Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Nature ; 616(7958): 814-821, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-37046086

RESUMO

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.


Assuntos
Envelhecimento , Longevidade , Elongação da Transcrição Genética , Animais , Humanos , Camundongos , Ratos , Envelhecimento/genética , Insulina/metabolismo , Longevidade/genética , RNA Polimerase II/genética , RNA Polimerase II/metabolismo , Transdução de Sinais , Drosophila melanogaster/genética , Caenorhabditis elegans/genética , RNA Circular , Somatomedinas , Nucleossomos , Histonas , Divisão Celular , Restrição Calórica
2.
Mol Cell ; 79(2): 268-279.e5, 2020 07 16.
Artigo em Inglês | MEDLINE | ID: mdl-32592682

RESUMO

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.


Assuntos
Drosophila/fisiologia , Insulina/fisiologia , Longevidade/genética , RNA Circular/fisiologia , Envelhecimento , Animais , Animais Geneticamente Modificados , Drosophila/genética , Proteínas de Drosophila/genética , Feminino , Masculino , Mutação , Neurônios/fisiologia , Sulfotransferases/genética , Transcriptoma
3.
Nucleic Acids Res ; 52(5): 2093-2111, 2024 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-38303573

RESUMO

Co-transcriptional processing of nascent pre-mRNAs by the spliceosome is vital to regulating gene expression and maintaining genome integrity. Here, we show that the deficiency of functional U5 small nuclear ribonucleoprotein particles (snRNPs) in Drosophila imaginal cells causes extensive transcriptome remodeling and accumulation of highly mutagenic R-loops, triggering a robust stress response and cell cycle arrest. Despite compromised proliferative capacity, the U5 snRNP-deficient cells increased protein translation and cell size, causing intra-organ growth disbalance before being gradually eliminated via apoptosis. We identify the Xrp1-Irbp18 heterodimer as the primary driver of transcriptional and cellular stress program downstream of U5 snRNP malfunction. Knockdown of Xrp1 or Irbp18 in U5 snRNP-deficient cells attenuated JNK and p53 activity, restored normal cell cycle progression and growth, and inhibited cell death. Reducing Xrp1-Irbp18, however, did not rescue the splicing defects, highlighting the requirement of accurate splicing for cellular and tissue homeostasis. Our work provides novel insights into the crosstalk between splicing and the DNA damage response and defines the Xrp1-Irbp18 heterodimer as a critical sensor of spliceosome malfunction and mediator of the stress-induced cellular senescence program.


The removal of introns and the joining of exons into mature mRNA by the spliceosome is crucial in regulating gene expression, simultaneously safeguarding genome integrity and enhancing proteome diversity in multicellular organisms. Spliceosome dysfunction is thus associated with various diseases and organismal aging. Our study describes the cascade of events in response to spliceosome dysfunction. We identified two transcription factors as drivers of a stress response program triggered by spliceosome dysfunction, which dramatically remodel gene expression to protect tissue integrity and induce a senescent-like state in damaged cells prior to their inevitable elimination. Together, we highlight the indispensable role of spliceosomes in maintaining homeostasis and implicate spliceosome dysfunction in senescent cell accumulation associated with the pathomechanisms of spliceopathies and aging.


Assuntos
Proteínas de Ligação a DNA , Ribonucleoproteína Nuclear Pequena U5 , Spliceossomos , Ribonucleoproteína Nuclear Pequena U4-U6/metabolismo , Ribonucleoproteína Nuclear Pequena U5/metabolismo , Precursores de RNA/genética , Precursores de RNA/metabolismo , Splicing de RNA/genética , Spliceossomos/genética , Spliceossomos/metabolismo , Animais , Drosophila melanogaster , Proteínas de Ligação a DNA/metabolismo
4.
Proc Natl Acad Sci U S A ; 116(42): 20817-20819, 2019 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-31570569

RESUMO

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.


