Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Am J Pathol ; 186(8): 2043-2054, 2016 08.
Article in English | MEDLINE | ID: mdl-27461362

ABSTRACT

Acute pancreatitis (AP) is a common and devastating gastrointestinal disorder that causes significant morbidity. The disease starts as local inflammation in the pancreas that may progress to systemic inflammation and complications. Protein tyrosine phosphatase 1B (PTP1B) is implicated in inflammatory signaling, but its significance in AP remains unclear. To investigate whether PTP1B may have a role in AP, we used pancreas PTP1B knockout (panc-PTP1B KO) mice and determined the effects of pancreatic PTP1B deficiency on cerulein- and arginine-induced acute pancreatitis. We report that PTP1B protein expression was increased in the early phase of AP in mice and rats. In addition, histological analyses of pancreas samples revealed enhanced features of AP in cerulein-treated panc-PTP1B KO mice compared with controls. Moreover, cerulein- and arginine-induced serum amylase and lipase were significantly higher in panc-PTP1B KO mice compared with controls. Similarly, pancreatic mRNA and serum concentrations of the inflammatory cytokines IL-1B, IL-6, and tumor necrosis factor-α were increased in panc-PTP1B KO mice compared with controls. Furthermore, panc-PTP1B KO mice exhibited enhanced cerulein- and arginine-induced NF-κB inflammatory response accompanied with increased mitogen-activated protein kinases activation and elevated endoplasmic reticulum stress. Notably, these effects were recapitulated in acinar cells treated with a pharmacological inhibitor of PTP1B. These findings reveal a novel role for pancreatic PTP1B in cerulein- and arginine-induced acute pancreatitis.


Subject(s)
Pancreatitis, Acute Necrotizing/metabolism , Pancreatitis, Acute Necrotizing/pathology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Animals , Disease Models, Animal , Male , Mice , Mice, Knockout , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction
2.
Biochim Biophys Acta Gen Subj ; 1861(11 Pt A): 2758-2765, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28757338

ABSTRACT

BACKGROUND: Diabetic nephropathy (DN) is the leading cause of renal failure, and podocyte dysfunction contributes to the pathogenesis of DN. Soluble epoxide hydrolase (sEH, encoded by Ephx2) is a conserved cytosolic enzyme whose inhibition has beneficial effects on renal function. The aim of this study is to investigate the contribution of sEH in podocytes to hyperglycemia-induced renal injury. MATERIALS AND METHODS: Mice with podocyte-specific sEH disruption (pod-sEHKO) were generated, and alterations in kidney function were determined under normoglycemia, and high-fat diet (HFD)- and streptozotocin (STZ)-induced hyperglycemia. RESULTS: sEH protein expression increased in murine kidneys under HFD- and STZ-induced hyperglycemia. sEH deficiency in podocytes preserved renal function and glucose control and mitigated hyperglycemia-induced renal injury. Also, podocyte sEH deficiency was associated with attenuated hyperglycemia-induced renal endoplasmic reticulum (ER) stress, inflammation and fibrosis, and enhanced autophagy. Moreover, these effects were recapitulated in immortalized murine podocytes treated with a selective sEH pharmacological inhibitor. Furthermore, pharmacological-induced elevation of ER stress or attenuation of autophagy in immortalized podocytes mitigated the protective effects of sEH inhibition. CONCLUSIONS: These findings establish sEH in podocytes as a significant contributor to renal function under hyperglycemia. GENERAL SIGNIFICANCE: These data suggest that sEH is a potential therapeutic target for podocytopathies.


Subject(s)
Diabetes Mellitus, Experimental/genetics , Diabetic Nephropathies/genetics , Epoxide Hydrolases/genetics , Hyperglycemia/genetics , Animals , Apoptosis/genetics , Autophagy/genetics , Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Experimental/pathology , Diabetic Nephropathies/enzymology , Diabetic Nephropathies/pathology , Endoplasmic Reticulum Stress/genetics , Enzyme Inhibitors/administration & dosage , Epoxide Hydrolases/antagonists & inhibitors , Humans , Hyperglycemia/enzymology , Hyperglycemia/pathology , Kidney/enzymology , Kidney/pathology , Mice , Podocytes/enzymology
3.
Biomed Pharmacother ; 175: 116590, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38653109

ABSTRACT

Alcohol-associated liver disease (ALD) is a leading factor of liver-related death worldwide. ALD has various manifestations that include steatosis, hepatitis, and cirrhosis and is currently without approved pharmacotherapies. The Src homology phosphatase 2 (Shp2) is a drug target in some cancers due to its positive regulation of Ras-mitogen-activated protein kinase signaling and cell proliferation. Shp2 pharmacological inhibition yields beneficial outcomes in animal disease models, but its impact on ALD remains unexplored. This study aims to investigate the effects of Shp2 inhibition and its validity using a preclinical mouse model of ALD. We report that the administration of SHP099, a potent and selective allosteric inhibitor of Shp2, partially ameliorated ethanol-induced hepatic injury, inflammation, and steatosis in mice. Additionally, Shp2 inhibition was associated with reduced ethanol-evoked activation of extracellular signal-regulated kinase (ERK), oxidative, and endoplasmic reticulum (ER) stress in the liver. Besides the liver, excessive alcohol consumption induces multi-organ injury and dysfunction, including the intestine. Notably, Shp2 inhibition diminished ethanol-induced intestinal inflammation and permeability, abrogated the reduction in tight junction protein expression, and the activation of ERK and stress signaling in the ileum. Collectively, Shp2 pharmacological inhibition mitigates the deleterious effects of ethanol in the liver and intestine in a mouse model of ALD. Given the multifactorial aspects underlying ALD pathogenesis, additional studies are needed to decipher the utility of Shp2 inhibition alone or as a component in a multitherapeutic regimen to combat this deadly malady.


Subject(s)
Disease Models, Animal , Ethanol , Liver Diseases, Alcoholic , Mice, Inbred C57BL , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Animals , Liver Diseases, Alcoholic/pathology , Liver Diseases, Alcoholic/prevention & control , Liver Diseases, Alcoholic/enzymology , Liver Diseases, Alcoholic/drug therapy , Mice , Male , Protein Tyrosine Phosphatase, Non-Receptor Type 11/antagonists & inhibitors , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Ethanol/toxicity , Liver/drug effects , Liver/pathology , Liver/enzymology , Liver/metabolism , Endoplasmic Reticulum Stress/drug effects , Oxidative Stress/drug effects
4.
Nat Metab ; 6(1): 61-77, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38195859

ABSTRACT

While pancreatic ß and α cells are considered the main drivers of blood glucose homeostasis through insulin and glucagon secretion, the contribution of δ cells and somatostatin (SST) secretion to glucose homeostasis remains unresolved. Here we provide a quantitative assessment of the physiological contribution of δ cells to the glycaemic set point in mice. Employing three orthogonal mouse models to remove SST signalling within the pancreas or transplanted islets, we demonstrate that ablating δ cells or SST leads to a sustained decrease in the glycaemic set point. This reduction coincides with a decreased glucose threshold for insulin response from ß cells, leading to increased insulin secretion to the same glucose challenge. Our data demonstrate that ß cells are sufficient to maintain stable glycaemia and reveal that the physiological role of δ cells is to provide tonic feedback inhibition that reduces the ß cell glucose threshold and consequently lowers the glycaemic set point in vivo.


Subject(s)
Islets of Langerhans , Somatostatin-Secreting Cells , Animals , Mice , Glucagon , Insulin , Glucose
5.
Environ Toxicol Pharmacol ; 98: 104070, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36682504

ABSTRACT

Given the paucity of research surrounding the effect of chronic paraquat on striatal neurotoxicity, there is a need for further investigation into the neurotoxic effects of paraquat in mouse striatum. Furthermore, while previous studies have shown that inhibiting soluble epoxide hydrolase mitigates MPTP-mediated endoplasmic reticulum stress in mouse striatum, its effect on paraquat toxicity is still unknown. Thus, this study attempts to observe changes in inflammatory and endoplasmic reticulum stress markers in mouse striatum following chronic paraquat administration to determine whether inhibiting soluble epoxide hydrolase mitigates paraquat-induced neurotoxicity and whether it can reduce TLR4-mediated inflammation in primary astrocytes and microglia. Our results show that while the pro-inflammatory effect of chronic paraquat is small, there is a significant induction of inflammatory and cellular stress markers, such as COX2 and CHOP, that can be mitigated through a prophylactic administration of a soluble epoxide hydrolase inhibitor.


Subject(s)
Neurotoxicity Syndromes , Paraquat , Mice , Animals , Epoxide Hydrolases , Rodentia , Inflammation
6.
J Nutr Sci Vitaminol (Tokyo) ; 68(4): 312-319, 2022.
Article in English | MEDLINE | ID: mdl-36047103

ABSTRACT

Tryptophan is an essential amino acid important as a protein building block, but it also serves as substrate for the generation of several bioactive compounds with important physiological roles. Furthermore, tryptophan has been reported to have a unique role as a nutritional signaling molecule that regulates protein synthesis in mouse and rat liver. In the present study, the acute effects of tryptophan on protein synthesis were confirmed and compared with those of leucine in rats. Eighteen hours fasted rats were orally administered of tryptophan or leucine at a dose of 135 mg/100 g body weight by gavage and then sacrificed 1 h after administration. The effects of tryptophan and leucine on the rate of protein synthesis were evaluated by the surface sensing of translation (SUnSET) method. We also examined the ability of tryptophan to induce activation of the mTOR pathway by measuring phosphorylation of 4E-BP1 and S6K1. Oral administration of tryptophan led to a stimulation of the rate of protein synthesis concomitant with activation of mTOR pathway in the liver, but not in skeletal muscle. We also investigated the sensitivity of liver protein synthesis to tryptophan administration. The half-maximal effective doses (ED50) of tryptophan in stimulating 4E-BP1 and S6K1 phosphorylation were both about 60% of daily intake. The effect of tryptophan on hepatic protein synthesis was similar to that of leucine on muscle protein synthesis, and the sensitivity of liver protein synthesis to tryptophan administration appeared to be almost the same or slightly lower than that of muscle protein synthesis to leucine administration.


Subject(s)
Intracellular Signaling Peptides and Proteins , Tryptophan , Animals , Carrier Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/pharmacology , Leucine/pharmacology , Liver/metabolism , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Phosphoproteins/metabolism , Phosphoproteins/pharmacology , Phosphorylation , Protein Biosynthesis , Rats , TOR Serine-Threonine Kinases/metabolism , Tryptophan/metabolism , Tryptophan/pharmacology
7.
Free Radic Biol Med ; 172: 48-57, 2021 08 20.
Article in English | MEDLINE | ID: mdl-34038767

ABSTRACT

Pancreatic ß-cells are crucial regulators of systemic glucose homeostasis, and their dysfunction and loss are central features in type 2 diabetes. Interventions that rectify ß-cell dysfunction and loss are essential to combat this deadly malady. In the current study, we sought to delineate the role of soluble epoxide hydrolase (sEH) in ß-cells under diet-induced metabolic stress. The expression of sEH was upregulated in murine and macaque diabetes models and islets of diabetic human patients. We postulated that hyperglycemia-induced elevation in sEH leads to a reduction in its substrates, epoxyeicosatrienoic acids (EETs), and attenuates the function of ß-cells. Genetic deficiency of sEH potentiated glucose-stimulated insulin secretion in mice, likely in a cell-autonomous manner, contributing to better systemic glucose control. Consistent with this observation, genetic and pharmacological inactivation of sEH and the treatment with EETs exhibited insulinotropic effects in isolated murine islets ex vivo. Additionally, sEH deficiency enhanced glucose sensing and metabolism with elevated ATP and cAMP concentrations. This phenotype was associated with attenuated oxidative stress and diminished ß-cell death in sEH deficient islets. Moreover, pharmacological inhibition of sEH in vivo mitigated, albeit partly, high fat diet-induced ß-cell loss and dedifferentiation. The current observations provide new insights into the role of sEH in ß-cells and information that may be leveraged for the development of a mechanism-based intervention to rectify ß-cell dysfunction and loss.


Subject(s)
Diabetes Mellitus, Type 2 , Hyperglycemia , Animals , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/genetics , Diet, High-Fat/adverse effects , Epoxide Hydrolases/genetics , Humans , Mice , Mice, Inbred C57BL , Pancreas
8.
Cell Mol Gastroenterol Hepatol ; 11(3): 815-830, 2021.
Article in English | MEDLINE | ID: mdl-33068774

ABSTRACT

BACKGROUND & AIMS: Alcohol-associated liver disease (ALD) is a significant cause of liver-related morbidity and mortality worldwide and with limited therapies. Soluble epoxide hydrolase (sEH; Ephx2) is a largely cytosolic enzyme that is highly expressed in the liver and is implicated in hepatic function, but its role in ALD is mostly unexplored. METHODS: To decipher the role of hepatic sEH in ALD, we generated mice with liver-specific sEH disruption (Alb-Cre; Ephx2fl/fl). Alb-Cre; Ephx2fl/fl and control (Ephx2fl/fl) mice were subjected to an ethanol challenge using the chronic plus binge model of ALD and hepatic injury, inflammation, and steatosis were evaluated under pair-fed and ethanol-fed states. In addition, we investigated the capacity of pharmacologic inhibition of sEH in the chronic plus binge mouse model. RESULTS: We observed an increase of hepatic sEH in mice upon ethanol consumption, suggesting that dysregulated hepatic sEH expression might be involved in ALD. Alb-Cre; Ephx2fl/fl mice presented efficient deletion of hepatic sEH with corresponding attenuation in sEH activity and alteration in the lipid epoxide/diol ratio. Consistently, hepatic sEH deficiency ameliorated ethanol-induced hepatic injury, inflammation, and steatosis. In addition, targeted metabolomics identified lipid mediators that were impacted significantly by hepatic sEH deficiency. Moreover, hepatic sEH deficiency was associated with a significant attenuation of ethanol-induced hepatic endoplasmic reticulum and oxidative stress. Notably, pharmacologic inhibition of sEH recapitulated the effects of hepatic sEH deficiency and abrogated injury, inflammation, and steatosis caused by ethanol feeding. CONCLUSIONS: These findings elucidated a role for sEH in ALD and validated a pharmacologic inhibitor of this enzyme in a preclinical mouse model as a potential therapeutic approach.


Subject(s)
Epoxide Hydrolases/metabolism , Ethanol/toxicity , Liver Diseases, Alcoholic/etiology , Liver/pathology , Phenylurea Compounds/therapeutic use , Piperidines/therapeutic use , Animals , Disease Models, Animal , Drug Evaluation, Preclinical , Epoxide Hydrolases/antagonists & inhibitors , Epoxide Hydrolases/genetics , Ethanol/administration & dosage , Female , Gene Expression Regulation/drug effects , Liver/enzymology , Liver/immunology , Liver Diseases, Alcoholic/drug therapy , Liver Diseases, Alcoholic/pathology , Mice , Mice, Transgenic , Phenylurea Compounds/pharmacology , Piperidines/pharmacology
9.
Nutrients ; 12(9)2020 Sep 01.
Article in English | MEDLINE | ID: mdl-32882842

ABSTRACT

Tryptophan has a unique role as a nutritional signaling molecule that regulates protein synthesis in mouse and rat liver. However, the mechanism underlying the stimulating actions of tryptophan on hepatic protein synthesis remains unclear. Proteomic and metabolomic analyses were performed to identify candidate proteins and metabolites likely to play a role in the stimulation of protein synthesis by tryptophan. Overnight-fasted rats were orally administered L-tryptophan and then sacrificed 1 or 3 h after administration. Four differentially expressed protein spots were detected in rat liver at 3 h after tryptophan administration, of which one was identified as an ornithine aminotransferase (OAT) precursor. OAT is the main catabolic enzyme for ornithine, and its expression was significantly decreased by tryptophan administration. The concentration of ornithine was increased in the liver at 3 h after tryptophan administration. Ornithine is a precursor for polyamine biosynthesis. Significantly increased concentrations of polyamines were found in the liver at 3 h after administration of tryptophan. Additionally, enhanced hepatic protein synthesis was demonstrated by oral administration of putrescine. We speculate that the increase in ornithine level through suppression of OAT expression by tryptophan administration may lead to accelerated polyamine synthesis, thereby promoting protein synthesis in the liver.


Subject(s)
Polyamines/metabolism , Protein Biosynthesis/drug effects , Tryptophan/pharmacology , Animals , Liver/metabolism , Metabolomics , Ornithine/metabolism , Ornithine-Oxo-Acid Transaminase/drug effects , Proteomics , Rats
10.
Redox Biol ; 36: 101658, 2020 09.
Article in English | MEDLINE | ID: mdl-32769011

ABSTRACT

Alcoholic liver disease (ALD) is a major health problem and a significant cause of liver-related death. Currently, the mainstay for ALD therapy is alcohol abstinence highlighting the need to develop pharmacotherapeutic approaches. Protein-tyrosine phosphatase 1B (PTP1B) is an established regulator of hepatic functions, but its role in ALD is mostly unexplored. In this study, we used mice with liver-specific PTP1B disruption as well as pharmacological inhibition to investigate the in vivo function of this phosphatase in ALD. We report upregulation of hepatic PTP1B in the chronic plus binge mouse model and, importantly, in liver biopsies of alcoholic hepatitis patients. Also, mice with hepatic PTP1B disruption attenuated ethanol-induced injury, inflammation, and steatosis compared with ethanol-fed control animals. Moreover, PTP1B deficiency was associated with decreased ethanol-induced oxidative stress in vivo and ex vivo. Further, pharmacological modulation of oxidative balance in hepatocytes identified diminished oxidative stress as a contributor to the salutary effects of PTP1B deficiency. Notably, PTP1B pharmacological inhibition elicited beneficial effects and mitigated hepatic injury, inflammation, and steatosis caused by ethanol feeding. In summary, these findings causally link hepatic PTP1B and ALD and define a potential therapeutic target for the management of this disease.


Subject(s)
Ethanol , Liver Diseases, Alcoholic , Animals , Ethanol/metabolism , Ethanol/toxicity , Hepatocytes , Humans , Liver/metabolism , Liver Diseases, Alcoholic/drug therapy , Liver Diseases, Alcoholic/genetics , Liver Diseases, Alcoholic/metabolism , Mice , Mice, Inbred C57BL , Oxidative Stress
11.
Metabolism ; 76: 56-69, 2017 11.
Article in English | MEDLINE | ID: mdl-28987240

ABSTRACT

OBJECTIVE: Diabetic nephropathy is one of the most devastating complications of diabetes, and growing evidence implicates podocyte dysfunction in disease pathogenesis. The objective of this study was to investigate the contribution of protein tyrosine phosphatase 1B (PTP1B) in podocytes to hyperglycemia-induced renal injury. METHODS: To determine the in vivo function of PTP1B in podocytes we generated mice with podocyte-specific PTP1B disruption (hereafter termed pod-PTP1B KO). Kidney functions were determined in control and pod-PTP1B KO mice under normoglycemia and high-fat diet (HFD)- and streptozotocin (STZ)-induced hyperglycemia. RESULTS: PTP1B expression increased in murine kidneys following HFD and STZ challenges. Under normoglycemia control and pod-PTP1B KO mice exhibited comparable renal functions. However, podocyte PTP1B disruption attenuated hyperglycemia-induced albuminuria and renal injury and preserved glucose control. Also, podocyte PTP1B disruption was accompanied with improved renal insulin signaling and enhanced autophagy with decreased inflammation and fibrosis. Moreover, the beneficial effects of podocyte PTP1B disruption in vivo were recapitulated in E11 murine podocytes with lentiviral-mediated PTP1B knockdown. Reconstitution of PTP1B in knockdown podocytes reversed the enhanced insulin signaling and autophagy suggesting that they were likely a consequence of PTP1B deficiency. Further, pharmacological attenuation of autophagy in PTP1B knockdown podocytes mitigated the protective effects of PTP1B deficiency. CONCLUSIONS: These findings demonstrate that podocyte PTP1B deficiency attenuates hyperglycemia-induced renal damage and suggest that PTP1B may present a therapeutic target in renal injury.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Diabetic Nephropathies/metabolism , Hyperglycemia/metabolism , Podocytes/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Animals , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Diabetic Nephropathies/genetics , Diabetic Nephropathies/pathology , Diet, High-Fat , Hyperglycemia/genetics , Hyperglycemia/pathology , Kidney/metabolism , Kidney/pathology , Mice , Mice, Knockout , Podocytes/pathology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Signal Transduction/physiology
12.
FEBS J ; 284(13): 1970-1986, 2017 07.
Article in English | MEDLINE | ID: mdl-28485854

ABSTRACT

Podocytes play an important role in maintaining glomerular function, and podocyte injury is a significant component in the pathogenesis of proteinuria. Soluble epoxide hydrolase (sEH) is a cytosolic enzyme whose genetic deficiency and pharmacological inhibition have beneficial effects on renal function, but its role in podocytes remains unexplored. The objective of this study was to investigate the contribution of sEH in podocytes to lipopolysaccharide (LPS)-induced kidney injury. We report increased sEH transcript and protein expression in murine podocytes upon LPS challenge. To determine the function of sEH in podocytes in vivo we generated podocyte-specific sEH-deficient (pod-sEHKO) mice. Following LPS challenge, podocyte sEH-deficient mice exhibited lower kidney injury, proteinuria, and blood urea nitrogen concentrations than controls suggestive of preserved renal function. Also, renal mRNA and serum concentrations of inflammatory cytokines IL-6, IL-1ß, and TNFα were significantly lower in LPS-treated pod-sEHKO than control mice. Moreover, podocyte sEH deficiency was associated with decreased LPS-induced NF-κB and MAPK activation and attenuated endoplasmic reticulum stress. Furthermore, the protective effects of podocyte sEH deficiency in vivo were recapitulated in E11 murine podocytes treated with a selective sEH pharmacological inhibitor. Altogether, these findings identify sEH in podocytes as a contributor to signaling events in acute renal injury and suggest that sEH inhibition may be of therapeutic value in proteinuria. ENZYMES: Soluble epoxide hydrolase: EC 3.3.2.10.


Subject(s)
Acute Kidney Injury/enzymology , Epoxide Hydrolases/deficiency , Podocytes/enzymology , Proteinuria/enzymology , Acute Kidney Injury/genetics , Animals , Blood Urea Nitrogen , Cells, Cultured , Cytokines/blood , Cytokines/genetics , Cytokines/metabolism , Epoxide Hydrolases/antagonists & inhibitors , Epoxide Hydrolases/genetics , Gene Expression Regulation/drug effects , Immunoblotting , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Lipopolysaccharides/pharmacology , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Fluorescence , Phenylurea Compounds/pharmacology , Piperidines/pharmacology , Proteinuria/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Solubility
13.
Cell Metab ; 26(3): 539-546.e5, 2017 Sep 05.
Article in English | MEDLINE | ID: mdl-28877457

ABSTRACT

Calorie restriction, without malnutrition, has been shown to increase lifespan and is associated with a shift away from glycolysis toward beta-oxidation. The objective of this study was to mimic this metabolic shift using low-carbohydrate diets and to determine the influence of these diets on longevity and healthspan in mice. C57BL/6 mice were assigned to a ketogenic, low-carbohydrate, or control diet at 12 months of age and were either allowed to live their natural lifespan or tested for physiological function after 1 or 14 months of dietary intervention. The ketogenic diet (KD) significantly increased median lifespan and survival compared to controls. In aged mice, only those consuming a KD displayed preservation of physiological function. The KD increased protein acetylation levels and regulated mTORC1 signaling in a tissue-dependent manner. This study demonstrates that a KD extends longevity and healthspan in mice.


Subject(s)
Diet, Ketogenic , Health , Longevity/physiology , Acetylation , Adaptation, Physiological , Animals , Diet, Carbohydrate-Restricted , Male , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice, Inbred C57BL , Organ Specificity , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL