drosophila melanogaster , Insulina/genética , Camundongos , Mitocôndrias/genética , Fosfatidilinositol 3-Quinases/genética
12.
PLoS Genet ; 15(7): e1008212, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31356597

RESUMO

Ageing populations pose one of the main public health crises of our time. Reprogramming gene expression by altering the activities of sequence-specific transcription factors (TFs) can ameliorate deleterious effects of age. Here we explore how a circuit of TFs coordinates pro-longevity transcriptional outcomes, which reveals a multi-tissue and multi-species role for an entire protein family: the E-twenty-six (ETS) TFs. In Drosophila, reduced insulin/IGF signalling (IIS) extends lifespan by coordinating activation of Aop, an ETS transcriptional repressor, and Foxo, a Forkhead transcriptional activator. Aop and Foxo bind the same genomic loci, and we show that, individually, they effect similar transcriptional programmes in vivo. In combination, Aop can both moderate or synergise with Foxo, dependent on promoter context. Moreover, Foxo and Aop oppose the gene-regulatory activity of Pnt, an ETS transcriptional activator. Directly knocking down Pnt recapitulates aspects of the Aop/Foxo transcriptional programme and is sufficient to extend lifespan. The lifespan-limiting role of Pnt appears to be balanced by a requirement for metabolic regulation in young flies, in which the Aop-Pnt-Foxo circuit determines expression of metabolic genes, and Pnt regulates lipolysis and responses to nutrient stress. Molecular functions are often conserved amongst ETS TFs, prompting us to examine whether other Drosophila ETS-coding genes may also affect ageing. We show that five out of eight Drosophila ETS TFs play a role in fly ageing, acting from a range of organs and cells including the intestine, adipose and neurons. We expand the repertoire of lifespan-limiting ETS TFs in C. elegans, confirming their conserved function in ageing and revealing that the roles of ETS TFs in physiology and lifespan are conserved throughout the family, both within and between species.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila/fisiologia , Proteínas do Olho/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Repressoras/metabolismo , Fatores de Transcrição/metabolismo , Tecido Adiposo/metabolismo , Animais , Proteínas de Ligação a DNA/genética , Drosophila/genética , Proteínas de Drosophila/genética , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Mucosa Intestinal/metabolismo , Lipólise , Longevidade , Redes e Vias Metabólicas , Proteínas do Tecido Nervoso/genética , Neurônios/metabolismo , Proteínas Proto-Oncogênicas/genética , Fatores de Transcrição/genética
13.
Aging (Albany NY) ; 9(12): 2521-2528, 2017 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-29207375

RESUMO

Lifespan and health in older age are strongly influenced by diet. Feeding Drosophila melanogaster diets high in sugar has increasingly been used as an experimental model to understand the physiological effects of unhealthy, contemporary human diets. Several metabolic parameters and physiological responses to nutrition are known to be dependent on the sex of the animal. However, sexual dimorphism in the responses to high-sugar diets in fruit flies has not been examined. Here we show that a high-sugar diet in Drosophila melanogaster elicits sexually dimorphic effects on feeding behaviour, starvation resistance and lifespan. Females feed less on such diets, while males feed more, and these feeding responses may have secondary consequences. Females, more than males, gain the ability to resist periods of starvation from high-sugar diets, indicating that the female response to excess sugar may be geared towards surviving food shortages in early life. At the same time, female lifespan is more susceptible to the detrimental effects of high sugar diets. Our study reveals differences between Drosophila sexes in their responses to sugar-rich diets, indicating the fruit fly could be used as a model to understand the sexually dimorphic features of human metabolic health.


Assuntos
Açúcares da Dieta/efeitos adversos , Comportamento Alimentar/fisiologia , Longevidade/fisiologia , Caracteres Sexuais , Animais , Drosophila melanogaster , Feminino , Masculino
14.
Nature ; 552(7684): 263-267, 2017 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-29186112

RESUMO

Three distinct RNA polymerases transcribe different classes of genes in the eukaryotic nucleus. RNA polymerase (Pol) III is the essential, evolutionarily conserved enzyme that generates short, non-coding RNAs, including tRNAs and 5S rRNA. The historical focus on transcription of protein-coding genes has left the roles of Pol III in organismal physiology relatively unexplored. Target of rapamycin kinase complex 1 (TORC1) regulates Pol III activity, and is also an important determinant of longevity. This raises the possibility that Pol III is involved in ageing. Here we show that Pol III limits lifespan downstream of TORC1. We find that a reduction in Pol III extends chronological lifespan in yeast and organismal lifespan in worms and flies. Inhibiting the activity of Pol III in the gut of adult worms or flies is sufficient to extend lifespan; in flies, longevity can be achieved by Pol III inhibition specifically in intestinal stem cells. The longevity phenotype is associated with amelioration of age-related gut pathology and functional decline, dampened protein synthesis and increased tolerance of proteostatic stress. Pol III acts on lifespan downstream of TORC1, and limiting Pol III activity in the adult gut achieves the full longevity benefit of systemic TORC1 inhibition. Hence, Pol III is a pivotal mediator of this key nutrient-signalling network for longevity; the growth-promoting anabolic activity of Pol III mediates the acceleration of ageing by TORC1. The evolutionary conservation of Pol III affirms its potential as a therapeutic target.


Assuntos
Longevidade/fisiologia , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , RNA Polimerase III/metabolismo , Envelhecimento/efeitos dos fármacos , Envelhecimento/fisiologia , Animais , Caenorhabditis elegans/efeitos dos fármacos , Caenorhabditis elegans/enzimologia , Caenorhabditis elegans/fisiologia , Drosophila melanogaster/efeitos dos fármacos , Drosophila melanogaster/enzimologia , Drosophila melanogaster/fisiologia , Evolução Molecular , Feminino , Alimentos , Intestinos/citologia , Intestinos/enzimologia , Longevidade/efeitos dos fármacos , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina/antagonistas & inibidores , Biossíntese de Proteínas , RNA Polimerase III/antagonistas & inibidores , RNA Polimerase III/deficiência , Saccharomyces cerevisiae/efeitos dos fármacos , Saccharomyces cerevisiae/enzimologia , Saccharomyces cerevisiae/fisiologia , Células-Tronco/citologia , Células-Tronco/enzimologia
15.
Cell Rep ; 21(3): 641-653, 2017 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-29045833

RESUMO

Reduced activity of nutrient-sensing signaling networks can extend organismal lifespan, yet the underlying biology remains unclear. We show that the anti-aging effects of rapamycin and reduced intestinal insulin/insulin growth factor (IGF) signaling (IIS) require the Drosophila FoxA transcription factor homolog Fork Head (FKH). Intestinal FKH induction extends lifespan, highlighting a role for the gut. FKH binds to and is phosphorylated by AKT and Target of Rapamycin. Gut-specific FKH upregulation improves gut barrier function in aged flies. Additionally, it increases the expression of nutrient transporters, as does lowered IIS. Evolutionary conservation of this effect of lowered IIS is suggested by the upregulation of related nutrient transporters in insulin receptor substrate 1 knockout mouse intestine. Our study highlights a critical role played by FKH in the gut in mediating anti-aging effects of reduced IIS. Malnutrition caused by poor intestinal absorption is a major problem in the elderly, and a better understanding of the mechanisms involved will have important therapeutic implications for human aging.


Assuntos
Drosophila melanogaster/metabolismo , Drosophila melanogaster/fisiologia , Alimentos , Fatores de Transcrição Forkhead/metabolismo , Absorção Intestinal , Mucosa Intestinal/metabolismo , Longevidade , Proteínas Nucleares/metabolismo , Animais , Restrição Calórica , Diferenciação Celular/efeitos dos fármacos , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Proteínas de Drosophila/metabolismo , Enterócitos/efeitos dos fármacos , Enterócitos/metabolismo , Feminino , Insulina/metabolismo , Proteínas Substratos do Receptor de Insulina/metabolismo , Absorção Intestinal/efeitos dos fármacos , Intestinos/citologia , Longevidade/efeitos dos fármacos , Proteínas de Membrana Transportadoras/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Sirolimo/farmacologia , Somatomedinas/metabolismo , Transcrição Gênica/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos
16.
Mol Syst Biol ; 13(9): 939, 2017 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-28916541

RESUMO

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.


Assuntos
Envelhecimento/metabolismo , Drosophila/metabolismo , Redes Reguladoras de Genes , Proteômica/métodos , Animais , Proteínas de Drosophila , Corpo Adiposo/metabolismo , Insulina/metabolismo , Mucosa Intestinal/metabolismo , Modelos Animais , Mutação , Especificidade de Órgãos , Proteínas Ribossômicas/metabolismo
17.
Cell Rep ; 18(2): 299-306, 2017 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-28076775

RESUMO

Consumption of unhealthy diets is exacerbating the burden of age-related ill health in aging populations. Such diets can program mammalian physiology to cause long-term, detrimental effects. Here, we show that, in Drosophila melanogaster, an unhealthy, high-sugar diet in early adulthood programs lifespan to curtail later-life survival despite subsequent dietary improvement. Excess dietary sugar promotes insulin-like signaling, inhibits dFOXO-the Drosophila homolog of forkhead box O (FOXO) transcription factors-and represses expression of dFOXO target genes encoding epigenetic regulators. Crucially, dfoxo is required both for transcriptional changes that mark the fly's dietary history and for nutritional programming of lifespan by excess dietary sugar, and this mechanism is conserved in Caenorhabditis elegans. Our study implicates FOXO factors, the evolutionarily conserved determinants of animal longevity, in the mechanisms of nutritional programming of animal lifespan.


Assuntos
Fenômenos Fisiológicos da Nutrição Animal , Dieta , Carboidratos da Dieta/farmacologia , Proteínas de Drosophila/antagonistas & inibidores , Drosophila melanogaster/fisiologia , Fatores de Transcrição Forkhead/antagonistas & inibidores , Longevidade , Animais , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/efeitos dos fármacos , Drosophila melanogaster/genética , Fatores de Transcrição Forkhead/metabolismo , Análise de Sobrevida , Transcrição Gênica/efeitos dos fármacos
18.
Cell Metab ; 23(3): 403-4, 2016 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-26959183

RESUMO

Age-related cognitive decline is one of the most haunting aspects of human aging. In a recent publication, Coleen Murphy and colleagues (Kaletsky et al., 2016) describe the transcriptional program that maintains youthful function of aging neurons in the nematode worm.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/genética , Fatores de Transcrição Forkhead/metabolismo , Insulina/metabolismo , Neurônios/metabolismo , Fenótipo , Transdução de Sinais , Somatomedinas/metabolismo , Transcriptoma , Animais
19.
Sci Rep ; 6: 23102, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26976084

RESUMO

Human Tau (hTau) is a highly soluble and natively unfolded protein that binds to microtubules within neurons. Its dysfunction and aggregation into insoluble paired helical filaments is involved in the pathogenesis of Alzheimer's disease (AD), constituting, together with accumulated ß-amyloid (Aß) peptides, a hallmark of the disease. Deciphering both the loss-of-function and toxic gain-of-function of hTau proteins is crucial to further understand the mechanisms leading to neurodegeneration in AD. As the fruit fly Drosophila melanogaster expresses Tau proteins (dTau) that are homologous to hTau, we aimed to better comprehend dTau functions by generating a specific tau knock-out (KO) fly line using homologous recombination. We observed that the specific removal of endogenous dTau proteins did not lead to overt, macroscopic phenotypes in flies. Indeed, survival, climbing ability and neuronal function were unchanged in tau KO flies. In addition, we did not find any overt positive or negative effect of dTau removal on human Aß-induced toxicity. Altogether, our results indicate that the absence of dTau proteins has no major functional impact on flies, and suggests that our tau KO strain is a relevant model to further investigate the role of dTau proteins in vivo, thereby giving additional insights into hTau functions.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Proteínas tau/genética , Animais , Animais Geneticamente Modificados , Drosophila melanogaster/fisiologia , Técnicas de Silenciamento de Genes , Expectativa de Vida , Locomoção , Neurônios/fisiologia
20.
Proc Natl Acad Sci U S A ; 113(5): 1321-6, 2016 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-26787908

RESUMO

Lifespan of laboratory animals can be increased by genetic, pharmacological, and dietary interventions. Increased expression of genes involved in xenobiotic metabolism, together with resistance to xenobiotics, are frequent correlates of lifespan extension in the nematode worm Caenorhabditis elegans, the fruit fly Drosophila, and mice. The Green Theory of Aging suggests that this association is causal, with the ability of cells to rid themselves of lipophilic toxins limiting normal lifespan. To test this idea, we experimentally increased resistance of Drosophila to the xenobiotic dichlordiphenyltrichlorethan (DDT), by artificial selection or by transgenic expression of a gene encoding a cytochrome P450. Although both interventions increased DDT resistance, neither increased lifespan. Furthermore, dietary restriction increased lifespan without increasing xenobiotic resistance, confirming that the two traits can be uncoupled. Reduced activity of the insulin/Igf signaling (IIS) pathway increases resistance to xenobiotics and extends lifespan in Drosophila, and can also increase longevity in C. elegans, mice, and possibly humans. We identified a nuclear hormone receptor, DHR96, as an essential mediator of the increased xenobiotic resistance of IIS mutant flies. However, the IIS mutants remained long-lived in the absence of DHR96 and the xenobiotic resistance that it conferred. Thus, in Drosophila IIS mutants, increased xenobiotic resistance and enhanced longevity are not causally connected. The frequent co-occurrence of the two traits may instead have evolved because, in nature, lowered IIS can signal the presence of pathogens. It will be important to determine whether enhanced xenobiotic metabolism is also a correlated, rather than a causal, trait in long-lived mice.


Assuntos
Proteínas de Drosophila/fisiologia , Drosophila/genética , Insulina/genética , Mutação , Receptores Citoplasmáticos e Nucleares/fisiologia , Xenobióticos/farmacologia , Animais , Resistência a Medicamentos , Expectativa de Vida , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...