Assuntos
Envelhecimento/efeitos dos fármacos , Drosophila/efeitos dos fármacos , Lítio/farmacologia , Longevidade/efeitos dos fármacos , Nutrientes/metabolismo , Piridonas/farmacologia , Pirimidinonas/farmacologia , Sirolimo/farmacologia , Idoso , Envelhecimento/metabolismo , Animais , Drosophila/genética , Drosophila/crescimento & desenvolvimento , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Combinação de Medicamentos , Feminino , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Quinase 3 da Glicogênio Sintase/genética , Quinase 3 da Glicogênio Sintase/metabolismo , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina/antagonistas & inibidores , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Pessoa de Meia-Idade , Transdução de Sinais/efeitos dos fármacos
5.
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
6.
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
7.
PLoS Biol ; 12(4): e1001824, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24690889

RESUMO

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.


Assuntos
Drosophila/metabolismo , Privação do Sono/metabolismo , Sono/fisiologia , Somatomedinas/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Envelhecimento , Animais , Ritmo Circadiano/genética , Ritmo Circadiano/fisiologia , Dopamina/biossíntese , Dopamina/metabolismo , Drosophila/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Feminino , Fatores de Transcrição Forkhead/metabolismo , Proteínas Inibidoras de Apoptose/genética , Hormônios de Inseto/metabolismo , Insulina/metabolismo , Octopamina/metabolismo , Oligopeptídeos/metabolismo , Ácido Pirrolidonocarboxílico/análogos & derivados , Ácido Pirrolidonocarboxílico/metabolismo , Receptor de Insulina/genética , Receptores Dopaminérgicos/biossíntese , Receptores de Glucagon/genética , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Transdução de Sinais , Sirolimo/farmacologia , Somatomedinas/biossíntese , Somatomedinas/genética , Serina-Treonina Quinases TOR/antagonistas & inibidores
8.
Nucleic Acids Res ; 42(22): 13920-38, 2014 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-25428350

RESUMO

Members of the pentatricopeptide repeat domain (PPR) protein family bind RNA and are important for post-transcriptional control of organelle gene expression in unicellular eukaryotes, metazoans and plants. They also have a role in human pathology, as mutations in the leucine-rich PPR-containing (LRPPRC) gene cause severe neurodegeneration. We have previously shown that the mammalian LRPPRC protein and its Drosophila melanogaster homolog DmLRPPRC1 (also known as bicoid stability factor) are necessary for mitochondrial translation by controlling stability and polyadenylation of mRNAs. We here report characterization of DmLRPPRC2, a second fruit fly homolog of LRPPRC, and show that it has a predominant mitochondrial localization and interacts with a stem-loop interacting RNA binding protein (DmSLIRP2). Ubiquitous downregulation of DmLrpprc2 expression causes respiratory chain dysfunction, developmental delay and shortened lifespan. Unexpectedly, decreased DmLRPPRC2 expression does not globally affect steady-state levels or polyadenylation of mitochondrial transcripts. However, some mitochondrial transcripts abnormally associate with the mitochondrial ribosomes and some products are dramatically overproduced and other ones decreased, which, in turn, results in severe deficiency of respiratory chain complexes. The function of DmLRPPRC2 thus seems to be to ensure that mitochondrial transcripts are presented to the mitochondrial ribosomes in an orderly fashion to avoid poorly coordinated translation.


Assuntos
Proteínas de Drosophila/fisiologia , Mitocôndrias/genética , Proteínas Mitocondriais/fisiologia , Biossíntese de Proteínas , Animais , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/crescimento & desenvolvimento , Drosophila melanogaster/metabolismo , Transporte de Elétrons , Longevidade , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Poliadenilação , RNA/metabolismo , Interferência de RNA , RNA Mitocondrial , Transcrição Gênica
9.
PLoS Genet ; 9(1): e1003178, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23300484

RESUMO

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.


Assuntos
Proteínas de Drosophila , Drosophila melanogaster/genética , Mitocôndrias , Proteínas Mitocondriais , Ribossomos , Animais , DNA Mitocondrial/genética , Proteínas de Drosophila/genética , Regulação da Expressão Gênica , Invertebrados/genética , Invertebrados/metabolismo , Camundongos , Mitocôndrias/genética , Mitocôndrias/metabolismo , Proteínas Mitocondriais/genética , Fosforilação Oxidativa , Ribossomos/genética , Ribossomos/metabolismo , Transcrição Gênica
10.
Front Cell Dev Biol ; 9: 625715, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33634126

RESUMO

Proteostasis collapses during aging resulting, among other things, in the accumulation of damaged and aggregated proteins. The proteasome is the main cellular proteolytic system and plays a fundamental role in the maintenance of protein homeostasis. Our previous work has demonstrated that senescence and aging are related to a decline in proteasome content and activities, while its activation extends lifespan in vitro and in vivo in various species. However, the mechanisms underlying this age-related decline of proteasome function and the down-regulation in expression of its subunits remain largely unclear. Here, we demonstrate that the Forkhead box-O1 (FoxO1) transcription factor directly regulates the expression of a 20S proteasome catalytic subunit and, hence, proteasome activity. Specifically, we demonstrate that knockout of FoxO1, but not of FoxO3, in mice severely impairs proteasome activity in several tissues, while depletion of IRS1 enhances proteasome function. Importantly, we show that FoxO1 directly binds on the promoter region of the rate-limiting catalytic ß5 proteasome subunit to regulate its expression. In summary, this study reveals the direct role of FoxO factors in the regulation of proteasome function and provides new insight into how FoxOs affect proteostasis and, in turn, longevity.

11.
Elife ; 102021 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-33879316

RESUMO

Reduced activity of the insulin/IGF signalling network increases health during ageing in multiple species. Diverse and tissue-specific mechanisms drive the health improvement. Here, we performed tissue-specific transcriptional and proteomic profiling of long-lived Drosophila dilp2-3,5 mutants, and identified tissue-specific regulation of >3600 transcripts and >3700 proteins. Most expression changes were regulated post-transcriptionally in the fat body, and only in mutants infected with the endosymbiotic bacteria, Wolbachia pipientis, which increases their lifespan. Bioinformatic analysis identified reduced co-translational ER targeting of secreted and membrane-associated proteins and increased DNA damage/repair response proteins. Accordingly, age-related DNA damage and genome instability were lower in fat body of the mutant, and overexpression of a minichromosome maintenance protein subunit extended lifespan. Proteins involved in carbohydrate metabolism showed altered expression in the mutant intestine, and gut-specific overexpression of a lysosomal mannosidase increased autophagy, gut homeostasis, and lifespan. These processes are candidates for combatting ageing-related decline in other organisms.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Insulina/fisiologia , Proteoma/metabolismo , Transdução de Sinais , Transcriptoma , Wolbachia/fisiologia , Animais , Drosophila melanogaster/metabolismo , Drosophila melanogaster/microbiologia , Corpo Adiposo/metabolismo , Perfilação da Expressão Gênica
12.
Aging Cell ; 19(1): e13043, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31721422

RESUMO

Reduced insulin/IGF signaling (IIS) extends lifespan in multiple organisms. Different processes in different tissues mediate this lifespan extension, with a set of interplays that remain unclear. We here show that, in Drosophila, reduced IIS activity modulates methionine metabolism, through tissue-specific regulation of glycine N-methyltransferase (Gnmt), and that this regulation is required for full IIS-mediated longevity. Furthermore, fat body-specific expression of Gnmt was sufficient to extend lifespan. Targeted metabolomics showed that reducing IIS activity led to a Gnmt-dependent increase in spermidine levels. We also show that both spermidine treatment and reduced IIS activity are sufficient to extend the lifespan of Drosophila, but only in the presence of Gnmt. This extension of lifespan was associated with increased levels of autophagy. Finally, we found that increased expression of Gnmt occurs in the liver of liver-specific IRS1 KO mice and is thus an evolutionarily conserved response to reduced IIS. The discovery of Gnmt and spermidine as tissue-specific modulators of IIS-mediated longevity may aid in developing future therapeutic treatments to ameliorate aging and prevent disease.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Glicina N-Metiltransferase/metabolismo , Insulina/metabolismo , Longevidade/efeitos dos fármacos , Espermidina/metabolismo , Animais , Transdução de Sinais
13.
BMC Dev Biol ; 8: 28, 2008 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-18366811

RESUMO

BACKGROUND: Dietary restriction (DR) results in increased longevity, reduced fecundity and reduced growth in many organisms. Though many studies have examined the effects of DR on longevity and fecundity, few have investigated the effects on growth. RESULTS: Here we use Caenorhabditis elegans to determine the mechanisms that regulate growth under DR. We show that rather than a reduction in cell number, decreased growth in wild type C. elegans under DR is correlated with lower levels of hypodermal polyploidization. We also show that mutants lacking wild type sensory ciliated neurons are small, exhibit hypo-polyploidization and more importantly, when grown under DR, reduce their levels of endoreduplication to a lesser extent than wild type, suggesting that these neurons are required for the regulation of hypodermal polyploidization in response to DR. Similarly, we also show that the cGMP-dependent protein kinase EGL-4 and the SMA/MAB signalling pathway regulate polyploidization under DR. CONCLUSION: We show C. elegans is capable of actively responding to food levels to regulate adult ploidy. We suggest this response is dependent on the SMA/MAB signalling pathway.


Assuntos
Caenorhabditis elegans/citologia , Caenorhabditis elegans/crescimento & desenvolvimento , Restrição Calórica , Derme/crescimento & desenvolvimento , Dieta , Poliploidia , Animais , Tamanho Corporal , Proteínas de Caenorhabditis elegans/metabolismo , Cílios/metabolismo , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Comportamento Alimentar , Fertilidade , Alimentos , Proteínas de Helminto/metabolismo , Estágios do Ciclo de Vida , Longevidade , Modelos Biológicos , Neurônios/metabolismo , Neuropeptídeos/metabolismo , Fator de Crescimento Transformador beta/metabolismo
14.
Elife ; 62017 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-28699890

RESUMO

Ageing is a progressive decline of intrinsic physiological functions. We examined the impact of ageing on the ultrastructure and function of mitochondria in mouse and fruit flies (Drosophila melanogaster) by electron cryo-tomography and respirometry. We discovered distinct age-related changes in both model organisms. Mitochondrial function and ultrastructure are maintained in mouse heart, whereas subpopulations of mitochondria from mouse liver show age-related changes in membrane morphology. Subpopulations of mitochondria from young and old mouse kidney resemble those described for apoptosis. In aged flies, respiratory activity is compromised and the production of peroxide radicals is increased. In about 50% of mitochondria from old flies, the inner membrane organization breaks down. This establishes a clear link between inner membrane architecture and functional decline. Mitochondria were affected by ageing to very different extents, depending on the organism and possibly on the degree to which tissues within the same organism are protected against mitochondrial damage.


Assuntos
Envelhecimento/fisiologia , Drosophila melanogaster/fisiologia , Mitocôndrias/ultraestrutura , Animais , Microscopia Crioeletrônica , DNA Mitocondrial/genética , Metabolismo Energético , Imageamento Tridimensional , Camundongos , Mitocôndrias/metabolismo , Especificidade de Órgãos , Tomografia
15.
Proc Biol Sci ; 273(1605): 3039-45, 2006 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-17015346

RESUMO

Manipulative parasites can alter the phenotype of intermediate hosts in various ways. However, it is unclear whether such changes are just by-products of infection or adaptive and enhance transmission to the final host. Here, we show that the alteration of serotonergic activity is functionally linked to the alteration of specific behaviour in the amphipod Gammarus pulex infected with acanthocephalan parasites. Pomphorhynchus laevis and, to a lesser extent, Pomphorhynchus tereticollis altered phototactism, but not geotactism, in G. pulex, whereas the reverse was true for Polymorphus minutus. Serotonin (5-hydroxytryptamine, 5-HT) injected to uninfected G. pulex mimicked the altered phototactism, but had no effect on geotactism. Photophilic G. pulex infected with P. laevis or P. tereticollis showed a 40% increase in brain 5-HT immunoreactivity compared to photophobic, uninfected individuals. In contrast, brain 5-HT immunoreactivity did not differ between P. minutus-infected and uninfected G. pulex. Finally, brain 5-HT immunoreactivity differed significantly among P. tereticollis-infected individuals in accordance with their degree of manipulation. Our results demonstrate that altered 5-HT activity is not the mere consequence of infection by acanthocephalans but is specifically linked to the disruption of host photophobic behaviour, whereas the alteration of other behaviours such as geotactism may rely on distinct physiological routes.


Assuntos
Acantocéfalos/fisiologia , Anfípodes/parasitologia , Comportamento Animal , Encéfalo/parasitologia , Serotonina/metabolismo , Anfípodes/metabolismo , Anfípodes/fisiologia , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Interações Hospedeiro-Parasita
16.
Sci Rep ; 6: 30290, 2016 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-27452396

RESUMO

Down-regulation of insulin/insulin-like growth factor signaling (IIS) can increase lifespan in C. elegans, Drosophila and mice. In C. elegans, reduced IIS results in down-regulation of translation, which itself can extend lifespan. However, the effect of reduced IIS on translation has yet to be determined in other multicellular organisms. Using two long-lived IIS models, namely Drosophila lacking three insulin-like peptides (dilp2-3,5(-/-)) and mice lacking insulin receptor substrate 1 (Irs1(-/-)), and two independent translation assays, polysome profiling and radiolabeled amino acid incorporation, we show that reduced IIS lowers translation in these organisms. In Drosophila, reduced IIS decreased polysome levels in fat body and gut, but reduced the rate of protein synthesis only in the fat body. Reduced IIS in mice decreased protein synthesis rate only in skeletal muscle, without reducing polysomes in any tissue. This lowered translation in muscle was independent of Irs1 loss in the muscle itself, but a secondary effect of Irs1 loss in the liver. In conclusion, down-regulation of translation is an evolutionarily conserved response to reduced IIS, but the tissues in which it occurs can vary between organisms. Furthermore, the mechanisms underlying lowered translation may differ in mice, possibly associated with the complexity of the regulatory processes.


Assuntos
Proteínas de Drosophila/genética , Proteínas Substratos do Receptor de Insulina/genética , Insulinas/genética , Peptídeos e Proteínas de Sinalização Intercelular/genética , Longevidade/genética , Biossíntese de Proteínas , Animais , Drosophila/genética , Insulina/genética , Insulina/metabolismo , Fator de Crescimento Insulin-Like I/genética , Fator de Crescimento Insulin-Like I/metabolismo , Camundongos , Estresse Oxidativo/genética , Transdução de Sinais/genética
17.
Cell Rep ; 15(3): 638-650, 2016 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-27068460

RESUMO

The quest to extend healthspan via pharmacological means is becoming increasingly urgent, both from a health and economic perspective. Here we show that lithium, a drug approved for human use, promotes longevity and healthspan. We demonstrate that lithium extends lifespan in female and male Drosophila, when administered throughout adulthood or only later in life. The life-extending mechanism involves the inhibition of glycogen synthase kinase-3 (GSK-3) and activation of the transcription factor nuclear factor erythroid 2-related factor (NRF-2). Combining genetic loss of the NRF-2 repressor Kelch-like ECH-associated protein 1 (Keap1) with lithium treatment revealed that high levels of NRF-2 activation conferred stress resistance, while low levels additionally promoted longevity. The discovery of GSK-3 as a therapeutic target for aging will likely lead to more effective treatments that can modulate mammalian aging and further improve health in later life.


Assuntos
Drosophila melanogaster/efeitos dos fármacos , Drosophila melanogaster/fisiologia , Quinase 3 da Glicogênio Sintase/metabolismo , Hormese/efeitos dos fármacos , Lítio/farmacologia , Longevidade/efeitos dos fármacos , Fator 2 Relacionado a NF-E2/metabolismo , Animais , Autofagia/efeitos dos fármacos , Restrição Calórica , Carboidratos da Dieta , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Feminino , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Metabolismo dos Lipídeos/efeitos dos fármacos , Masculino , Modelos Biológicos , Estresse Fisiológico/efeitos dos fármacos , Análise de Sobrevida , Transcrição Gênica/efeitos dos fármacos , Xenobióticos/farmacologia
18.
Front Aging Neurosci ; 6: 190, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25126078

RESUMO

The greatest risk factor for Alzheimer's disease (AD) is age, and changes in the ageing nervous system are likely contributors to AD pathology. Amyloid beta (Aß) accumulation, which occurs as a result of the amyloidogenic processing of amyloid precursor protein (APP), is thought to initiate the pathogenesis of AD, eventually leading to neuronal cell death. Previously, we developed an adult-onset Drosophila model of AD. Mutant Aß42 accumulation led to increased mortality and neuronal dysfunction in the adult flies. Furthermore, we showed that lithium reduced Aß42 protein, but not mRNA, and was able to rescue Aß42-induced toxicity. In the current study, we investigated the mechanism/s by which lithium modulates Aß42 protein levels and Aß42 induced toxicity in the fly model. We found that lithium caused a reduction in protein synthesis in Drosophila and hence the level of Aß42. At both the low and high doses tested, lithium rescued the locomotory defects induced by Aß42, but it rescued lifespan only at lower doses, suggesting that long-term, high-dose lithium treatment may have induced toxicity. Lithium also down-regulated translation in the fission yeast Schizosaccharomyces pombe associated with increased chronological lifespan. Our data highlight a role for lithium and reduced protein synthesis as potential therapeutic targets for AD pathogenesis.

19.
Fly (Austin) ; 3(4): 278-80, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19875944

RESUMO

Altering the cellular response to internal and external stressors is essential for survival, hence the process of translation is exquisitely regulated to rapidly change the proteomic profile upon physiological challenges. We recently reported that genetic and pharmacological manipulation of translation may be beneficial in the treatment of Parkinson disease (PD). Using two Drosophila models of PD, we showed that altering the regulation of protein translation is sufficient to ameliorate the phenotypes of these models, including neurodegeneration, mitochondrial defects and behavioral deficits. Previous studies implicating translation regulation in lifespan extension further implicates this as an important mechanism that can mediate cell protective pathways, not just for age-related diseases such as PD, but also of aging itself. As such, translation regulation represents a convergent target for therapeutic interventions. Here we highlight the therapeutic potential of translation regulation in disease and describe how determining profiles of protein synthesis may help in the fight for disease prevention and healthy aging.


Assuntos
Doença de Parkinson/terapia , Biossíntese de Proteínas , Animais , Modelos Animais de Doenças , Drosophila
20.
Nat Neurosci ; 12(9): 1129-35, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19684592

RESUMO

Mutations in PINK1 and PARK2 cause autosomal recessive parkinsonism, a neurodegenerative disorder that is characterized by the loss of dopaminergic neurons. To discover potential therapeutic pathways, we identified factors that genetically interact with Drosophila park and Pink1. We found that overexpression of the translation inhibitor Thor (4E-BP) can suppress all of the pathologic phenotypes, including degeneration of dopaminergic neurons in Drosophila. 4E-BP is activated in vivo by the TOR inhibitor rapamycin, which could potently suppress pathology in Pink1 and park mutants. Rapamycin also ameliorated mitochondrial defects in cells from individuals with PARK2 mutations. Recently, 4E-BP was shown to be inhibited by the most common cause of parkinsonism, dominant mutations in LRRK2. We also found that loss of the Drosophila LRRK2 homolog activated 4E-BP and was also able to suppress Pink1 and park pathology. Thus, in conjunction with recent findings, our results suggest that pharmacologic stimulation of 4E-BP activity may represent a viable therapeutic approach for multiple forms of parkinsonism.


Assuntos
Dopamina/metabolismo , Proteínas de Drosophila/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Neurônios/fisiologia , Fármacos Neuroprotetores/farmacologia , Fatores de Iniciação de Peptídeos/metabolismo , Sirolimo/farmacologia , Animais , Animais Geneticamente Modificados , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Drosophila , Proteínas de Drosophila/genética , Fibroblastos/efeitos dos fármacos , Fibroblastos/fisiologia , Glutationa Transferase/metabolismo , Humanos , Locomoção/efeitos dos fármacos , Locomoção/fisiologia , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/fisiologia , Músculos/efeitos dos fármacos , Músculos/ultraestrutura , Degeneração Neural/tratamento farmacológico , Degeneração Neural/fisiopatologia , Neurônios/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/genética , Ubiquitina-Proteína Ligases/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA