Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Cell ; 166(3): 784-784.e1, 2016 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-27471972

RESUMO

Aging is characterized by loss of homeostasis across multiple tissues. The nervous system governs whole-body homeostasis by communicating external and internal signals to peripheral tissues. Here, we highlight neuronal mechanisms and downstream outputs that regulate aging and longevity. Targeting these neuronal pathways may be a novel strategy to promote healthy aging. To view this SnapShot, open or download the PDF.


Assuntos
Envelhecimento/fisiologia , Fenômenos Fisiológicos do Sistema Nervoso , Humanos , Sistema Nervoso , Vias Neurais
2.
Cell ; 160(5): 842-855, 2015 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-25723162

RESUMO

Low energy states delay aging in multiple species, yet mechanisms coordinating energetics and longevity across tissues remain poorly defined. The conserved energy sensor AMP-activated protein kinase (AMPK) and its corresponding phosphatase calcineurin modulate longevity via the CREB regulated transcriptional coactivator (CRTC)-1 in C. elegans. We show that CRTC-1 specifically uncouples AMPK/calcineurin-mediated effects on lifespan from pleiotropic side effects by reprogramming mitochondrial and metabolic function. This pro-longevity metabolic state is regulated cell nonautonomously by CRTC-1 in the nervous system. Neuronal CRTC-1/CREB regulates peripheral metabolism antagonistically with the functional PPARα ortholog, NHR-49, drives mitochondrial fragmentation in distal tissues, and suppresses the effects of AMPK on systemic mitochondrial metabolism and longevity via a cell-nonautonomous catecholamine signal. These results demonstrate that while both local and distal mechanisms combine to modulate aging, distal regulation overrides local contribution. Targeting central perception of energetic state is therefore a potential strategy to promote healthy aging.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/fisiologia , Catecolaminas/metabolismo , Mitocôndrias/metabolismo , Neurônios/metabolismo , Transdução de Sinais , Transativadores/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Caenorhabditis elegans/citologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Longevidade , Receptores Citoplasmáticos e Nucleares/metabolismo
3.
Cell ; 160(1-2): 132-44, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25542313

RESUMO

Dietary restriction (DR) without malnutrition encompasses numerous regimens with overlapping benefits including longevity and stress resistance, but unifying nutritional and molecular mechanisms remain elusive. In a mouse model of DR-mediated stress resistance, we found that sulfur amino acid (SAA) restriction increased expression of the transsulfuration pathway (TSP) enzyme cystathionine γ-lyase (CGL), resulting in increased hydrogen sulfide (H2S) production and protection from hepatic ischemia reperfusion injury. SAA supplementation, mTORC1 activation, or chemical/genetic CGL inhibition reduced H2S production and blocked DR-mediated stress resistance. In vitro, the mitochondrial protein SQR was required for H2S-mediated protection during nutrient/oxygen deprivation. Finally, TSP-dependent H2S production was observed in yeast, worm, fruit fly, and rodent models of DR-mediated longevity. Together, these data are consistent with evolutionary conservation of TSP-mediated H2S as a mediator of DR benefits with broad implications for clinical translation. PAPERFLICK:


Assuntos
Dieta , Sulfeto de Hidrogênio/metabolismo , Animais , Evolução Biológica , Caenorhabditis elegans/fisiologia , Restrição Calórica , Cistationina gama-Liase/metabolismo , Cisteína/metabolismo , Drosophila melanogaster/fisiologia , Feminino , Rim/irrigação sanguínea , Rim/lesões , Expectativa de Vida , Fígado/irrigação sanguínea , Fígado/lesões , Masculino , Metionina/metabolismo , Camundongos Knockout , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Traumatismo por Reperfusão , Transdução de Sinais , Estresse Fisiológico , Transcriptoma , Leveduras/fisiologia
4.
PLoS Genet ; 19(9): e1010938, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37721956

RESUMO

mTORC1 (mechanistic target of rapamycin complex 1) is a metabolic sensor that promotes growth when nutrients are abundant. Ubiquitous inhibition of mTORC1 extends lifespan in multiple organisms but also disrupts several anabolic processes resulting in stunted growth, slowed development, reduced fertility, and disrupted metabolism. However, it is unclear if these pleiotropic effects of mTORC1 inhibition can be uncoupled from longevity. Here, we utilize the auxin-inducible degradation (AID) system to restrict mTORC1 inhibition to C. elegans neurons. We find that neuron-specific degradation of RAGA-1, an upstream activator of mTORC1, or LET-363, the ortholog of mammalian mTOR, is sufficient to extend lifespan in C. elegans. Unlike raga-1 loss of function genetic mutations or somatic AID of RAGA-1, neuronal AID of RAGA-1 robustly extends lifespan without impairing body size, developmental rate, brood size, or neuronal function. Moreover, while degradation of RAGA-1 in all somatic tissues alters the expression of thousands of genes, demonstrating the widespread effects of mTORC1 inhibition, degradation of RAGA-1 in neurons only results in around 200 differentially expressed genes with a specific enrichment in metabolism and stress response. Notably, our work demonstrates that targeting mTORC1 specifically in the nervous system in C. elegans uncouples longevity from growth and reproductive impairments, and that many canonical effects of low mTORC1 activity are not required to promote healthy aging. These data challenge previously held ideas about the mechanisms of mTORC1 lifespan extension and underscore the potential of promoting longevity by neuron-specific mTORC1 modulation.


Assuntos
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Animais , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Caenorhabditis elegans/metabolismo , Longevidade/genética , Complexos Multiproteicos/genética , Reprodução/genética , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Mamíferos/metabolismo
5.
Nature ; 544(7649): 185-190, 2017 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-28379943

RESUMO

Chromatin and metabolic states both influence lifespan, but how they interact in lifespan regulation is largely unknown. The COMPASS chromatin complex, which trimethylates lysine 4 on histone H3 (H3K4me3), regulates lifespan in Caenorhabditis elegans. However, the mechanism by which H3K4me3 modifiers affect longevity, and whether this mechanism involves metabolic changes, remain unclear. Here we show that a deficiency in H3K4me3 methyltransferase, which extends lifespan, promotes fat accumulation in worms with a specific enrichment of mono-unsaturated fatty acids (MUFAs). This fat metabolism switch in H3K4me3 methyltransferase-deficient worms is mediated at least in part by the downregulation of germline targets, including S6 kinase, and by the activation of an intestinal transcriptional network that upregulates delta-9 fatty acid desaturases. Notably, the accumulation of MUFAs is necessary for the lifespan extension of H3K4me3 methyltransferase-deficient worms, and dietary MUFAs are sufficient to extend lifespan. Given the conservation of lipid metabolism, dietary or endogenous MUFAs could extend lifespan and healthspan in other species, including mammals.


Assuntos
Caenorhabditis elegans/fisiologia , Gorduras na Dieta/farmacologia , Ácidos Graxos Insaturados/metabolismo , Histonas/metabolismo , Longevidade/efeitos dos fármacos , Longevidade/fisiologia , Lisina/metabolismo , Envelhecimento/efeitos dos fármacos , Envelhecimento/metabolismo , Animais , Caenorhabditis elegans/efeitos dos fármacos , Caenorhabditis elegans/enzimologia , Caenorhabditis elegans/genética , Gorduras na Dieta/administração & dosagem , Gorduras na Dieta/metabolismo , Regulação para Baixo , Ácidos Graxos Dessaturases/genética , Ácidos Graxos Dessaturases/metabolismo , Ácidos Graxos Insaturados/administração & dosagem , Ácidos Graxos Insaturados/farmacologia , Regulação Enzimológica da Expressão Gênica , Células Germinativas/enzimologia , Células Germinativas/metabolismo , Histona-Lisina N-Metiltransferase/deficiência , Histona-Lisina N-Metiltransferase/metabolismo , Histonas/química , Mucosa Intestinal/metabolismo , Intestinos/enzimologia , Metabolismo dos Lipídeos/efeitos dos fármacos , Metilação , Proteínas Quinases S6 Ribossômicas 70-kDa/deficiência , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Estearoil-CoA Dessaturase , Regulação para Cima
7.
Nature ; 541(7635): 102-106, 2017 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-27919065

RESUMO

Ageing is driven by a loss of transcriptional and protein homeostasis and is the key risk factor for multiple chronic diseases. Interventions that attenuate or reverse systemic dysfunction associated with age therefore have the potential to reduce overall disease risk in the elderly. Precursor mRNA (pre-mRNA) splicing is a fundamental link between gene expression and the proteome, and deregulation of the splicing machinery is linked to several age-related chronic illnesses. However, the role of splicing homeostasis in healthy ageing remains unclear. Here we demonstrate that pre-mRNA splicing homeostasis is a biomarker and predictor of life expectancy in Caenorhabditis elegans. Using transcriptomics and in-depth splicing analysis in young and old animals fed ad libitum or subjected to dietary restriction, we find defects in global pre-mRNA splicing with age that are reduced by dietary restriction via splicing factor 1 (SFA-1; the C. elegans homologue of SF1, also known as branchpoint binding protein, BBP). We show that SFA-1 is specifically required for lifespan extension by dietary restriction and by modulation of the TORC1 pathway components AMPK, RAGA-1 and RSKS-1/S6 kinase. We also demonstrate that overexpression of SFA-1 is sufficient to extend lifespan. Together, these data demonstrate a role for RNA splicing homeostasis in dietary restriction longevity and suggest that modulation of specific spliceosome components may prolong healthy ageing.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Restrição Calórica , Longevidade/genética , Longevidade/fisiologia , Complexos Multiproteicos/metabolismo , Fatores de Processamento de RNA/metabolismo , Splicing de RNA , Serina-Treonina Quinases TOR/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Envelhecimento/genética , Animais , Proteínas de Caenorhabditis elegans/genética , Genoma/genética , Homeostase , Alvo Mecanístico do Complexo 1 de Rapamicina , Precursores de RNA/genética , Precursores de RNA/metabolismo , Fatores de Processamento de RNA/genética , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Transcriptoma
8.
EMBO Rep ; 20(12): e48395, 2019 12 05.
Artigo em Inglês | MEDLINE | ID: mdl-31667999

RESUMO

Mitochondria are organized in the cell in the form of a dynamic, interconnected network. Mitochondrial dynamics, regulated by mitochondrial fission, fusion, and trafficking, ensure restructuring of this complex reticulum in response to nutrient availability, molecular signals, and cellular stress. Aberrant mitochondrial structures have long been observed in aging and age-related diseases indicating that mitochondrial dynamics are compromised as cells age. However, the specific mechanisms by which aging affects mitochondrial dynamics and whether these changes are causally or casually associated with cellular and organismal aging is not clear. Here, we review recent studies that show specifically how mitochondrial fission, fusion, and trafficking are altered with age. We discuss factors that change with age to directly or indirectly influence mitochondrial dynamics while examining causal roles for altered mitochondrial dynamics in healthy aging and underlying functional outputs that might affect longevity. Lastly, we propose that altered mitochondrial dynamics might not just be a passive consequence of aging but might constitute an adaptive mechanism to mitigate age-dependent cellular impairments and might be targeted to increase longevity and promote healthy aging.


Assuntos
Envelhecimento Saudável/fisiologia , Longevidade/fisiologia , Dinâmica Mitocondrial/fisiologia , Proteínas Quinases Ativadas por AMP/fisiologia , Envelhecimento/fisiologia , Animais , Senescência Celular/fisiologia , Interações entre Hospedeiro e Microrganismos/fisiologia , Humanos , Insulina/fisiologia , Microbiota/fisiologia , Modelos Biológicos , Organelas/fisiologia , Transdução de Sinais , Sirtuínas/fisiologia , Somatomedinas/fisiologia , Serina-Treonina Quinases TOR/fisiologia
9.
Trends Genet ; 33(5): 303-321, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28365140

RESUMO

Advances in public health in the past century have seen a sharp increase in human life expectancy. With these changes have come an increased prevalence of age-related pathologies and health burdens in the elderly. Patient age is the biggest risk factor for multiple chronic conditions that often occur simultaneously within a single individual. An alternative to disease-centric therapeutic approaches is that of 'geroscience', which aims to define molecular mechanisms that link age to overall disease risk. One such mechanism is deregulation of CREB-regulated transcriptional coactivators (CRTCs). Initially identified for their role in modulating CREB transcription, the past 5 years has seen an expansion in knowledge of new cellular regulators and roles of CRTCs beyond CREB. CRTCs have been shown to modulate organismal aging in Caenorhabditis elegans and to impact on age-related diseases in humans. We discuss CRTC deregulation as a new driver of aging that integrates the link between age and disease risk.


Assuntos
Envelhecimento/genética , Transativadores/genética , Transcrição Gênica , Envelhecimento/patologia , Animais , Modelos Animais de Doenças , Humanos , Fosforilação , Fatores de Risco
10.
Hum Genet ; 139(3): 357-369, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31834493

RESUMO

Alternative pre-mRNA splicing increases the complexity of the proteome that can be generated from the available genomic coding sequences. Dysregulation of the splicing process has been implicated in a vast repertoire of diseases. However, splicing has recently been linked to both the aging process itself and pro-longevity interventions. This review focuses on recent research towards defining RNA splicing as a new hallmark of aging. We highlight dysfunctional alternative splicing events that contribute to the aging phenotype across multiple species, along with recent efforts toward deciphering mechanistic roles for RNA splicing in the regulation of aging and longevity. Further, we discuss recent research demonstrating a direct requirement for specific splicing factors in pro-longevity interventions, and specifically how nutrient signaling pathways interface to splicing factor regulation and downstream splicing targets. Finally, we review the emerging potential of using splicing profiles as a predictor of biological age and life expectancy. Understanding the role of RNA splicing components and downstream targets altered in aging may provide opportunities to develop therapeutics and ultimately extend healthy lifespan in humans.


Assuntos
Envelhecimento/genética , Processamento Alternativo/genética , Longevidade/genética , Animais , Humanos , Fenótipo , Fatores de Processamento de RNA/genética
12.
Proc Natl Acad Sci U S A ; 111(42): E4458-67, 2014 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-25288734

RESUMO

Mild inhibition of mitochondrial respiration extends the lifespan of many species. In Caenorhabditis elegans, reactive oxygen species (ROS) promote longevity by activating hypoxia-inducible factor 1 (HIF-1) in response to reduced mitochondrial respiration. However, the physiological role and mechanism of ROS-induced longevity are poorly understood. Here, we show that a modest increase in ROS increases the immunity and lifespan of C. elegans through feedback regulation by HIF-1 and AMP-activated protein kinase (AMPK). We found that activation of AMPK as well as HIF-1 mediates the longevity response to ROS. We further showed that AMPK reduces internal levels of ROS, whereas HIF-1 amplifies the levels of internal ROS under conditions that increase ROS. Moreover, mitochondrial ROS increase resistance to various pathogenic bacteria, suggesting a possible association between immunity and long lifespan. Thus, AMPK and HIF-1 may control immunity and longevity tightly by acting as feedback regulators of ROS.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/fisiologia , Retroalimentação Fisiológica , Espécies Reativas de Oxigênio/metabolismo , Fatores de Transcrição/metabolismo , Envelhecimento , Animais , Caenorhabditis elegans/imunologia , Respiração Celular , Homeostase , Ferro/química , Longevidade/fisiologia , Mitocôndrias/metabolismo , Mutação , Paraquat/química , Fosforilação
14.
Nat Commun ; 15(1): 3070, 2024 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-38594249

RESUMO

Cellular response to redox imbalance is crucial for organismal health. microRNAs are implicated in stress responses. ALG-1, the C. elegans ortholog of human AGO2, plays an essential role in microRNA processing and function. Here we investigated the mechanisms governing ALG-1 expression in C. elegans and the players controlling lifespan and stress resistance downstream of ALG-1. We show that upregulation of ALG-1 is a shared feature in conditions linked to increased longevity (e.g., germline-deficient glp-1 mutants). ALG-1 knockdown reduces lifespan and oxidative stress resistance, while overexpression enhances survival against pro-oxidant agents but not heat or reductive stress. R02D3.7 represses alg-1 expression, impacting oxidative stress resistance at least in part via ALG-1. microRNAs upregulated in glp-1 mutants (miR-87-3p, miR-230-3p, and miR-235-3p) can target genes in the protein disulfide isomerase pathway and protect against oxidative stress. This study unveils a tightly regulated network involving transcription factors and microRNAs which controls organisms' ability to withstand oxidative stress.


Assuntos
Proteínas de Caenorhabditis elegans , MicroRNAs , Animais , Humanos , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Estresse Oxidativo/genética , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo
15.
Nat Commun ; 14(1): 7564, 2023 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-37985670

RESUMO

Even slight imbalance between the growth rate of different organs can accumulate to a large deviation from their appropriate size during development. Here, we use live imaging of the pharynx of C. elegans to ask if and how organ size scaling nevertheless remains uniform among individuals. Growth trajectories of hundreds of individuals reveal that pharynxes grow by a near constant volume per larval stage that is independent of their initial size, such that undersized pharynxes catch-up in size during development. Tissue-specific depletion of RAGA-1, an activator of mTOR and growth, shows that maintaining correct pharynx-to-body size proportions involves a bi-directional coupling between pharynx size and body growth. In simulations, this coupling cannot be explained by limitation of food uptake alone, and genetic experiments reveal an involvement of the mechanotransducing transcriptional co-regulator yap-1. Our data suggests that mechanotransduction coordinates pharynx growth with other tissues, ensuring body plan uniformity among individuals.


Assuntos
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Humanos , Animais , Caenorhabditis elegans/genética , Faringe/metabolismo , Mecanotransdução Celular , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Sinalização YAP
16.
Cell Rep Methods ; 3(3): 100433, 2023 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-37056370

RESUMO

Here, we introduce a single-copy knockin translating ribosome immunoprecipitation (SKI TRIP) toolkit, a collection of Caenorhabditis elegans strains engineered by CRISPR in which tissue-specific expression of FLAG-tagged ribosomal subunit protein RPL-22 is driven by cassettes present in single copy from defined sites in the genome. Through in-depth characterization of the effects of the FLAG tag in animals in which endogenous RPL-22 has been tagged, we show that it incorporates into actively translating ribosomes and efficiently and cleanly pulls down cell-type-specific transcripts. Importantly, the presence of the tag does not impact overall mRNA translation, create bias in transcript use, or cause changes to fitness of the animal. We propose SKI TRIP use for the study of tissue-specific differences in translation and for investigating processes that are acutely sensitive to changes in translation like development or aging.


Assuntos
Caenorhabditis elegans , Biossíntese de Proteínas , Animais , Caenorhabditis elegans/genética , RNA Mensageiro/genética , Biossíntese de Proteínas/genética , Ribossomos/genética , Proteínas Ribossômicas/genética , Imunoprecipitação
17.
Nat Aging ; 3(11): 1358-1371, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37946042

RESUMO

Loss of function during aging is accompanied by transcriptional drift, altering gene expression and contributing to a variety of age-related diseases. CREB-regulated transcriptional coactivators (CRTCs) have emerged as key regulators of gene expression that might be targeted to promote longevity. Here we define the role of the Caenorhabditis elegans CRTC-1 in the epigenetic regulation of longevity. Endogenous CRTC-1 binds chromatin factors, including components of the COMPASS complex, which trimethylates lysine 4 on histone H3 (H3K4me3). CRISPR editing of endogenous CRTC-1 reveals that the CREB-binding domain in neurons is specifically required for H3K4me3-dependent longevity. However, this effect is independent of CREB but instead acts via the transcription factor AP-1. Strikingly, CRTC-1 also mediates global histone acetylation levels, and this acetylation is essential for H3K4me3-dependent longevity. Indeed, overexpression of an acetyltransferase enzyme is sufficient to promote longevity in wild-type worms. CRTCs, therefore, link energetics to longevity by critically fine-tuning histone acetylation and methylation to promote healthy aging.


Assuntos
Caenorhabditis elegans , Longevidade , Animais , Caenorhabditis elegans/genética , Epigênese Genética , Histonas/química , Longevidade/genética , Fatores de Transcrição/genética
18.
Nat Cell Biol ; 25(5): 672-684, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37127715

RESUMO

Dietary mono-unsaturated fatty acids (MUFAs) are linked to longevity in several species. But the mechanisms by which MUFAs extend lifespan remain unclear. Here we show that an organelle network involving lipid droplets and peroxisomes is critical for MUFA-induced longevity in Caenorhabditis elegans. MUFAs upregulate the number of lipid droplets in fat storage tissues. Increased lipid droplet number is necessary for MUFA-induced longevity and predicts remaining lifespan. Lipidomics datasets reveal that MUFAs also modify the ratio of membrane lipids and ether lipids-a signature associated with decreased lipid oxidation. In agreement with this, MUFAs decrease lipid oxidation in middle-aged individuals. Intriguingly, MUFAs upregulate not only lipid droplet number but also peroxisome number. A targeted screen identifies genes involved in the co-regulation of lipid droplets and peroxisomes, and reveals that induction of both organelles is optimal for longevity. Our study uncovers an organelle network involved in lipid homeostasis and lifespan regulation, opening new avenues for interventions to delay aging.


Assuntos
Longevidade , Peroxissomos , Humanos , Pessoa de Meia-Idade , Animais , Longevidade/genética , Gotículas Lipídicas , Ácidos Graxos Insaturados , Caenorhabditis elegans/genética , Ácidos Graxos
19.
Nat Cell Biol ; 24(6): 906-916, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35681008

RESUMO

Lysosomes are key cellular organelles that metabolize extra- and intracellular substrates. Alterations in lysosomal metabolism are implicated in ageing-associated metabolic and neurodegenerative diseases. However, how lysosomal metabolism actively coordinates the metabolic and nervous systems to regulate ageing remains unclear. Here we report a fat-to-neuron lipid signalling pathway induced by lysosomal metabolism and its longevity-promoting role in Caenorhabditis elegans. We discovered that induced lysosomal lipolysis in peripheral fat storage tissue upregulates the neuropeptide signalling pathway in the nervous system to promote longevity. This cell-non-autonomous regulation is mediated by a specific polyunsaturated fatty acid, dihomo-γ-linolenic acid, and LBP-3 lipid chaperone protein transported from the fat storage tissue to neurons. LBP-3 binds to dihomo-γ-linolenic acid, and acts through NHR-49 nuclear receptor and NLP-11 neuropeptide in neurons to extend lifespan. These results reveal lysosomes as a signalling hub to coordinate metabolism and ageing, and lysosomal signalling mediated inter-tissue communication in promoting longevity.


Assuntos
Proteínas de Caenorhabditis elegans , Neuropeptídeos , Ácido 8,11,14-Eicosatrienoico/metabolismo , Animais , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Longevidade/genética , Lisossomos/metabolismo , Neurônios/metabolismo , Neuropeptídeos/metabolismo
20.
Nat Commun ; 13(1): 967, 2022 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-35181679

RESUMO

Inhibition of the master growth regulator mTORC1 (mechanistic target of rapamycin complex 1) slows ageing across phyla, in part by reducing protein synthesis. Various stresses globally suppress protein synthesis through the integrated stress response (ISR), resulting in preferential translation of the transcription factor ATF-4. Here we show in C. elegans that inhibition of translation or mTORC1 increases ATF-4 expression, and that ATF-4 mediates longevity under these conditions independently of ISR signalling. ATF-4 promotes longevity by activating canonical anti-ageing mechanisms, but also by elevating expression of the transsulfuration enzyme CTH-2 to increase hydrogen sulfide (H2S) production. This H2S boost increases protein persulfidation, a protective modification of redox-reactive cysteines. The ATF-4/CTH-2/H2S pathway also mediates longevity and increased stress resistance from mTORC1 suppression. Increasing H2S levels, or enhancing mechanisms that H2S influences through persulfidation, may represent promising strategies for mobilising therapeutic benefits of the ISR, translation suppression, or mTORC1 inhibition.


Assuntos
Fator 4 Ativador da Transcrição/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Sulfeto de Hidrogênio/metabolismo , Longevidade/genética , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Fator 4 Ativador da Transcrição/genética , Animais , Animais Geneticamente Modificados , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/genética , Transdução de Sinais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA