Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Circulation ; 145(25): 1853-1866, 2022 06 21.
Article in English | MEDLINE | ID: mdl-35616058

ABSTRACT

BACKGROUND: The insulin-like growth factor 1 (IGF1) pathway is a key regulator of cellular metabolism and aging. Although its inhibition promotes longevity across species, the effect of attenuated IGF1 signaling on cardiac aging remains controversial. METHODS: We performed a lifelong study to assess cardiac health and lifespan in 2 cardiomyocyte-specific transgenic mouse models with enhanced versus reduced IGF1 receptor (IGF1R) signaling. Male mice with human IGF1R overexpression or dominant negative phosphoinositide 3-kinase mutation were examined at different life stages by echocardiography, invasive hemodynamics, and treadmill coupled to indirect calorimetry. In vitro assays included cardiac histology, mitochondrial respiration, ATP synthesis, autophagic flux, and targeted metabolome profiling, and immunoblots of key IGF1R downstream targets in mouse and human explanted failing and nonfailing hearts, as well. RESULTS: Young mice with increased IGF1R signaling exhibited superior cardiac function that progressively declined with aging in an accelerated fashion compared with wild-type animals, resulting in heart failure and a reduced lifespan. In contrast, mice with low cardiac IGF1R signaling exhibited inferior cardiac function early in life, but superior cardiac performance during aging, and increased maximum lifespan, as well. Mechanistically, the late-life detrimental effects of IGF1R activation correlated with suppressed autophagic flux and impaired oxidative phosphorylation in the heart. Low IGF1R activity consistently improved myocardial bioenergetics and function of the aging heart in an autophagy-dependent manner. In humans, failing hearts, but not those with compensated hypertrophy, displayed exaggerated IGF1R expression and signaling activity. CONCLUSIONS: Our findings indicate that the relationship between IGF1R signaling and cardiac health is not linear, but rather biphasic. Hence, pharmacological inhibitors of the IGF1 pathway, albeit unsuitable for young individuals, might be worth considering in older adults.


Subject(s)
Insulin-Like Growth Factor I , Longevity , Aged , Animals , Health Promotion , Humans , Insulin-Like Growth Factor I/metabolism , Male , Mice , Myocytes, Cardiac/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism
3.
Cell Rep ; 35(2): 108985, 2021 04 13.
Article in English | MEDLINE | ID: mdl-33852843

ABSTRACT

Decreased cognitive performance is a hallmark of brain aging, but the underlying mechanisms and potential therapeutic avenues remain poorly understood. Recent studies have revealed health-protective and lifespan-extending effects of dietary spermidine, a natural autophagy-promoting polyamine. Here, we show that dietary spermidine passes the blood-brain barrier in mice and increases hippocampal eIF5A hypusination and mitochondrial function. Spermidine feeding in aged mice affects behavior in homecage environment tasks, improves spatial learning, and increases hippocampal respiratory competence. In a Drosophila aging model, spermidine boosts mitochondrial respiratory capacity, an effect that requires the autophagy regulator Atg7 and the mitophagy mediators Parkin and Pink1. Neuron-specific Pink1 knockdown abolishes spermidine-induced improvement of olfactory associative learning. This suggests that the maintenance of mitochondrial and autophagic function is essential for enhanced cognition by spermidine feeding. Finally, we show large-scale prospective data linking higher dietary spermidine intake with a reduced risk for cognitive impairment in humans.


Subject(s)
Aging/genetics , Autophagy-Related Protein 7/genetics , Cognitive Dysfunction/genetics , Dietary Supplements , Protein Kinases/genetics , Spermidine/pharmacology , Ubiquitin-Protein Ligases/genetics , Aging/metabolism , Animals , Autophagy-Related Protein 7/metabolism , Brain/cytology , Brain/drug effects , Brain/growth & development , Brain/metabolism , Cognition/drug effects , Cognition/physiology , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/physiopathology , Cognitive Dysfunction/prevention & control , Drosophila melanogaster/drug effects , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Female , Gene Expression Regulation , Humans , Learning/drug effects , Learning/physiology , Male , Mice , Mitochondria/drug effects , Mitochondria/genetics , Mitochondria/metabolism , Neurons/drug effects , Neurons/metabolism , Oxidative Phosphorylation/drug effects , Protein Kinases/metabolism , Signal Transduction , Spatial Memory/drug effects , Spatial Memory/physiology , Ubiquitin-Protein Ligases/metabolism
4.
Sci Transl Med ; 13(580)2021 02 10.
Article in English | MEDLINE | ID: mdl-33568522

ABSTRACT

Heart failure with preserved ejection fraction (HFpEF) is a highly prevalent and intractable form of cardiac decompensation commonly associated with diastolic dysfunction. Here, we show that diastolic dysfunction in patients with HFpEF is associated with a cardiac deficit in nicotinamide adenine dinucleotide (NAD+). Elevating NAD+ by oral supplementation of its precursor, nicotinamide, improved diastolic dysfunction induced by aging (in 2-year-old C57BL/6J mice), hypertension (in Dahl salt-sensitive rats), or cardiometabolic syndrome (in ZSF1 obese rats). This effect was mediated partly through alleviated systemic comorbidities and enhanced myocardial bioenergetics. Simultaneously, nicotinamide directly improved cardiomyocyte passive stiffness and calcium-dependent active relaxation through increased deacetylation of titin and the sarcoplasmic reticulum calcium adenosine triphosphatase 2a, respectively. In a long-term human cohort study, high dietary intake of naturally occurring NAD+ precursors was associated with lower blood pressure and reduced risk of cardiac mortality. Collectively, these results suggest NAD+ precursors, and especially nicotinamide, as potential therapeutic agents to treat diastolic dysfunction and HFpEF in humans.


Subject(s)
Heart Failure , Animals , Cohort Studies , Heart Failure/drug therapy , Humans , Mice , Mice, Inbred C57BL , Niacinamide/pharmacology , Niacinamide/therapeutic use , Rats , Rats, Inbred Dahl , Stroke Volume
5.
Annu Rev Nutr ; 40: 135-159, 2020 09 23.
Article in English | MEDLINE | ID: mdl-32634331

ABSTRACT

Natural polyamines (spermidine and spermine) are small, positively charged molecules that are ubiquitously found within organisms and cells. They exert numerous (intra)cellular functions and have been implicated to protect against several age-related diseases. Although polyamine levels decline in a complex age-dependent, tissue-, and cell type-specific manner, they are maintained in healthy nonagenarians and centenarians. Increased polyamine levels, including through enhanced dietary intake, have been consistently linked to improved health and reduced overall mortality. In preclinical models, dietary supplementation with spermidine prolongs life span and health span. In this review, we highlight salient aspects of nutritional polyamine intake and summarize the current knowledge of organismal and cellular uptake and distribution of dietary (and gastrointestinal) polyamines and their impact on human health. We further summarize clinical and epidemiological studies of dietary polyamines.


Subject(s)
Food Analysis , Nutritive Value , Spermidine/metabolism , Animals , Diet , Humans
6.
Cell Death Differ ; 27(10): 2904-2920, 2020 10.
Article in English | MEDLINE | ID: mdl-32376874

ABSTRACT

The pharmacological targeting of polyamine metabolism is currently under the spotlight for its potential in the prevention and treatment of several age-associated disorders. Here, we report the finding that triethylenetetramine dihydrochloride (TETA), a copper-chelator agent that can be safely administered to patients for the long-term treatment of Wilson disease, exerts therapeutic benefits in animals challenged with hypercaloric dietary regimens. TETA reduced obesity induced by high-fat diet, excessive sucrose intake, or leptin deficiency, as it reduced glucose intolerance and hepatosteatosis, but induced autophagy. Mechanistically, these effects did not involve the depletion of copper from plasma or internal organs. Rather, the TETA effects relied on the activation of an energy-consuming polyamine catabolism, secondary to the stabilization of spermidine/spermine N1-acetyltransferase-1 (SAT1) by TETA, resulting in enhanced enzymatic activity of SAT. All the positive effects of TETA on high-fat diet-induced metabolic syndrome were lost in SAT1-deficient mice. Altogether, these results suggest novel health-promoting effects of TETA that might be taken advantage of for the prevention or treatment of obesity.


Subject(s)
Acetyltransferases/metabolism , Chelating Agents/pharmacology , Obesity/drug therapy , Trientine/analogs & derivatives , Animals , Diet, High-Fat , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/chemically induced
8.
EMBO Mol Med ; 11(11): e10469, 2019 11 07.
Article in English | MEDLINE | ID: mdl-31609086

ABSTRACT

Caloric restriction mimetics (CRMs) are natural or synthetic compounds that mimic the health-promoting and longevity-extending effects of caloric restriction. CRMs provoke the deacetylation of cellular proteins coupled to an increase in autophagic flux in the absence of toxicity. Here, we report the identification of a novel candidate CRM, namely 3,4-dimethoxychalcone (3,4-DC), among a library of polyphenols. When added to several different human cell lines, 3,4-DC induced the deacetylation of cytoplasmic proteins and stimulated autophagic flux. At difference with other well-characterized CRMs, 3,4-DC, however, required transcription factor EB (TFEB)- and E3 (TFE3)-dependent gene transcription and mRNA translation to trigger autophagy. 3,4-DC stimulated the translocation of TFEB and TFE3 into nuclei both in vitro and in vivo, in hepatocytes and cardiomyocytes. 3,4-DC induced autophagy in vitro and in mouse organs, mediated autophagy-dependent cardioprotective effects, and improved the efficacy of anticancer chemotherapy in vivo. Altogether, our results suggest that 3,4-DC is a novel CRM with a previously unrecognized mode of action.


Subject(s)
Autophagy/drug effects , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cardiotonic Agents/metabolism , Chalcones/metabolism , Transcription Factors/metabolism , Transcription, Genetic/drug effects , Acetylation , Animal Structures/pathology , Animals , Cardiotonic Agents/administration & dosage , Cell Line , Chalcones/administration & dosage , Hepatocytes/drug effects , Humans , Mice , Myocytes, Cardiac/drug effects , Protein Processing, Post-Translational , Protein Transport
9.
FASEB J ; 33(12): 13808-13824, 2019 12.
Article in English | MEDLINE | ID: mdl-31638418

ABSTRACT

N-acetylaspartate (NAA) is synthesized by aspartate N-acetyltransferase (gene: Nat8l) from acetyl-coenzyme A and aspartate. In the brain, NAA is considered an important energy metabolite for lipid synthesis. However, the role of NAA in peripheral tissues remained elusive. Therefore, we characterized the metabolic phenotype of knockout (ko) and adipose tissue-specific (ako) Nat8l-ko mice as well as NAA-supplemented mice on various diets. We identified an important role of NAA availability in the brain during adolescence, as 75% of Nat8l-ko mice died on fat-free diet (FFD) after weaning but could be rescued by NAA supplementation. In adult life, NAA deficiency promotes a beneficial metabolic phenotype, as Nat8l-ko and Nat8l-ako mice showed reduced body weight, increased energy expenditure, and improved glucose tolerance on chow, high-fat, and FFDs. Furthermore, Nat8l-deficient adipocytes exhibited increased mitochondrial respiration, ATP synthesis, and an induction of browning. Conversely, NAA-treated wild-type mice showed reduced adipocyte respiration and lipolysis and increased de novo lipogenesis, culminating in reduced energy expenditure, glucose tolerance, and insulin sensitivity. Mechanistically, our data point to a possible role of NAA as modulator of pancreatic insulin secretion and suggest NAA as a critical energy metabolite for adipocyte and whole-body energy homeostasis.-Hofer, D. C., Zirkovits, G., Pelzmann, H. J., Huber, K., Pessentheiner, A. R., Xia, W., Uno, K., Miyazaki, T., Kon, K., Tsuneki, H., Pendl, T., Al Zoughbi, W., Madreiter-Sokolowski, C. T., Trausinger, G., Abdellatif, M., Schoiswohl, G., Schreiber, R., Eisenberg, T., Magnes, C., Sedej, S., Eckhardt, M., Sasahara, M., Sasaoka, T., Nitta, A., Hoefler, G., Graier, W. F., Kratky, D., Auwerx, J., Bogner-Strauss, J. G. N-acetylaspartate availability is essential for juvenile survival on fat-free diet and determines metabolic health.


Subject(s)
Aspartic Acid/analogs & derivatives , Acetyl Coenzyme A/metabolism , Acetyltransferases/metabolism , Adipocytes/metabolism , Animals , Aspartic Acid/metabolism , Brain/metabolism , Diet, Fat-Restricted , Energy Metabolism/physiology , Insulin Resistance/physiology , Lipolysis/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism
10.
Cell Metab ; 30(3): 462-476.e6, 2019 09 03.
Article in English | MEDLINE | ID: mdl-31471173

ABSTRACT

Caloric restriction and intermittent fasting are known to prolong life- and healthspan in model organisms, while their effects on humans are less well studied. In a randomized controlled trial study (ClinicalTrials.gov identifier: NCT02673515), we show that 4 weeks of strict alternate day fasting (ADF) improved markers of general health in healthy, middle-aged humans while causing a 37% calorie reduction on average. No adverse effects occurred even after >6 months. ADF improved cardiovascular markers, reduced fat mass (particularly the trunk fat), improving the fat-to-lean ratio, and increased ß-hydroxybutyrate, even on non-fasting days. On fasting days, the pro-aging amino-acid methionine, among others, was periodically depleted, while polyunsaturated fatty acids were elevated. We found reduced levels sICAM-1 (an age-associated inflammatory marker), low-density lipoprotein, and the metabolic regulator triiodothyronine after long-term ADF. These results shed light on the physiological impact of ADF and supports its safety. ADF could eventually become a clinically relevant intervention.


Subject(s)
Aging/blood , Fasting/adverse effects , Fasting/blood , Healthy Aging/blood , 3-Hydroxybutyric Acid/blood , Adult , Biomarkers/blood , Body Mass Index , Caloric Restriction/adverse effects , Energy Intake/physiology , Fatty Acids, Unsaturated/blood , Female , Healthy Volunteers , Humans , Intercellular Adhesion Molecule-1/blood , Lipoproteins, LDL/blood , Male , Middle Aged , Pilot Projects , Prospective Studies , Triiodothyronine/blood , Weight Loss
11.
J Biol Chem ; 294(32): 12020-12039, 2019 08 09.
Article in English | MEDLINE | ID: mdl-31209110

ABSTRACT

Autophagy, a membrane-dependent catabolic process, ensures survival of aging cells and depends on the cellular energetic status. Acetyl-CoA carboxylase 1 (Acc1) connects central energy metabolism to lipid biosynthesis and is rate-limiting for the de novo synthesis of lipids. However, it is unclear how de novo lipogenesis and its metabolic consequences affect autophagic activity. Here, we show that in aging yeast, autophagy levels highly depend on the activity of Acc1. Constitutively active Acc1 (acc1S/A ) or a deletion of the Acc1 negative regulator, Snf1 (yeast AMPK), shows elevated autophagy levels, which can be reversed by the Acc1 inhibitor soraphen A. Vice versa, pharmacological inhibition of Acc1 drastically reduces cell survival and results in the accumulation of Atg8-positive structures at the vacuolar membrane, suggesting late defects in the autophagic cascade. As expected, acc1S/A cells exhibit a reduction in acetate/acetyl-CoA availability along with elevated cellular lipid content. However, concomitant administration of acetate fails to fully revert the increase in autophagy exerted by acc1S/A Instead, administration of oleate, while mimicking constitutively active Acc1 in WT cells, alleviates the vacuolar fusion defects induced by Acc1 inhibition. Our results argue for a largely lipid-dependent process of autophagy regulation downstream of Acc1. We present a versatile genetic model to investigate the complex relationship between acetate metabolism, lipid homeostasis, and autophagy and propose Acc1-dependent lipogenesis as a fundamental metabolic path downstream of Snf1 to maintain autophagy and survival during cellular aging.


Subject(s)
Acetyl-CoA Carboxylase/metabolism , Autophagy , Lipogenesis , Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Acetates/metabolism , Acetyl-CoA Carboxylase/antagonists & inhibitors , Acetyl-CoA Carboxylase/genetics , Autophagy/drug effects , Macrolides/pharmacology , Mutagenesis, Site-Directed , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Saccharomyces cerevisiae Proteins/genetics
12.
Nat Commun ; 10(1): 651, 2019 02 19.
Article in English | MEDLINE | ID: mdl-30783116

ABSTRACT

Ageing constitutes the most important risk factor for all major chronic ailments, including malignant, cardiovascular and neurodegenerative diseases. However, behavioural and pharmacological interventions with feasible potential to promote health upon ageing remain rare. Here we report the identification of the flavonoid 4,4'-dimethoxychalcone (DMC) as a natural compound with anti-ageing properties. External DMC administration extends the lifespan of yeast, worms and flies, decelerates senescence of human cell cultures, and protects mice from prolonged myocardial ischaemia. Concomitantly, DMC induces autophagy, which is essential for its cytoprotective effects from yeast to mice. This pro-autophagic response induces a conserved systemic change in metabolism, operates independently of TORC1 signalling and depends on specific GATA transcription factors. Notably, we identify DMC in the plant Angelica keiskei koidzumi, to which longevity- and health-promoting effects are ascribed in Asian traditional medicine. In summary, we have identified and mechanistically characterised the conserved longevity-promoting effects of a natural anti-ageing drug.


Subject(s)
Aging/drug effects , Autophagy/drug effects , Flavonoids/pharmacology , Longevity/drug effects , Aging/physiology , Angelica/chemistry , Animals , Caenorhabditis elegans/drug effects , Cation Transport Proteins/genetics , Cell Death/drug effects , Cell Line/drug effects , Drosophila melanogaster/drug effects , Flavonoids/administration & dosage , GATA Transcription Factors/drug effects , Gene Expression Regulation/drug effects , Humans , Longevity/physiology , Male , Mechanistic Target of Rapamycin Complex 1/metabolism , Medicine, East Asian Traditional , Mice , Mice, Inbred C57BL , Myocardial Ischemia/drug therapy , Plant Extracts/pharmacology , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Signal Transduction , Sirolimus/pharmacology , Transcription Factors/drug effects , Transcription Factors/genetics
13.
Front Mol Neurosci ; 11: 318, 2018.
Article in English | MEDLINE | ID: mdl-30233317

ABSTRACT

Huntington's disease (HD) is a neurodegenerative disorder that leads to progressive neuronal loss, provoking impaired motor control, cognitive decline, and dementia. So far, HD remains incurable, and available drugs are effective only for symptomatic management. HD is caused by a mutant form of the huntingtin protein, which harbors an elongated polyglutamine domain and is highly prone to aggregation. However, many aspects underlying the cytotoxicity of mutant huntingtin (mHTT) remain elusive, hindering the efficient development of applicable interventions to counteract HD. An important strategy to obtain molecular insights into human disorders in general is the use of eukaryotic model organisms, which are easy to genetically manipulate and display a high degree of conservation regarding disease-relevant cellular processes. The budding yeast Saccharomyces cerevisiae has a long-standing and successful history in modeling a plethora of human maladies and has recently emerged as an effective tool to study neurodegenerative disorders, including HD. Here, we summarize some of the most important contributions of yeast to HD research, specifically concerning the elucidation of mechanistic features of mHTT cytotoxicity and the potential of yeast as a platform to screen for pharmacological agents against HD.

14.
FEMS Yeast Res ; 18(6)2018 09 01.
Article in English | MEDLINE | ID: mdl-29905792

ABSTRACT

In the search for interventions against aging and age-related diseases, biological screening platforms are indispensable tools to identify anti-aging compounds among large substance libraries. The budding yeast, Saccharomyces cerevisiae, has emerged as a powerful chemical and genetic screening platform, as it combines a rapid workflow with experimental amenability and the availability of a wide range of genetic mutant libraries. Given the amount of conserved genes and aging mechanisms between yeast and human, testing candidate anti-aging substances in yeast gene-deletion or overexpression collections, or de novo derived mutants, has proven highly successful in finding potential molecular targets. Yeast-based studies, for example, have led to the discovery of the polyphenol resveratrol and the natural polyamine spermidine as potential anti-aging agents. Here, we present strategies for pharmacological anti-aging screens in yeast, discuss common pitfalls and summarize studies that have used yeast for drug discovery and target identification.


Subject(s)
Aging/drug effects , Drug Discovery , Models, Biological , Saccharomyces cerevisiae/drug effects , Small Molecule Libraries/pharmacology , Aging/genetics , Aging/pathology , Gene Library , Genetic Testing , Humans , Microbial Viability/drug effects , Microbial Viability/genetics , Phenotype , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development
15.
Aging (Albany NY) ; 10(1): 19-33, 2018 01 08.
Article in English | MEDLINE | ID: mdl-29315079

ABSTRACT

Supplementation of spermidine, an autophagy-inducing agent, has been shown to protect against neurodegeneration and cognitive decline in aged animal models. The present translational study aimed to determine safety and tolerability of a wheat germ extract containing enhanced spermidine concentrations. In a preclinical toxicity study, supplementation of spermidine using this extract did not result in morbidities or changes in behavior in BALBc/Rj mice during the 28-days repeated-dose tolerance study. Post mortem examination of the mice organs showed no increase in tumorigenic and fibrotic events. In the human cohort (participants with subjective cognitive decline, n=30, 60 to 80 years of age), a 3-month randomized, placebo-controlled, double-blind Phase II trial was conducted with supplementation of the spermidine-rich plant extract (dosage: 1.2 mg/day). No differences were observed between spermidine and placebo-treated groups in vital signs, weight, clinical chemistry and hematological parameters of safety, as well as in self-reported health status at the end of intervention. Compliance rates above 85% indicated excellent tolerability. The data demonstrate that spermidine supplementation using a spermidine-rich plant extract is safe and well-tolerated in mice and older adults. These findings allow for longer-term intervention studies in humans to investigate the impact of spermidine treatment on cognition and brain integrity.


Subject(s)
Cognition/drug effects , Plant Extracts/administration & dosage , Plant Extracts/pharmacology , Spermidine/pharmacology , Administration, Oral , Aged , Aged, 80 and over , Aging , Animals , Cognitive Dysfunction/drug therapy , Double-Blind Method , Female , Humans , Male , Mice , Mice, Inbred BALB C , Middle Aged , Plant Extracts/adverse effects , Spermidine/administration & dosage , Spermidine/adverse effects
16.
Autophagy ; 13(4): 767-769, 2017 Apr 03.
Article in English | MEDLINE | ID: mdl-28118075

ABSTRACT

Loss of cardiac macroautophagy/autophagy impairs heart function, and evidence accumulates that an increased autophagic flux may protect against cardiovascular disease. We therefore tested the protective capacity of the natural autophagy inducer spermidine in animal models of aging and hypertension, which both represent major risk factors for the development of cardiovascular disease. Dietary spermidine elicits cardioprotective effects in aged mice through enhancing cardiac autophagy and mitophagy. In salt-sensitive rats, spermidine supplementation also delays the development of hypertensive heart disease, coinciding with reduced arterial blood pressure. The high blood pressure-lowering effect likely results from improved global arginine bioavailability and protection from hypertension-associated renal damage. The polyamine spermidine is naturally present in human diets, though to a varying amount depending on food type and preparation. In humans, high dietary spermidine intake correlates with reduced blood pressure and decreased risk of cardiovascular disease and related death. Altogether, spermidine represents a cardio- and vascular-protective autophagy inducer that can be readily integrated in common diets.


Subject(s)
Hypertension/drug therapy , Spermidine/therapeutic use , Animals , Humans , Mice, Inbred C57BL , Models, Biological , Rats, Inbred Dahl , Spermidine/pharmacology
17.
Nat Med ; 22(12): 1428-1438, 2016 12.
Article in English | MEDLINE | ID: mdl-27841876

ABSTRACT

Aging is associated with an increased risk of cardiovascular disease and death. Here we show that oral supplementation of the natural polyamine spermidine extends the lifespan of mice and exerts cardioprotective effects, reducing cardiac hypertrophy and preserving diastolic function in old mice. Spermidine feeding enhanced cardiac autophagy, mitophagy and mitochondrial respiration, and it also improved the mechano-elastical properties of cardiomyocytes in vivo, coinciding with increased titin phosphorylation and suppressed subclinical inflammation. Spermidine feeding failed to provide cardioprotection in mice that lack the autophagy-related protein Atg5 in cardiomyocytes. In Dahl salt-sensitive rats that were fed a high-salt diet, a model for hypertension-induced congestive heart failure, spermidine feeding reduced systemic blood pressure, increased titin phosphorylation and prevented cardiac hypertrophy and a decline in diastolic function, thus delaying the progression to heart failure. In humans, high levels of dietary spermidine, as assessed from food questionnaires, correlated with reduced blood pressure and a lower incidence of cardiovascular disease. Our results suggest a new and feasible strategy for protection against cardiovascular disease.


Subject(s)
Aging/drug effects , Autophagy/drug effects , Blood Pressure/drug effects , Heart/drug effects , Longevity/drug effects , Mitochondria, Heart/drug effects , Mitophagy/drug effects , Myocytes, Cardiac/drug effects , Spermidine/pharmacology , Adult , Aged , Aging/immunology , Aging/metabolism , Animals , Autophagy-Related Protein 5/genetics , Cardiomegaly/diagnostic imaging , Cardiotonic Agents/pharmacology , Cardiovascular Diseases/epidemiology , Chromatography, High Pressure Liquid , Connectin/drug effects , Connectin/metabolism , Cytokines/drug effects , Cytokines/immunology , Diastole , Diet/statistics & numerical data , Echocardiography , Female , Gene Expression/drug effects , Glucose Tolerance Test , Heart/diagnostic imaging , Heart Failure , Humans , Immunoblotting , Inflammation , Male , Mass Spectrometry , Mice , Middle Aged , Mitochondria, Heart/metabolism , Phosphorylation/drug effects , Prospective Studies , Rats , Rats, Inbred Dahl , Surveys and Questionnaires
18.
PLoS One ; 10(3): e0120250, 2015.
Article in English | MEDLINE | ID: mdl-25767889

ABSTRACT

The target of rapamycin complex 1 (TORC1) is an evolutionarily conserved sensor of nutrient availability. Genetic and pharmacological studies in the yeast Saccharomyces cerevisiae have provided mechanistic insights on the regulation of TORC1 signaling in response to nutrients. Using a highly specific antibody that recognizes phosphorylation of the bona fide TORC1 target ribosomal protein S6 (Rps6) in yeast, we found that nutrients rapidly induce Rps6 phosphorylation in a TORC1-dependent manner. Moreover, we demonstrate that Ypk3, an AGC kinase which exhibits high homology to human S6 kinase (S6K), is required for the phosphorylation of Rps6 in vivo. Rps6 phosphorylation is completely abolished in cells lacking Ypk3 (ypk3Δ), whereas Sch9, previously reported to be the yeast ortholog of S6K, is dispensable for Rps6 phosphorylation. Phosphorylation-deficient mutations in regulatory motifs of Ypk3 abrogate Rps6 phosphorylation, and complementation of ypk3Δ cells with human S6 kinase restores Rps6 phosphorylation in a rapamycin-sensitive manner. Our findings demonstrate that Ypk3 is a critical component of the TORC1 pathway and that the use of a phospho-S6 specific antibody offers a valuable tool to identify new nutrient-dependent and rapamycin-sensitive targets in vivo.


Subject(s)
Cyclic Nucleotide-Regulated Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Ribosomal Protein S6 Kinases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Transcription Factors/metabolism , Analysis of Variance , Escherichia coli , Immunoblotting , Phosphorylation , Plasmids/genetics , Polymerase Chain Reaction
19.
Autophagy ; 10(6): 1143-5, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24879160

ABSTRACT

The multifaceted process of aging inevitably leads to disturbances in cellular metabolism and protein homeostasis. To meet this challenge, cells make use of autophagy, which is probably one of the most important pathways preserving cellular protection under stressful conditions. Thus, efficient autophagic flux is required for healthy aging in many if not all eukaryotic organisms. The regulation of autophagy itself is affected by changing metabolic conditions, but the precise metabolic circuitries are poorly understood. Recently, we found that the nucleocytosolic pool of acetyl-coenzyme A (AcCoA) functions as a major and dominant suppressor of cytoprotective autophagy during aging. Here, we propose an epigenetic mechanism for AcCoA-mediated autophagy suppression that causally involves the regulation of histone acetylation and changes in the autophagy-relevant transcriptome.


Subject(s)
Autophagy , Coenzyme A Ligases/metabolism , Drosophila Proteins/metabolism , Longevity , Animals
20.
Autophagy ; 10(7): 1335-7, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24904996

ABSTRACT

As the major lysosomal degradation pathway, autophagy represents the guardian of cellular homeostasis, removing damaged and potentially harmful material and replenishing energy reserves in conditions of starvation. Given its vast physiological importance, autophagy is crucially involved in the process of aging and associated pathologies. Although the regulation of autophagy strongly depends on nutrient availability, specific metabolites that modulate autophagic responses are poorly described. Recently, we revealed nucleo-cytosolic acetyl-coenzyme A (AcCoA) as a phylogenetically conserved inhibitor of starvation-induced and age-associated autophagy. AcCoA is the sole acetyl-group donor for protein acetylation, explaining why pharmacological or genetic manipulations that modify the concentrations of nucleo-cytosolic AcCoA directly affect the levels of protein acetylation. The acetylation of histones and cytosolic proteins inversely correlates with the rate of autophagy in yeast and mammalian cells, respectively, despite the fact that the routes of de novo AcCoA synthesis differ across phyla. Thus, we propose nucleo-cytosolic AcCoA to act as a conserved metabolic rheostat, linking the cellular metabolic state to the regulation of autophagy via effects on protein acetylation.


Subject(s)
Acetyl Coenzyme A/metabolism , Autophagy , Longevity , Metabolic Networks and Pathways , Animals , Cell Nucleus/metabolism , Cytosol/metabolism , Humans , Mice , Models, Biological , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...