Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters











Publication year range
1.
Clin Nutr ; 43(10): 2236-2248, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39182436

ABSTRACT

BACKGROUND & AIMS: Unhealthy lifestyles, such as chronic consumption of a Western Diet (WD), have been associated with increased systemic inflammation and oxidative stress (OS), a condition that may favour cognitive dysfunctions during aging. Polyphenols, such as rosmarinic acid (RA) may buffer low-grade inflammation and OS, characterizing the aging brain that is sustained by WD, promoting healthspan. The aim of this study was to evaluate the ability of RA to prevent cognitive decline in a mouse model of WD-driven unhealthy aging and to gain knowledge on the specific molecular pathways modulated within the brain. METHODS: Aged male and female C57Bl/6N mice were supplemented either with RA or vehicle for 6 weeks. Following 2 weeks on RA they started being administered either with WD or control diet (CD). Successively all mice were tested for cognitive abilities in the Morris water maze (MWM) and emotionality in the elevated plus maze (EPM). Glucose and lipid homeostasis were assessed in trunk blood while the hippocampus was dissected out for RNAseq transcriptomic analysis. RESULTS: RA prevented insulin resistance in males while protecting both males and females from WD-dependent memory impairment. In the hippocampus, RA modulated OS pathways in males and immune- and sex hormones-related signalling cascades (Lhb and Lhcgr genes) in females. Moreover, RA overall resulted in an upregulation of Glp1r, recently identified as a promising target to prevent metabolic derangements. In addition, we also found an RA-dependent enrichment in nuclear transcription factors, such as NF-κB, GR and STAT3, that have been recently suggested to promote healthspan and longevity by modulating inflammatory and cell survival pathways. CONCLUSIONS: Oral RA supplementation may promote brain and metabolic plasticity during aging through antioxidant and immune-modulating properties possibly affecting the post-reproductive hormonal milieu in a sex-dependent fashion. Thus, its supplementation should be considered in the context of precision medicine as a possible strategy to preserve cognitive functions and to counteract metabolic derangements.


Subject(s)
Aging , Cinnamates , Depsides , Diet, Western , Hippocampus , Mice, Inbred C57BL , Rosmarinic Acid , Animals , Depsides/pharmacology , Male , Female , Cinnamates/pharmacology , Mice , Diet, Western/adverse effects , Aging/drug effects , Hippocampus/metabolism , Hippocampus/drug effects , Sex Factors , Cognitive Dysfunction/prevention & control , Oxidative Stress/drug effects , Cognition/drug effects , Insulin Resistance , Disease Models, Animal , Maze Learning/drug effects , Antioxidants/pharmacology
2.
Nat Commun ; 14(1): 8142, 2023 Dec 08.
Article in English | MEDLINE | ID: mdl-38065964

ABSTRACT

To ameliorate or even prevent signatures of aging in ultimately humans, we here report the identification of a previously undescribed polyacetylene contained in the root of carrots (Daucus carota), hereafter named isofalcarintriol, which we reveal as potent promoter of longevity in the nematode C. elegans. We assign the absolute configuration of the compound as (3 S,8 R,9 R,E)-heptadeca-10-en-4,6-diyne-3,8,9-triol, and develop a modular asymmetric synthesis route for all E-isofalcarintriol stereoisomers. At the molecular level, isofalcarintriol affects cellular respiration in mammalian cells, C. elegans, and mice, and interacts with the α-subunit of the mitochondrial ATP synthase to promote mitochondrial biogenesis. Phenotypically, this also results in decreased mammalian cancer cell growth, as well as improved motility and stress resistance in C. elegans, paralleled by reduced protein accumulation in nematodal models of neurodegeneration. In addition, isofalcarintriol supplementation to both wild-type C57BL/6NRj mice on high-fat diet, and aged mice on chow diet results in improved glucose metabolism, increased exercise endurance, and attenuated parameters of frailty at an advanced age. Given these diverse effects on health parameters in both nematodes and mice, isofalcarintriol might become a promising mitohormesis-inducing compound to delay, ameliorate, or prevent aging-associated diseases in humans.


Subject(s)
Caenorhabditis elegans , Daucus carota , Humans , Animals , Mice , Caenorhabditis elegans/metabolism , Mitochondria/metabolism , Mice, Inbred C57BL , Aging , Longevity , Polyynes/metabolism , Mammals
3.
Nutrients ; 15(15)2023 Jul 28.
Article in English | MEDLINE | ID: mdl-37571303

ABSTRACT

A growing body of evidence suggests that regular consumption of natural products might promote healthy aging; however, their mechanisms of action are still unclear. Rosmarinic acid (RA) is a polyphenol holding anti-inflammatory, antioxidant and neuroprotective properties. The aim of this study was to characterise the efficacy of an oral administration of RA in promoting healthspan in a mouse model of physiological aging. Aged C57Bl/6 male and female (24-month-old) mice were either administered with RA (500 mg/Kg) or a vehicle in drinking bottles for 52 days while 3-month-old mice receiving the same treatment were used as controls. All subjects were assessed for cognitive abilities in the Morris water maze (MWM) and for emotionality in the elevated-plus maze test (EPM). Brain-derived Neurotrophic Factor (BDNF) protein levels were evaluated in the hippocampus. Since the interaction between metabolic signals and cerebral functions plays a pivotal role in the etiopathogenesis of cognitive decline, the glycaemic and lipid profiles of the mice were also assessed. RA enhanced learning and memory in 24-month-old mice, an effect that was associated to improved glucose homeostasis. By contrast, the lipid profile was disrupted in young adults. This effect was associated with worse glycaemic control in males and with reduced BDNF levels in females, suggesting powerful sex-dependent effects and raising a note of caution for RA administration in young healthy adult subjects.


Subject(s)
Brain-Derived Neurotrophic Factor , Oxidative Stress , Male , Mice , Female , Animals , Brain-Derived Neurotrophic Factor/metabolism , Cognition , Hippocampus/metabolism , Mice, Inbred Strains , Glucose/metabolism , Lipids , Mice, Inbred C57BL , Rosmarinic Acid
4.
Sci Rep ; 12(1): 21050, 2022 12 06.
Article in English | MEDLINE | ID: mdl-36473898

ABSTRACT

Due to intact reactive oxygen species homeostasis and glucose metabolism, C57BL/6NRj mice are especially suitable to study cellular alterations in metabolism. We applied Nuclear Magnetic resonance spectroscopy to analyze five different tissues of this mouse strain during aging and included female and male mice aged 3, 6, 12, and 24 months. Metabolite signatures allowed separation between the age groups in all tissues, and we identified the most prominently changing metabolites in female and male tissues. A refined analysis of individual metabolite levels during aging revealed an early onset of age-related changes at 6 months, sex-specific differences in the liver, and a biphasic pattern for various metabolites in the brain, heart, liver, and lung. In contrast, a linear decrease of amino acids was apparent in muscle tissues. Based on these results, we assume that age-related metabolic alterations happen at a comparably early aging state and are potentially associated with a metabolic switch. Moreover, identified differences between female and male tissues stress the importance of distinguishing between sexes when studying age-related changes and developing new treatment approaches. Besides, metabolomic features seem to be highly dependent on the genetic background of mouse strains.


Subject(s)
Aging , Mice, Inbred C57BL , Animals , Female , Male , Mice , Mice, Inbred C57BL/metabolism
5.
J Biol Chem ; 298(7): 102085, 2022 07.
Article in English | MEDLINE | ID: mdl-35636511

ABSTRACT

Inhibition of gene expression in Caenorhabditis elegans, a versatile model organism for studying the genetics of development and aging, is achievable by feeding nematodes with bacteria expressing specific dsRNAs. Overexpression of hypoxia-inducible factor 1 (hif-1) or heat-shock factor 1 (hsf-1) by conventional transgenesis has previously been shown to promote nematodal longevity. However, it is unclear whether other methods of gene overexpression are feasible, particularly with the advent of CRISPR-based techniques. Here, we show that feeding C. elegans engineered to stably express a Cas9-derived synthetic transcription factor with bacteria expressing promoter-specific single guide RNAs (sgRNAs) also allows activation of gene expression. We demonstrate that CRISPR activation via ingested sgRNAs specific for the respective promoter regions of hif-1 or hsf-1 increases gene expression and extends lifespan of C. elegans. Furthermore, and as an in silico resource for future studies aiming to use CRISPR activation in C. elegans, we provide predicted promoter-specific sgRNA target sequences for >13,000 C. elegans genes with experimentally defined transcription start sites. We anticipate that the approach and components described herein will help to facilitate genome-wide gene overexpression studies, for example, to identify modulators of aging or other phenotypes of interest, by enabling induction of transcription by feeding of sgRNA-expressing bacteria to nematodes.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Animals , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , Clustered Regularly Interspaced Short Palindromic Repeats , Eating , Longevity/genetics , RNA, Small Untranslated , CRISPR-Cas Systems
6.
Nat Commun ; 13(1): 107, 2022 01 10.
Article in English | MEDLINE | ID: mdl-35013237

ABSTRACT

Aging is impacted by interventions across species, often converging on metabolic pathways. Transcription factors regulate longevity yet approaches for their pharmacological modulation to exert geroprotection remain sparse. We show that increased expression of the transcription factor Grainyhead 1 (GRH-1) promotes lifespan and pathogen resistance in Caenorhabditis elegans. A compound screen identifies FDA-approved drugs able to activate human GRHL1 and promote nematodal GRH-1-dependent longevity. GRHL1 activity is regulated by post-translational lysine methylation and the phosphoinositide (PI) 3-kinase C2A. Consistently, nematodal longevity following impairment of the PI 3-kinase or insulin/IGF-1 receptor requires grh-1. In BXD mice, Grhl1 expression is positively correlated with lifespan and insulin sensitivity. In humans, GRHL1 expression positively correlates with insulin receptor signaling and also with lifespan. Fasting blood glucose levels, including in individuals with type 2 diabetes, are negatively correlated with GRHL1 expression. Thereby, GRH-1/GRHL1 is identified as a pharmacologically malleable transcription factor impacting insulin signaling and lifespan.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Class II Phosphatidylinositol 3-Kinases/genetics , Diabetes Mellitus, Type 2/genetics , Insulin-Like Growth Factor I/genetics , Insulin/metabolism , Longevity/genetics , Repressor Proteins/genetics , Transcription Factors/genetics , Animals , Animals, Genetically Modified , Blood Glucose/metabolism , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/genetics , Caenorhabditis elegans/growth & development , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , Class II Phosphatidylinositol 3-Kinases/metabolism , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Gene Expression Regulation , Humans , Insulin Resistance , Insulin-Like Growth Factor I/metabolism , Longevity/drug effects , Methylation , Mice , Papaverine/pharmacology , Repressor Proteins/metabolism , Signal Transduction , Transcription Factors/metabolism , Vorinostat/pharmacology
7.
Aging (Albany NY) ; 13(19): 22629-22648, 2021 10 04.
Article in English | MEDLINE | ID: mdl-34607977

ABSTRACT

Green tea catechins are associated with a delay in aging. We have designed the current study to investigate the impact and to unveil the target of the most abundant green tea catechins, epigallocatechin gallate (EGCG) and epicatechin gallate (ECG). Experiments were performed in Caenorhabditis elegans to analyze cellular metabolism, ROS homeostasis, stress resistance, physical exercise capacity, health- and lifespan, and the underlying signaling pathways. Besides, we examined the impact of EGCG and ECG in isolated murine mitochondria. A concentration of 2.5 µM EGCG and ECG enhanced health- and lifespan as well as stress resistance in C. elegans. Catechins hampered mitochondrial respiration in C. elegans after 6-12 h and the activity of complex I in isolated rodent mitochondria. The impaired mitochondrial respiration was accompanied by a transient drop in ATP production and a temporary increase in ROS levels in C. elegans. After 24 h, mitochondrial respiration and ATP levels got restored, and ROS levels even dropped below control conditions. The lifespan increases induced by EGCG and ECG were dependent on AAK-2/AMPK and SIR-2.1/SIRT1, as well as on PMK-1/p38 MAPK, SKN-1/NRF2, and DAF-16/FOXO. Long-term effects included significantly diminished fat content and enhanced SOD and CAT activities, required for the positive impact of catechins on lifespan. In summary, complex I inhibition by EGCG and ECG induced a transient drop in cellular ATP levels and a temporary ROS burst, resulting in SKN-1 and DAF-16 activation. Through adaptative responses, catechins reduced fat content, enhanced ROS defense, and improved healthspan in the long term.


Subject(s)
Catechin/analogs & derivatives , Electron Transport Complex I/antagonists & inhibitors , Longevity/drug effects , Tea/chemistry , Animals , Caenorhabditis elegans , Catechin/chemistry , Catechin/pharmacology , Electron Transport Complex I/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Stress, Physiological/drug effects
8.
Nat Metab ; 3(5): 588-589, 2021 May.
Article in English | MEDLINE | ID: mdl-34031593
9.
10.
Nat Commun ; 11(1): 2080, 2020 04 29.
Article in English | MEDLINE | ID: mdl-32350271

ABSTRACT

Excessive insulin signaling through the insulin receptor (IR) may play a role in the pathogenesis of diet-induced metabolic disease, including obesity and type 2 diabetes. Here we investigate whether heterozygous impairment of insulin receptor (IR) expression limited to peripheral, i.e. non-CNS, tissues of adult mice impacts the development of high-fat diet-induced metabolic deterioration. While exhibiting some features of insulin resistance, PerIRKO+/- mice display a hepatic energy deficit accompanied by induction of energy-sensing AMPK, mitochondrial biogenesis, PPARα, unexpectedly leading to protection from, and reversal of hepatic lipid accumulation (steatosis hepatis, NAFLD). Consistently, and unlike in control mice, the PPARα activator fenofibrate fails to further affect hepatic lipid accumulation in PerIRKO+/- mice. Taken together, and opposing previously established diabetogenic features of insulin resistance, incomplete impairment of insulin signaling may mimic central aspects of calorie restriction to limit hepatic lipid accumulation during conditions of metabolic stress.


Subject(s)
Diet, High-Fat/adverse effects , Fasting/metabolism , Fatty Liver/etiology , Fatty Liver/prevention & control , Receptor, Insulin/metabolism , Animals , Body Composition , Energy Metabolism , Feeding Behavior , Glucose/metabolism , Homeostasis , Insulin Resistance , Liver/metabolism , Liver/pathology , Male , Mice, Inbred C57BL , Mice, Knockout
11.
Redox Biol ; 32: 101448, 2020 05.
Article in English | MEDLINE | ID: mdl-32203922

ABSTRACT

Physiological aging is a complex process, influenced by a plethora of genetic and environmental factors. While being far from fully understood, a number of common aging hallmarks have been elucidated in recent years. Among these, transcriptomic alterations are hypothesized to represent a crucial early manifestation of aging. Accordingly, several transcription factors (TFs) have previously been identified as important modulators of lifespan in evolutionarily distant model organisms. Based on a set of TFs conserved between nematodes, zebrafish, mice, and humans, we here perform a RNA interference (RNAi) screen in C. elegans to discover evolutionarily conserved TFs impacting aging. We identify a basic helix-loop-helix TF, named HLH-2 in nematodes (Tcf3/E2A in mammals), to exert a pronounced lifespan-extending effect in C. elegans upon impairment. We further show that its impairment impacts cellular energy metabolism, increases parameters of healthy aging, and extends nematodal lifespan in a ROS-dependent manner. We then identify arginine kinases, orthologues of mammalian creatine kinases, as a target of HLH-2 transcriptional regulation, serving to mediate the healthspan-promoting effects observed upon impairment of hlh-2 expression. Consistently, HLH-2 is shown to epistatically interact with core components of known lifespan-regulating pathways, i.e. AAK-2/AMPK and LET-363/mTOR, as well as the aging-related TFs SKN-1/Nrf2 and HSF-1. Lastly, single-nucelotide polymorphisms (SNPs) in Tcf3/E2A are associated with exceptional longevity in humans. Together, these findings demonstrate that HLH-2 regulates energy metabolism via arginine kinases and thereby affects the aging phenotype dependent on ROS-signaling and established canonical effectors.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Longevity/genetics , Mice , Oxidation-Reduction , Transcription Factors/genetics , Transcription Factors/metabolism , Zebrafish/genetics , Zebrafish/metabolism
12.
Genes (Basel) ; 10(7)2019 07 13.
Article in English | MEDLINE | ID: mdl-31337008

ABSTRACT

Mitochondrial complex I-the largest enzyme complex of the mitochondrial oxidative phosphorylation machinery-has been proposed to contribute to a variety of age-related pathological alterations as well as longevity. The enzyme complex-consisting proteins are encoded by both nuclear (nDNA) and mitochondrial DNA (mtDNA). While some association studies of mtDNA encoded complex I genes and lifespan in humans have been reported, experimental evidence and the functional consequence of such variants is limited to studies using invertebrate models. Here, we present experimental evidence that a homoplasmic mutation in the mitochondrially encoded complex I gene mt-Nd2 modulates lifespan by altering cellular tryptophan levels and, consequently, ageing-related pathways in mice. A conplastic mouse strain carrying a mutation at m.4738C > A in mt-Nd2 lived slightly, but significantly, shorter than the controls did. The same mutation led to a higher susceptibility to glucose intolerance induced by high-fat diet feeding. These phenotypes were not observed in mice carrying a mutation in another mtDNA encoded complex I gene, mt-Nd5, suggesting the functional relevance of particular mutations in complex I to ageing and age-related diseases.


Subject(s)
Longevity/genetics , Maternal Inheritance , Mitochondrial Proteins/genetics , NADH Dehydrogenase/genetics , Animals , DNA, Mitochondrial , Diet, High-Fat , Female , Glucose Intolerance , Male , Metabolic Networks and Pathways/genetics , Mice, Inbred C57BL , Mutation , Stress, Physiological , Tryptophan/metabolism
13.
Sci Rep ; 8(1): 5872, 2018 04 12.
Article in English | MEDLINE | ID: mdl-29651131

ABSTRACT

Mutations in mitochondrial DNA (mtDNA) lead to heteroplasmy, i.e., the intracellular coexistence of wild-type and mutant mtDNA strands, which impact a wide spectrum of diseases but also physiological processes, including endurance exercise performance in athletes. However, the phenotypic consequences of limited levels of naturally arising heteroplasmy have not been experimentally studied to date. We hence generated a conplastic mouse strain carrying the mitochondrial genome of an AKR/J mouse strain (B6-mtAKR) in a C57BL/6 J nuclear genomic background, leading to >20% heteroplasmy in the origin of light-strand DNA replication (OriL). These conplastic mice demonstrate a shorter lifespan as well as dysregulation of multiple metabolic pathways, culminating in impaired glucose metabolism, compared to that of wild-type C57BL/6 J mice carrying lower levels of heteroplasmy. Our results indicate that physiologically relevant differences in mtDNA heteroplasmy levels at a single, functionally important site impair the metabolic health and lifespan in mice.


Subject(s)
DNA Replication/genetics , DNA, Mitochondrial/genetics , Longevity/genetics , Mitochondria/genetics , Animals , Glucose/genetics , Glucose/metabolism , Humans , Metabolic Networks and Pathways/genetics , Mice , Mitochondria/pathology , Mutation
14.
Cell Metab ; 27(4): 914-925.e5, 2018 04 03.
Article in English | MEDLINE | ID: mdl-29551589

ABSTRACT

Whether and how regulation of genes and pathways contributes to physiological aging is topic of intense scientific debate. By performing an RNA expression-based screen for genes downregulated during aging of three different species, we identified glycine-C-acetyltransferase (GCAT, EC 2.3.1.29). Impairing gcat expression promotes the lifespan of C. elegans by interfering with threonine catabolism to promote methylglyoxal (MGO; CAS 78-98-8) formation in an amine oxidase-dependent manner. MGO is a reactive dicarbonyl inducing diabetic complications in mammals by causing oxidative stress and damaging cellular components, including proteins. While high concentrations of MGO consistently exert toxicity in nematodes, we unexpectedly find that low-dose MGO promotes lifespan, resembling key mediators of gcat impairment. These were executed by the ubiquitin-proteasome system, namely PBS-3 and RPN-6.1 subunits, regulated by the stress-responsive transcriptional regulators SKN-1/NRF2 and HSF-1. Taken together, GCAT acts as an evolutionary conserved aging-related gene by orchestrating an unexpected nonlinear impact of proteotoxic MGO on longevity.


Subject(s)
Acetyltransferases/physiology , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Proteasome Endopeptidase Complex/metabolism , Pyruvaldehyde/metabolism , Threonine/metabolism , Acetyltransferases/genetics , Animals , DNA-Binding Proteins/metabolism , Longevity/physiology , Oxidative Stress , Signal Transduction , Transcription Factors/metabolism
15.
Aging Cell ; 16(4): 761-772, 2017 08.
Article in English | MEDLINE | ID: mdl-28544360

ABSTRACT

Impaired insulin/IGF1 signalling has been shown to extend lifespan in model organisms ranging from yeast to mammals. Here we sought to determine the effect of targeted disruption of the insulin receptor (IR) in non-neuronal tissues of adult mice on the lifespan. We induced hemizygous (PerIRKO+/- ) or homozygous (PerIRKO-/- ) disruption of the IR in peripheral tissue of 15-weeks-old mice using a tamoxifen-inducible Cre transgenic mouse with only peripheral tissue expression, and subsequently monitored glucose metabolism, insulin signalling and spontaneous death rates over 4 years. Complete peripheral IR disruption resulted in a diabetic phenotype with increased blood glucose and plasma insulin levels in young mice. Although blood glucose levels returned to normal, and fat mass was reduced in aged PerIRKO-/- mice, their lifespan was reduced. By contrast, heterozygous disruption had no effect on lifespan. This was despite young male PerIRKO+/- mice showing reduced fat mass and mild increase in hepatic insulin sensitivity. In conflict with findings in metazoans like Caenorhabditis elegans and Drosophila melanogaster, our results suggest that heterozygous impairment of the insulin signalling limited to peripheral tissues of adult mice fails to extend lifespan despite increased systemic insulin sensitivity, while homozygous impairment shortens lifespan.


Subject(s)
Adipose Tissue/metabolism , Insulin/metabolism , Longevity/genetics , Receptor, Insulin/genetics , Signal Transduction , Animals , Blood Glucose/metabolism , Gene Expression , Heterozygote , Homozygote , Integrases/genetics , Integrases/metabolism , Male , Mice , Mice, Knockout , Mutation , Receptor, Insulin/deficiency
16.
Cell Syst ; 2(5): 312-22, 2016 05 25.
Article in English | MEDLINE | ID: mdl-27211858

ABSTRACT

We present a genome-scale model of Caenorhabditis elegans metabolism along with the public database ElegCyc (http://elegcyc.bioinf.uni-jena.de:1100), which represents a reference for metabolic pathways in the worm and allows for the visualization as well as analysis of omics datasets. Our model reflects the metabolic peculiarities of C. elegans that make it distinct from other higher eukaryotes and mammals, including mice and humans. We experimentally verify one of these peculiarities by showing that the lifespan-extending effect of L-tryptophan supplementation is dose dependent (hormetic). Finally, we show the utility of our model for analyzing omics datasets through predicting changes in amino acid concentrations after genetic perturbations and analyzing metabolic changes during normal aging as well as during two distinct, reactive oxygen species (ROS)-related lifespan-extending treatments. Our analyses reveal a notable similarity in metabolic adaptation between distinct lifespan-extending interventions and point to key pathways affecting lifespan in nematodes.


Subject(s)
Caenorhabditis elegans , Databases, Genetic , Animals , Caenorhabditis elegans Proteins , Genome , Hormesis , Humans , Longevity , Mice , Reactive Oxygen Species
17.
Nat Commun ; 6: 10043, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26620638

ABSTRACT

Ageing has been defined as a global decline in physiological function depending on both environmental and genetic factors. Here we identify gene transcripts that are similarly regulated during physiological ageing in nematodes, zebrafish and mice. We observe the strongest extension of lifespan when impairing expression of the branched-chain amino acid transferase-1 (bcat-1) gene in C. elegans, which leads to excessive levels of branched-chain amino acids (BCAAs). We further show that BCAAs reduce a LET-363/mTOR-dependent neuro-endocrine signal, which we identify as DAF-7/TGFß, and that impacts lifespan depending on its related receptors, DAF-1 and DAF-4, as well as ultimately on DAF-16/FoxO and HSF-1 in a cell-non-autonomous manner. The transcription factor HLH-15 controls and epistatically synergizes with BCAT-1 to modulate physiological ageing. Lastly and consistent with previous findings in rodents, nutritional supplementation of BCAAs extends nematodal lifespan. Taken together, BCAAs act as periphery-derived metabokines that induce a central neuro-endocrine response, culminating in extended healthspan.


Subject(s)
Aging/metabolism , Amino Acids, Branched-Chain/metabolism , Caenorhabditis elegans/metabolism , Aging/genetics , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/growth & development , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Female , Longevity , Male , Mice/genetics , Mice/growth & development , Mice/metabolism , Mice, Inbred C57BL , Transaminases/genetics , Transaminases/metabolism , Zebrafish/genetics , Zebrafish/growth & development , Zebrafish/metabolism
18.
Cell Metab ; 21(3): 355-6, 2015 Mar 03.
Article in English | MEDLINE | ID: mdl-25738453

ABSTRACT

MOTS-c, a mitochondrially encoded open reading frame-derived peptide recently discovered by Lee et al. 2015 (this issue of Cell Metabolism) promotes biosynthesis of an endogenous AMP analog, AICAR. As AICAR activates AMPK, the discovery of MOTS-c offers an unexpected therapeutic option to be exploited toward the prevention of type 2 diabetes and delaying of the aging processes.


Subject(s)
Homeostasis/physiology , Insulin Resistance/physiology , Mitochondria/metabolism , Obesity/metabolism , Peptides/metabolism , Animals , Humans , Male
19.
Proc Natl Acad Sci U S A ; 111(40): 14512-7, 2014 Oct 07.
Article in English | MEDLINE | ID: mdl-25246545

ABSTRACT

In addition to signaling through the classical tyrosine kinase pathway, recent studies indicate that insulin receptors (IRs) and insulin-like growth factor 1 (IGF1) receptors (IGF1Rs) can emit signals in the unoccupied state through some yet-to-be-defined noncanonical pathways. Here we show that cells lacking both IRs and IGF1Rs exhibit a major decrease in expression of multiple imprinted genes and microRNAs, which is partially mimicked by inactivation of IR alone in mouse embryonic fibroblasts or in vivo in brown fat in mice. This down-regulation is accompanied by changes in DNA methylation of differentially methylated regions related to these loci. Different from a loss of imprinting pattern, loss of IR and IGF1R causes down-regulated expression of both maternally and paternally expressed imprinted genes and microRNAs, including neighboring reciprocally imprinted genes. Thus, the unoccupied IR and IGF1R generate previously unidentified signals that control expression of imprinted genes and miRNAs through transcriptional mechanisms that are distinct from classical imprinting control.


Subject(s)
Gene Expression/genetics , Genomic Imprinting/genetics , Receptor, IGF Type 1/deficiency , Receptor, Insulin/deficiency , Adipose Tissue, Brown/cytology , Adipose Tissue, Brown/metabolism , Animals , Azacitidine/pharmacology , Cell Line, Transformed , Cells, Cultured , DNA Methylation/drug effects , DNA Methylation/genetics , Embryo, Mammalian/cytology , Enzyme Inhibitors/pharmacology , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression/drug effects , Gene Expression Profiling , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Insulin/pharmacology , Insulin-Like Growth Factor I/pharmacology , Mice, Knockout , MicroRNAs/genetics , Oligonucleotide Array Sequence Analysis , Receptor, IGF Type 1/genetics , Receptor, Insulin/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics
20.
Nat Commun ; 5: 3563, 2014 Apr 08.
Article in English | MEDLINE | ID: mdl-24714520

ABSTRACT

D-Glucosamine (GlcN) is a freely available and commonly used dietary supplement potentially promoting cartilage health in humans, which also acts as an inhibitor of glycolysis. Here we show that GlcN, independent of the hexosamine pathway, extends Caenorhabditis elegans life span by impairing glucose metabolism that activates AMP-activated protein kinase (AMPK/AAK-2) and increases mitochondrial biogenesis. Consistent with the concept of mitohormesis, GlcN promotes increased formation of mitochondrial reactive oxygen species (ROS) culminating in increased expression of the nematodal amino acid-transporter 1 (aat-1) gene. Ameliorating mitochondrial ROS formation or impairment of aat-1-expression abolishes GlcN-mediated life span extension in an NRF2/SKN-1-dependent fashion. Unlike other calorie restriction mimetics, such as 2-deoxyglucose, GlcN extends life span of ageing C57BL/6 mice, which show an induction of mitochondrial biogenesis, lowered blood glucose levels, enhanced expression of several murine amino-acid transporters, as well as increased amino-acid catabolism. Taken together, we provide evidence that GlcN extends life span in evolutionary distinct species by mimicking a low-carbohydrate diet.


Subject(s)
Aging/drug effects , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/physiology , Glucosamine/pharmacology , Longevity/drug effects , Animals , Female , Hep G2 Cells , Humans , Male , Mice , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL