Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Trends Genet ; 39(8): 587-592, 2023 08.
Article in English | MEDLINE | ID: mdl-37244757
2.
J Physiol ; 601(11): 2139-2163, 2023 06.
Article in English | MEDLINE | ID: mdl-36086823

ABSTRACT

Low-protein (LP) diets are associated with a decreased risk of diabetes in humans, and promote leanness and glycaemic control in both rodents and humans. While the effects of an LP diet on glycaemic control are mediated by reduced levels of the branched-chain amino acids, we have observed that reducing dietary levels of the other six essential amino acids leads to changes in body composition. Here, we find that dietary histidine plays a key role in the response to an LP diet in male C57BL/6J mice. Specifically reducing dietary levels of histidine by 67% reduces the weight gain of young, lean male mice, reducing both adipose and lean mass without altering glucose metabolism, and rapidly reverses diet-induced obesity and hepatic steatosis in diet-induced obese male mice, increasing insulin sensitivity. This normalization of metabolic health was associated not with caloric restriction or increased activity, but with increased energy expenditure. Surprisingly, the effects of histidine restriction do not require the energy balance hormone Fgf21. Histidine restriction that was started in midlife promoted leanness and glucose tolerance in aged males but not females, but did not affect frailty or lifespan in either sex. Finally, we demonstrate that variation in dietary histidine levels helps to explain body mass index differences in humans. Overall, our findings demonstrate that dietary histidine is a key regulator of weight and body composition in male mice and in humans, and suggest that reducing dietary histidine may be a translatable option for the treatment of obesity. KEY POINTS: Protein restriction (PR) promotes metabolic health in rodents and humans and extends rodent lifespan. Restriction of specific individual essential amino acids can recapitulate the benefits of PR. Reduced histidine promotes leanness and increased energy expenditure in male mice. Reduced histidine does not extend the lifespan of mice when begun in midlife. Dietary levels of histidine are positively associated with body mass index in humans.


Subject(s)
Histidine , Thinness , Male , Humans , Animals , Mice , Aged , Histidine/metabolism , Mice, Inbred C57BL , Diet , Obesity/metabolism , Proteins , Energy Metabolism
3.
Nat Metab ; 3(10): 1327-1341, 2021 10.
Article in English | MEDLINE | ID: mdl-34663973

ABSTRACT

Calorie restriction (CR) promotes healthy ageing in diverse species. Recently, it has been shown that fasting for a portion of each day has metabolic benefits and promotes lifespan. These findings complicate the interpretation of rodent CR studies, in which animals typically eat only once per day and rapidly consume their food, which collaterally imposes fasting. Here we show that a prolonged fast is necessary for key metabolic, molecular and geroprotective effects of a CR diet. Using a series of feeding regimens, we dissect the effects of calories and fasting, and proceed to demonstrate that fasting alone recapitulates many of the physiological and molecular effects of CR. Our results shed new light on how both when and how much we eat regulate metabolic health and longevity, and demonstrate that daily prolonged fasting, and not solely reduced caloric intake, is likely responsible for the metabolic and geroprotective benefits of a CR diet.


Subject(s)
Aging/metabolism , Caloric Restriction , Animals , Longevity/physiology , Mice
4.
Cell Metab ; 33(5): 905-922.e6, 2021 05 04.
Article in English | MEDLINE | ID: mdl-33887198

ABSTRACT

Low-protein diets promote metabolic health in rodents and humans, and the benefits of low-protein diets are recapitulated by specifically reducing dietary levels of the three branched-chain amino acids (BCAAs), leucine, isoleucine, and valine. Here, we demonstrate that each BCAA has distinct metabolic effects. A low isoleucine diet reprograms liver and adipose metabolism, increasing hepatic insulin sensitivity and ketogenesis and increasing energy expenditure, activating the FGF21-UCP1 axis. Reducing valine induces similar but more modest metabolic effects, whereas these effects are absent with low leucine. Reducing isoleucine or valine rapidly restores metabolic health to diet-induced obese mice. Finally, we demonstrate that variation in dietary isoleucine levels helps explain body mass index differences in humans. Our results reveal isoleucine as a key regulator of metabolic health and the adverse metabolic response to dietary BCAAs and suggest reducing dietary isoleucine as a new approach to treating and preventing obesity and diabetes.


Subject(s)
Amino Acids, Branched-Chain/metabolism , Diet , Isoleucine/metabolism , Valine/metabolism , Adipose Tissue, White/metabolism , Animals , Body Mass Index , Diet/veterinary , Energy Metabolism , Fibroblast Growth Factors/deficiency , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Humans , Liver/metabolism , Male , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/metabolism , Obesity/pathology , Protein Serine-Threonine Kinases/metabolism , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism
5.
BMC Biol ; 19(1): 31, 2021 02 16.
Article in English | MEDLINE | ID: mdl-33593351

ABSTRACT

BACKGROUND: Proper regulation of feeding is important for an organism's well-being and survival and involves a motivational component directing the search for food. Dissecting the molecular and neural mechanisms of motivated feeding behavior requires assays that allow quantification of both motivation and food intake. Measurements of motivated behavior usually involve assessing physical effort or overcoming an aversive stimulus. Food intake in Drosophila can be determined in a number of ways, including by measuring the time a fly's proboscis interacts with a food source associated with an electrical current in the fly liquid-food interaction counter (FLIC). Here, we show that electrical current flowing through flies during this interaction is aversive, and we describe a modified assay to measure motivation in Drosophila. RESULTS: Food intake is reduced during the interaction with FLIC when the electrical current is turned on, which provides a confounding variable in studies of motivated behavior. Based on the FLIC, we engineer a novel assay, the fly liquid-food electroshock assay (FLEA), which allows for current adjustments for each feeding well. Using the FLEA, we show that both external incentives and internal motivational state can serve as drivers for flies to overcome higher current (electric shock) to obtain superior food. Unlike similar assays in which bitterness is the aversive stimulus for the fly to overcome, we show that current perception is not discounted as flies become more food-deprived. Finally, we use genetically manipulated flies to show that neuropeptide F, an orthologue of mammalian NPY previously implicated in regulation of feeding motivation, is required for sensory processing of electrical current. CONCLUSION: The FLEA is therefore a novel assay to accurately measure incentive motivation in Drosophila. Using the FLEA, we also show that neuropeptide F is required for proper perception or processing of an electroshock, a novel function for this neuropeptide involved in the processing of external and internal stimuli.


Subject(s)
Drosophila melanogaster/physiology , Electroshock , Insect Proteins/metabolism , Neuropeptides/metabolism , Animals , Avoidance Learning/physiology , Feeding Behavior/physiology , Food/classification , Male , Taste Perception/physiology
6.
Mol Cell ; 78(2): 210-223.e8, 2020 04 16.
Article in English | MEDLINE | ID: mdl-32208170

ABSTRACT

S-adenosylmethionine (SAM) is the methyl-donor substrate for DNA and histone methyltransferases that regulate epigenetic states and subsequent gene expression. This metabolism-epigenome link sensitizes chromatin methylation to altered SAM abundance, yet the mechanisms that allow organisms to adapt and protect epigenetic information during life-experienced fluctuations in SAM availability are unknown. We identified a robust response to SAM depletion that is highlighted by preferential cytoplasmic and nuclear mono-methylation of H3 Lys 9 (H3K9) at the expense of broad losses in histone di- and tri-methylation. Under SAM-depleted conditions, H3K9 mono-methylation preserves heterochromatin stability and supports global epigenetic persistence upon metabolic recovery. This unique chromatin response was robust across the mouse lifespan and correlated with improved metabolic health, supporting a significant role for epigenetic adaptation to SAM depletion in vivo. Together, these studies provide evidence for an adaptive response that enables epigenetic persistence to metabolic stress.


Subject(s)
DNA Methylation/genetics , Heterochromatin/genetics , Metabolome/genetics , S-Adenosylmethionine/metabolism , Animals , Cell Nucleus/genetics , Cell Nucleus/metabolism , Chromatin/genetics , Cytoplasm/genetics , Cytoplasm/metabolism , Epigenesis, Genetic/genetics , Gene Expression Regulation/genetics , HCT116 Cells , Heterochromatin/metabolism , Histone-Lysine N-Methyltransferase/genetics , Histones/genetics , Humans , Methionine/genetics , Mice , Protein Processing, Post-Translational/genetics , Proteomics/methods
7.
Cell Rep ; 29(1): 236-248.e3, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31577953

ABSTRACT

Calorie restriction (CR) extends the healthspan and lifespan of diverse species. In mammals, a broadly conserved metabolic effect of CR is improved insulin sensitivity, which may mediate the beneficial effects of a CR diet. This model has been challenged by the identification of interventions that extend lifespan and healthspan yet promote insulin resistance. These include rapamycin, which extends mouse lifespan yet induces insulin resistance by disrupting mTORC2 (mechanistic target of rapamycin complex 2). Here, we induce insulin resistance by genetically disrupting adipose mTORC2 via tissue-specific deletion of the mTORC2 component Rictor (AQ-RKO). Loss of adipose mTORC2 blunts the metabolic adaptation to CR and prevents whole-body sensitization to insulin. Despite this, AQ-RKO mice subject to CR experience the same increase in fitness and lifespan on a CR diet as wild-type mice. We conclude that the CR-induced improvement in insulin sensitivity is dispensable for the effects of CR on fitness and longevity.


Subject(s)
Adiposity/physiology , Insulin Resistance/physiology , Insulin/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Adiposity/drug effects , Animals , Caloric Restriction/methods , Energy Intake/drug effects , Energy Intake/physiology , Humans , Longevity/drug effects , Longevity/physiology , Mice , Mice, Inbred C57BL , Signal Transduction/drug effects , Signal Transduction/physiology , Sirolimus/pharmacology
8.
Nat Commun ; 10(1): 3194, 2019 07 19.
Article in English | MEDLINE | ID: mdl-31324799

ABSTRACT

Rapamycin, an inhibitor of mechanistic Target Of Rapamycin Complex 1 (mTORC1), extends lifespan and shows strong potential for the treatment of age-related diseases. However, rapamycin exerts metabolic and immunological side effects mediated by off-target inhibition of a second mTOR-containing complex, mTOR complex 2. Here, we report the identification of DL001, a FKBP12-dependent rapamycin analog 40x more selective for mTORC1 than rapamycin. DL001 inhibits mTORC1 in cell culture lines and in vivo in C57BL/6J mice, in which DL001 inhibits mTORC1 signaling without impairing glucose homeostasis and with substantially reduced or no side effects on lipid metabolism and the immune system. In cells, DL001 efficiently represses elevated mTORC1 activity and restores normal gene expression to cells lacking a functional tuberous sclerosis complex. Our results demonstrate that highly selective pharmacological inhibition of mTORC1 can be achieved in vivo, and that selective inhibition of mTORC1 significantly reduces the side effects associated with conventional rapalogs.


Subject(s)
Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Sirolimus/analogs & derivatives , Sirolimus/pharmacology , Animals , Cell Line , Drug Discovery , Gene Expression/drug effects , Humans , Immune System/drug effects , Lipid Metabolism/drug effects , Mice , Mice, Inbred C57BL , Proteomics , Signal Transduction/drug effects , Sirolimus/chemistry , TOR Serine-Threonine Kinases , Tuberous Sclerosis
9.
Sci Rep ; 9(1): 67, 2019 01 11.
Article in English | MEDLINE | ID: mdl-30635612

ABSTRACT

Obesity and type 2 diabetes are increasing in prevalence around the world, and there is a clear need for new and effective strategies to promote metabolic health. A low protein (LP) diet improves metabolic health in both rodents and humans, but the mechanisms that underlie this effect remain unknown. The gut microbiome has recently emerged as a potent regulator of host metabolism and the response to diet. Here, we demonstrate that a LP diet significantly alters the taxonomic composition of the gut microbiome at the phylum level, altering the relative abundance of Actinobacteria, Bacteroidetes, and Firmicutes. Transcriptional profiling suggested that any impact of the microbiome on liver metabolism was likely independent of the microbiome-farnesoid X receptor (FXR) axis. We therefore tested the ability of a LP diet to improve metabolic health following antibiotic ablation of the gut microbiota. We found that a LP diet promotes leanness, increases energy expenditure, and improves glycemic control equally well in mice treated with antibiotics as in untreated control animals. Our results demonstrate that the beneficial effects of a LP diet on glucose homeostasis, energy balance, and body composition are unlikely to be mediated by diet-induced changes in the taxonomic composition of the gut microbiome.


Subject(s)
Amino Acids/metabolism , Biota , Diet/methods , Intestines/microbiology , Metabolism , Animals , Gene Expression Profiling , Mice
10.
FASEB J ; 32(6): 3471-3482, 2018 06.
Article in English | MEDLINE | ID: mdl-29401631

ABSTRACT

Obesity and diabetes are major challenges to global health, and there is an urgent need for interventions that promote weight loss. Dietary restriction of methionine promotes leanness and improves metabolic health in mice and humans. However, poor long-term adherence to this diet limits its translational potential. In this study, we develop a short-term methionine deprivation (MD) regimen that preferentially reduces fat mass, restoring normal body weight and glycemic control to diet-induced obese mice of both sexes. The benefits of MD do not accrue from calorie restriction, but instead result from increased energy expenditure. MD promotes increased energy expenditure in a sex-specific manner, inducing the fibroblast growth factor (Fgf)-21-uncoupling protein (Ucp)-1 axis only in males. Methionine is an agonist of the protein kinase mechanistic target of rapamycin complex (mTORC)-1, which has been proposed to play a key role in the metabolic response to amino acid-restricted diets. In our study, we used a mouse model of constitutive hepatic mTORC1 activity and demonstrate that suppression of hepatic mTORC1 signaling is not required for the metabolic effects of MD. Our study sheds new light on the mechanisms by which dietary methionine regulates metabolic health and demonstrates the translational potential of MD for the treatment of obesity and type 2 diabetes.-Yu, D., Yang, S. E., Miller, B. R., Wisinski, J. A., Sherman, D. S., Brinkman, J. A., Tomasiewicz, J. L., Cummings, N. E., Kimple, M. E., Cryns, V. L., Lamming, D. W. Short-term methionine deprivation improves metabolic health via sexually dimorphic, mTORC1-independent mechanisms.


Subject(s)
Energy Metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Methionine/deficiency , Obesity/metabolism , Sex Characteristics , Animals , Caloric Restriction , Female , Fibroblast Growth Factors/metabolism , Male , Mice , Obesity/diet therapy , Obesity/pathology , Uncoupling Protein 1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...