Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Biol Chem ; 299(2): 102835, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36581203

RESUMO

Tetraspanins are transmembrane signaling and proinflammatory proteins. Prior work demonstrates that the tetraspanin, CD53/TSPAN25/MOX44, mediates B-cell development and lymphocyte migration to lymph nodes and is implicated in various inflammatory diseases. However, CD53 is also expressed in highly metabolic tissues, including adipose and liver; yet its function outside the lymphoid compartment is not defined. Here, we show that CD53 demarcates the nutritional and inflammatory status of hepatocytes. High-fat exposure and inflammatory stimuli induced CD53 in vivo in liver and isolated primary hepatocytes. In contrast, restricting hepatocyte glucose flux through hepatocyte glucose transporter 8 deletion or through trehalose treatment blocked CD53 induction in fat- and fructose-exposed contexts. Furthermore, germline CD53 deletion in vivo blocked Western diet-induced dyslipidemia and hepatic inflammatory transcriptomic activation. Surprisingly, metabolic protection in CD53 KO mice was more pronounced in the presence of an inciting inflammatory event. CD53 deletion attenuated tumor necrosis factor alpha-induced and fatty acid + lipopolysaccharide-induced cytokine gene expression and hepatocyte triglyceride accumulation in isolated murine hepatocytes. In vivo, CD53 deletion in nonalcoholic steatohepatitis diet-fed mice blocked peripheral adipose accumulation and adipose inflammation, insulin tolerance, and liver lipid accumulation. We then defined a stabilized and trehalase-resistant trehalose polymer that blocks hepatocyte CD53 expression in basal and over-fed contexts. The data suggest that CD53 integrates inflammatory and metabolic signals in response to hepatocyte nutritional status and that CD53 blockade may provide a means by which to attenuate pathophysiology in diseases that integrate overnutrition and inflammation, such as nonalcoholic steatohepatitis and type 2 diabetes.


Assuntos
Hepatócitos , Hepatopatia Gordurosa não Alcoólica , Tetraspanina 25 , Animais , Camundongos , Dieta Hiperlipídica , Hepatócitos/metabolismo , Inflamação/genética , Inflamação/metabolismo , Fígado/metabolismo , Camundongos Endogâmicos C57BL , Hepatopatia Gordurosa não Alcoólica/metabolismo , Obesidade/metabolismo , Tetraspanina 25/metabolismo , Tetraspaninas/genética , Tetraspaninas/metabolismo , Trealose/metabolismo
2.
Gastroenterology ; 158(5): 1402-1416.e2, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31838076

RESUMO

BACKGROUND & AIMS: Trehalose is a disaccharide that might be used in the treatment of cardiometabolic diseases. However, trehalose consumption promotes the expansion of Clostridioides difficile ribotypes that metabolize trehalose via trehalose-6-phosphate hydrolase. Furthermore, brush border and renal trehalases can reduce the efficacy of trehalose by cleaving it into monosaccharides. We investigated whether a trehalase-resistant analogue of trehalose (lactotrehalose) has the same metabolic effects of trehalose without expanding C difficile. METHODS: We performed studies with HEK293 and Caco2 cells, primary hepatocytes from mice, and human intestinal organoids. Glucose transporters were overexpressed in HEK293 cells, and glucose tra2nsport was quantified. Primary hepatocytes were cultured with or without trehalose or lactotrehalose, and gene expression patterns were analyzed. C57B6/J mice were given oral antibiotics and trehalose or lactotrehalose in drinking water, or only water (control), followed by gavage with the virulent C difficile ribotype 027 (CD027); fecal samples were analyzed for toxins A (ToxA) or B (ToxB) by enzyme-linked immunosorbent assay. Other mice were given trehalose or lactotrehalose in drinking water for 2 days before placement on a chow or 60% fructose diet for 10 days. Liver tissues were collected and analyzed by histologic, serum biochemical, RNA sequencing, autophagic flux, and thermogenesis analyses. We quantified portal trehalose and lactotrehalose bioavailability by gas chromatography mass spectrometry. Fecal microbiomes were analyzed by 16S ribosomal RNA sequencing and principal component analyses. RESULTS: Lactotrehalose and trehalose each blocked glucose transport in HEK293 cells and induced a gene expression pattern associated with fasting in primary hepatocytes. Compared with mice on the chow diet, mice on the high-fructose diet had increased circulating cholesterol, higher ratios of liver weight-to-body weight, hepatic lipid accumulation (steatosis), and liver gene expression patterns of carbohydrate-responsive de novo lipogenesis. Mice given lactotrehalose while on the high-fructose diet did not develop any of these features and had increased whole-body caloric expenditure compared with mice given trehalose or water and fed a high-fructose diet. Livers from mice given lactotrehalose had increased transcription of genes that regulate mitochondrial energy metabolism compared with liver from mice given trehalose or controls. Lactotrehalose was bioavailable in venous and portal circulation and fecal samples. Lactotrehalose reduced fecal markers of microbial branched-chain amino acid biosynthesis and increased expression of microbial genes that regulate insulin signaling. In mice given antibiotics followed by CD027, neither lactotrehalose nor trehalose increased levels of the bacteria or its toxin in stool-in fact, trehalose reduced the abundance of CD027 in stool. Lactotrehalose and trehalose reduced markers of inflammation in rectal tissue after CD027 infection. CONCLUSIONS: Lactotrehalose is a trehalase-resistant analogue that increases metabolic parameters, compared with trehalose, without increasing the abundance or virulence of C difficile strain CD027. Trehalase-resistant trehalose analogues might be developed as next-generation fasting-mimetics for the treatment of diabetes and nonalcoholic fatty liver disease.


Assuntos
Clostridioides difficile/isolamento & purificação , Infecções por Clostridium/prevenção & controle , Metabolismo Energético/efeitos dos fármacos , Trealose/farmacologia , Animais , Proteínas de Bactérias/metabolismo , Células CACO-2 , Clostridioides difficile/enzimologia , Infecções por Clostridium/diagnóstico , Infecções por Clostridium/microbiologia , Diabetes Mellitus/tratamento farmacológico , Diabetes Mellitus/metabolismo , Dissacaridases/metabolismo , Modelos Animais de Doenças , Jejum/metabolismo , Fezes/microbiologia , Glucose/metabolismo , Células HEK293 , Hepatócitos , Humanos , Mucosa Intestinal/citologia , Lipogênese/efeitos dos fármacos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Camundongos , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , Hepatopatia Gordurosa não Alcoólica/metabolismo , Cultura Primária de Células , Trealose/análogos & derivados , Trealose/uso terapêutico
3.
Am J Physiol Renal Physiol ; 316(5): F814-F822, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-30724105

RESUMO

Iron is a critical nutrient required by hosts and pathogens. Uropathogenic Escherichia coli (UPEC), the principal causative agent of urinary tract infections (UTIs), chelate iron for their survival and persistence. Here, we demonstrate that dietary modulation of iron availability limits UPEC burden in a mouse model of UTI. Mice on a low-iron diet exhibit reduced systemic and bladder mucosal iron availability and harbor significantly lower bacterial burden, concomitant with dampened inflammation. Hepcidin is a master regulator of iron that controls iron-dependent UPEC intracellular growth. Hepcidin-deficient mice ( Hamp1-/-) exhibit accumulation of iron deposits, persistent bacterial burden in the bladder, and a heightened inflammatory response to UTI. However, a low-iron dietary regimen reversed the iron overload and increased bacterial burden phenotypes in Hamp1-/- mice. Thus modulation of iron levels via diet can reduce UPEC infection and persistence, which may have significant implications for clinical management of UTI.


Assuntos
Infecções por Escherichia coli/dietoterapia , Ferro da Dieta/metabolismo , Bexiga Urinária/microbiologia , Infecções Urinárias/dietoterapia , Escherichia coli Uropatogênica/patogenicidade , Animais , Carga Bacteriana , Modelos Animais de Doenças , Infecções por Escherichia coli/metabolismo , Infecções por Escherichia coli/microbiologia , Ferritinas/metabolismo , Hepcidinas/genética , Hepcidinas/metabolismo , Interações Hospedeiro-Patógeno , Interleucina-6/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Bexiga Urinária/metabolismo , Infecções Urinárias/metabolismo , Infecções Urinárias/microbiologia
4.
Cell Mol Gastroenterol Hepatol ; 17(5): 801-820, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38280549

RESUMO

BACKGROUND & AIMS: Restoring hepatic and peripheral insulin sensitivity is critical to prevent or reverse metabolic syndrome and type 2 diabetes. Glucose homeostasis comprises in part the complex regulation of hepatic glucose production and insulin-mediated glucose uptake and oxidation in peripheral tissues. We previously identified hepatocyte arginase 2 (Arg2) as an inducible ureahydrolase that improves glucose homeostasis and enhances glucose oxidation in multiple obese, insulin-resistant models. We therefore examined structure-function determinants through which hepatocyte Arg2 governs systemic insulin action and glucose oxidation. METHODS: To do this, we generated mice expressing wild-type murine Arg2, enzymatically inactive Arg2 (Arg2H160F) and Arg2 lacking its putative mitochondrial targeting sequence (Arg2Δ1-22). We expressed these hepatocyte-specific constructs in obese, diabetic (db/db) mice and performed genetic complementation analyses in hepatocyte-specific Arg2-deficent (Arg2LKO) mice. RESULTS: We show that Arg2 attenuates hepatic steatosis, independent of mitochondrial localization or ureahydrolase activity, and that enzymatic arginase activity is dispensable for Arg2 to augment total body energy expenditure. In contrast, mitochondrial localization and ureahydrolase activity were required for Arg2-mediated reductions in fasting glucose and insulin resistance indices. Mechanistically, Arg2Δ1-22 and Arg2H160F failed to suppress glucose appearance during hyperinsulinemic-euglycemic clamping. Quantification of heavy-isotope-labeled glucose oxidation further revealed that mistargeting or ablating Arg2 enzymatic function abrogates Arg2-induced peripheral glucose oxidation. CONCLUSION: We conclude that the metabolic effects of Arg2 extend beyond its enzymatic activity, yet hepatocyte mitochondrial ureahydrolysis drives hepatic and peripheral oxidative metabolism. The data define a structure-based mechanism mediating hepatocyte Arg2 function and nominate hepatocyte mitochondrial ureahydrolysis as a key determinant of glucose oxidative capacity in mammals.


Assuntos
Arginase , Diabetes Mellitus Tipo 2 , Camundongos , Animais , Arginase/genética , Arginase/metabolismo , Glucose , Hepatócitos/metabolismo , Obesidade/metabolismo , Insulina , Mamíferos/metabolismo
5.
Cell Rep Med ; 3(1): 100498, 2022 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-35106510

RESUMO

Obesity is a multi-systemic disorder of energy balance. Despite intense investigation, the determinants of energy homeostasis remain incompletely understood, and efficacious treatments against obesity and its complications are lacking. Here, we demonstrate that conferred arginine iminohydrolysis by the bacterial virulence factor and arginine deiminase, arcA, promotes mammalian energy expenditure and insulin sensitivity and reverses dyslipidemia, hepatic steatosis, and inflammation in obese mice. Extending this, pharmacological arginine catabolism via pegylated arginine deiminase (ADI-PEG 20) recapitulates these metabolic effects in dietary and genetically obese models. These effects require hepatic and whole-body expression of the autophagy complex protein BECN1 and hepatocyte-specific FGF21 secretion. Single-cell ATAC sequencing further reveals BECN1-dependent hepatocyte chromatin accessibility changes in response to ADI-PEG 20. The data thus reveal an unexpected therapeutic utility for arginine catabolism in modulating energy metabolism by activating systemic autophagy, which is now exploitable through readily available pharmacotherapy.


Assuntos
Arginina/metabolismo , Autofagia , Metabolismo Energético , Hidrolases/química , Hidrolases/metabolismo , Polietilenoglicóis/química , Animais , Proteína Beclina-1/metabolismo , Dependovirus/metabolismo , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patologia , Dieta Ocidental , Dislipidemias/patologia , Fígado Gorduroso/patologia , Fatores de Crescimento de Fibroblastos/metabolismo , Glucose/metabolismo , Hepatócitos/metabolismo , Homeostase , Resistência à Insulina , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Termogênese
6.
Nat Commun ; 13(1): 1074, 2022 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-35228549

RESUMO

Calorie restriction abates aging and cardiometabolic disease by activating metabolic signaling pathways, including nicotinamide adenine dinucleotide (NAD+) biosynthesis and salvage. Nicotinamide phosphoribosyltransferase (NAMPT) is rate-limiting in NAD+ salvage, yet hepatocyte NAMPT actions during fasting and metabolic duress remain unclear. We demonstrate that hepatocyte NAMPT is upregulated in fasting mice, and in isolated hepatocytes subjected to nutrient withdrawal. Mice lacking hepatocyte NAMPT exhibit defective FGF21 activation and thermal regulation during fasting, and are sensitized to diet-induced glucose intolerance. Hepatocyte NAMPT overexpression induced FGF21 and adipose browning, improved glucose homeostasis, and attenuated dyslipidemia in obese mice. Hepatocyte SIRT1 deletion reversed hepatocyte NAMPT effects on dark-cycle thermogenesis, and hepatic FGF21 expression, but SIRT1 was dispensable for NAMPT insulin-sensitizing, anti-dyslipidemic, and light-cycle thermogenic effects. Hepatocyte NAMPT thus conveys key aspects of the fasting response, which selectively dissociate through hepatocyte SIRT1. Modulating hepatocyte NAD+ is thus a potential mechanism through which to attenuate fasting-responsive disease.


Assuntos
Nicotinamida Fosforribosiltransferase , Sirtuína 1 , Animais , Citocinas/metabolismo , Hepatócitos/metabolismo , Fígado/metabolismo , Camundongos , NAD/metabolismo , Nicotinamida Fosforribosiltransferase/genética , Nicotinamida Fosforribosiltransferase/metabolismo , Sirtuína 1/genética , Sirtuína 1/metabolismo
7.
Nat Commun ; 10(1): 1587, 2019 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-30962478

RESUMO

Caloric restriction and intermittent fasting are emerging therapeutic strategies against obesity, insulin resistance and their complications. However, the effectors that drive this response are not completely defined. Here we identify arginase 2 (Arg2) as a fasting-induced hepatocyte factor that protects against hepatic and peripheral fat accumulation, hepatic inflammatory responses, and insulin and glucose intolerance in obese murine models. Arg2 is upregulated in fasting conditions and upon treatment with the hepatocyte glucose transporter inhibitor trehalose. Hepatocyte-specific Arg2 overexpression enhances basal thermogenesis, and protects from weight gain, insulin resistance, glucose intolerance, hepatic steatosis and hepatic inflammation in diabetic mouse models. Arg2 suppresses expression of the regulator of G-protein signalling (RGS) 16, and genetic RGS16 reconstitution reverses the effects of Arg2 overexpression. We conclude that hepatocyte Arg2 is a critical effector of the hepatic glucose fasting response and define a therapeutic target to mitigate the complications of obesity and non-alcoholic fatty liver disease.


Assuntos
Arginase/metabolismo , Jejum/fisiologia , Fígado/metabolismo , Termogênese/fisiologia , Animais , Arginase/genética , Restrição Calórica , Colesterol/genética , Colesterol/metabolismo , Diabetes Mellitus Experimental/metabolismo , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Teste de Tolerância a Glucose , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Resistência à Insulina/fisiologia , Fígado/enzimologia , Masculino , Camundongos Endogâmicos C57BL , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/metabolismo , Proteínas RGS/genética , Proteínas RGS/metabolismo , Termogênese/genética , Trealose/farmacologia
8.
Autophagy ; 14(11): 1959-1975, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29996716

RESUMO

The macroautophagy/autophagy-inducing disaccharide, trehalose, has been proposed to be a promising therapeutic agent against neurodegenerative and cardiometabolic diseases. We recently showed that trehalose attenuates hepatic steatosis in part by blocking hepatocyte glucose transport to induce hepatocyte autophagic flux. However, although every major demonstration of trehalose action invokes activating autophagic flux as its primary function, the mechanism of action of trehalose in whole-body energy metabolism remains poorly defined. Here, we demonstrate that trehalose induces hepatocyte TFEB (transcription factor EB)-dependent thermogenesis in vivo, concomitant with upregulation of hepatic and white adipose expression of UCP1 (uncoupling protein 1 [mitochondrial, protein carrier]). Mechanistically, we provide evidence that hepatocyte fasting transcriptional and metabolic responses depend upon PPARGC1A (peroxisome proliferative activated receptor, gamma, coactivator 1 alpha), TFEB, and FGF21 (fibroblast growth factor 21) signaling. Strikingly, hepatocyte-selective TFEB knockdown abrogated trehalose induction of thermogenesis and white adipose tissue UCP1 upregulation in vivo. In contrast, we found that trehalose action on thermogenesis was independent of LEP (leptin) and the autophagy pathway, as there was robust thermogenic induction in trehalose-treated ob/ob, Becn1, Atg16l1, and Epg5 mutant mice. We conclude that trehalose induces metabolically favorable effects on whole-body thermogenesis in part via hepatocyte-centered fasting-like mechanisms that appear to be independent of autophagic flux. Our findings elucidate a novel mechanism by which trehalose acts as a metabolic therapeutic agent by activating hepatic fasting responses. More broadly, the hepatic glucose fasting response may be of clinical utility against overnutrition-driven disease, such as obesity and type 2 diabetes mellitus.


Assuntos
Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/fisiologia , Hepatócitos/efeitos dos fármacos , Termogênese/efeitos dos fármacos , Termogênese/genética , Trealose/farmacologia , Animais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Células Cultivadas , Metabolismo Energético/efeitos dos fármacos , Metabolismo Energético/genética , Proteínas Facilitadoras de Transporte de Glucose/antagonistas & inibidores , Proteínas Facilitadoras de Transporte de Glucose/metabolismo , Hepatócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética
9.
JCI Insight ; 3(16)2018 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-30135298

RESUMO

The hepatic glucose fasting response is gaining traction as a therapeutic pathway to enhance hepatic and whole-host metabolism. However, the mechanisms underlying these metabolic effects remain unclear. Here, we demonstrate the epidermal-type lipoxygenase, eLOX3 (encoded by its gene, Aloxe3), is a potentially novel effector of the therapeutic fasting response. We show that Aloxe3 is activated during fasting, glucose withdrawal, or trehalose/trehalose analogue treatment. Hepatocyte-specific Aloxe3 expression reduced weight gain and hepatic steatosis in diet-induced and genetically obese (db/db) mouse models. Aloxe3 expression, moreover, enhanced basal thermogenesis and abrogated insulin resistance in db/db diabetic mice. Targeted metabolomics demonstrated accumulation of the PPARγ ligand 12-KETE in hepatocytes overexpressing Aloxe3. Strikingly, PPARγ inhibition reversed hepatic Aloxe3-mediated insulin sensitization, suppression of hepatocellular ATP production and oxygen consumption, and gene induction of PPARγ coactivator-1α (PGC1α) expression. Moreover, hepatocyte-specific PPARγ deletion reversed the therapeutic effect of hepatic Aloxe3 expression on diet-induced insulin intolerance. Aloxe3 is, therefore, a potentially novel effector of the hepatocellular fasting response that leverages both PPARγ-mediated and pleiotropic effects to augment hepatic and whole-host metabolism, and it is, thus, a promising target to ameliorate metabolic disease.


Assuntos
Jejum/fisiologia , Insulina/metabolismo , Lipoxigenase/metabolismo , Síndrome Metabólica/metabolismo , PPAR gama/metabolismo , Animais , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Hepatócitos/metabolismo , Humanos , Lipoxigenase/genética , Fígado/citologia , Fígado/metabolismo , Síndrome Metabólica/dietoterapia , Síndrome Metabólica/etiologia , Camundongos , Camundongos Transgênicos , PPAR gama/genética
10.
Endocrinology ; 159(5): 2110-2126, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29596655

RESUMO

The adaptive fasting response is invoked as a promising cardiometabolic and neurodegenerative therapeutic pathway. We and others have defined the carbohydrate transporter glucose transporter 8 (GLUT8) as a critical regulator of hepatic and whole-organism metabolic homeostasis in the overfed and diabetic states. However, the functions of this critical transporter in the physiological fasting response remain poorly understood. Here, we tested the hypothesis that GLUT8 modulates the adaptive hepatic fasting response. We demonstrate that mice with targeted Slc2a8 disruption exhibit enhanced thermogenesis, ketogenesis, and peripheral lipid mobilization during fasting. These metabolic enhancements were observed in the context of mildly impaired hepatic mitochondrial oxidative metabolism in vivo and in vitro. Mechanistically, we show that hepatic peroxisome proliferator-activated receptor α (PPARα) and its transcriptional fasting response target hepatokine, fibroblast growth factor (FGF)21, are cell-autonomously hyperactivated in GLUT8-deficient liver and in isolated primary murine hepatocytes during nutrient depletion. Hepatic PPARα knockdown in GLUT8-deficient mice normalized the enhanced ketogenic and FGF21 secretory responses and decreased mitochondrial respiratory function without altering the hyperthermic response to fasting. Our data demonstrate that hepatocyte GLUT8 regulates adaptive fasting in part through regulation of the PPARα signaling cascade. Moreover, the ketotic and thermic responses to fasting are differentially encoded within the GLUT8-PPARα communication axis. GLUT8 therefore represents a therapeutic target that can be leveraged against cardiometabolic disease.


Assuntos
Jejum/metabolismo , Fatores de Crescimento de Fibroblastos/genética , Proteínas Facilitadoras de Transporte de Glucose/genética , Hepatócitos/metabolismo , Corpos Cetônicos/biossíntese , Fígado/metabolismo , Mitocôndrias Hepáticas/metabolismo , PPAR alfa/metabolismo , Animais , Técnicas de Silenciamento de Genes , Proteínas Facilitadoras de Transporte de Glucose/deficiência , Metabolismo dos Lipídeos/genética , Masculino , Camundongos , Transdução de Sinais , Termogênese/genética
11.
Sci Rep ; 6: 38586, 2016 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-27922102

RESUMO

Trehalose is a disaccharide demonstrated to mitigate disease burden in multiple murine neurodegenerative models. We recently revealed that trehalose rapidly induces hepatic autophagy and abrogates hepatic steatosis by inhibiting hexose transport via the SLC2A family of facilitative transporters. Prior studies, however, postulate that intracellular trehalose is sufficient to induce cellular autophagy. The objective of the current study was to identify the means by which trehalose accesses the hepatocyte cytoplasm, and define the distal signaling mechanisms by which trehalose induces autophagy. We provide gas chromatographic/mass spectrometric, fluorescence microscopic and radiolabeled uptake evidence that trehalose traverses the plasma membrane via SLC2A8 (GLUT8), a homolog of the trehalose transporter-1 (Tret1). Moreover, GLUT8-deficient hepatocytes and GLUT8-deficient mice exposed to trehalose resisted trehalose-induced AMP-activated protein kinase (AMPK) phosphorylation and autophagic induction in vitro and in vivo. Although trehalose profoundly attenuated mTORC1 signaling, trehalose-induced mTORC1 suppression was insufficient to activate autophagy in the absence of AMPK or GLUT8. Strikingly, transient, heterologous Tret1 overexpression reconstituted autophagic flux and AMPK signaling defects in GLUT8-deficient hepatocyte cultures. Together, these data suggest that cytoplasmic trehalose access is carrier-mediated, and that GLUT8 is a mammalian trehalose transporter required for hepatocyte trehalose-induced autophagy and signal transduction.


Assuntos
Autofagia , Proteínas Facilitadoras de Transporte de Glucose/metabolismo , Trealose/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Sequência de Aminoácidos , Animais , Autofagia/efeitos dos fármacos , Transporte Biológico , Linhagem Celular , Ácidos Graxos/metabolismo , Glucose/metabolismo , Proteínas Facilitadoras de Transporte de Glucose/química , Proteínas Facilitadoras de Transporte de Glucose/genética , Hepatócitos/metabolismo , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Camundongos , Camundongos Knockout , Modelos Biológicos , Modelos Moleculares , Conformação Molecular , Fosforilação , Ligação Proteica , Transdução de Sinais , Trealose/química , Trealose/farmacologia , Triglicerídeos/metabolismo
12.
Sci Signal ; 9(416): ra21, 2016 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-26905426

RESUMO

Trehalose is a naturally occurring disaccharide that has gained attention for its ability to induce cellular autophagy and mitigate diseases related to pathological protein aggregation. Despite decades of ubiquitous use as a nutraceutical, preservative, and humectant, its mechanism of action remains elusive. We showed that trehalose inhibited members of the SLC2A (also known as GLUT) family of glucose transporters. Trehalose-mediated inhibition of glucose transport induced AMPK (adenosine 5'-monophosphate-activated protein kinase)-dependent autophagy and regression of hepatic steatosis in vivo and a reduction in the accumulation of lipid droplets in primary murine hepatocyte cultures. Our data indicated that trehalose triggers beneficial cellular autophagy by inhibiting glucose transport.


Assuntos
Autofagia , Fígado Gorduroso/metabolismo , Proteínas Facilitadoras de Transporte de Glucose/metabolismo , Trealose/metabolismo , Proteínas Quinases Ativadas por AMP/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Fígado Gorduroso/genética , Fígado Gorduroso/patologia , Proteínas Facilitadoras de Transporte de Glucose/genética , Células HEK293 , Células Hep G2 , Humanos , Camundongos , Camundongos Knockout
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA