Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
ACS Nano ; 18(27): 17969-17986, 2024 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-38920100

RESUMEN

Obesity is becoming a worldwide pandemic. Interfacial engineering of food lipid is expected to inhibit diet-induced obesity without damage to the eating enjoyment brought by high-fat diets. Unfortunately, this strategy has not been achieved yet. After screening different plant proteins, bromelain and papain were found to form wormlike and long-straight protein fibrils, respectively. The conversion of long-straight amyloid-like fibrils to wormlike fibrils was demonstrated in the fibrillation of bromelain. Using oil-in-water high internal phase emulsions (HIPEs) as a proof of concept, bromelain fibrils showed dramatically stronger interfacial stabilization capabilities than papain fibrils with high application potentials in the real-world formulation of high-fat food products such as mayonnaise. Compared with papain fibrils, oral administration of HIPEs stabilized by bromelain fibrils resulted in substantially higher fecal lipid contents and significantly decreased expression levels of the genes related to lipid absorption and transport in the intestine, including CD36, FATP-2, FATP-4, and APOA-4, without a difference in intervening gut microbiota. Consequently, dramatically less lipid absorption in the small intestine, markedly smaller chylomicron particles in the plasma, lower serum triglycerides, and controlled energy and lipid metabolism, as well as the inhibition of adipose expansion and overweight, were observed in the group with gavage of HIPEs stabilized by the bromelain fibrils rather than the papain fibrils. Furthermore, with the same calorie, substitution of all the fat in the standard high-fat feed of mice with the HIPEs emulsified by the bromelain fibrils showed a significantly stronger effect than the ones prepared by the papain fibrils on preventing high-fat-diet (HFD)-induced obesity including alleviation of adipose expansion and inflammation as well as fatty liver, also via inhibiting the absorption and transport of lipid in the intestine. The effect is ascribed to the suppressed lipolysis caused by a more compact and elastic interfacial layer formed by the wormlike fibrils than that of the long-straight fibrils, which are resistant to gastric environments and replacement by bile acids in digestion. Therefore, we provide an appealing and general strategy for controlling obesity by reducing the supply of free fatty acids (FAs) for absorption in the enteric lumen through protein fibril polymorphisms at the interface.


Asunto(s)
Obesidad , Papaína , Animales , Obesidad/metabolismo , Ratones , Papaína/metabolismo , Papaína/química , Bromelaínas/farmacología , Bromelaínas/química , Bromelaínas/metabolismo , Ratones Endogámicos C57BL , Masculino , Dieta Alta en Grasa , Emulsiones/química , Tejido Adiposo/metabolismo , Tejido Adiposo/efectos de los fármacos , Metabolismo de los Lípidos/efectos de los fármacos
2.
Proc Natl Acad Sci U S A ; 120(23): e2219419120, 2023 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-37252972

RESUMEN

Prolyl hydroxylase domain (PHD) enzymes change HIF activity according to oxygen signal; whether it is regulated by other physiological conditions remains largely unknown. Here, we report that PHD3 is induced by fasting and regulates hepatic gluconeogenesis through interaction and hydroxylation of CRTC2. Pro129 and Pro615 hydroxylation of CRTC2 following PHD3 activation is necessary for its association with cAMP-response element binding protein (CREB) and nuclear translocation, and enhanced binding to promoters of gluconeogenic genes by fasting or forskolin. CRTC2 hydroxylation-stimulated gluconeogenic gene expression is independent of SIK-mediated phosphorylation of CRTC2. Liver-specific knockout of PHD3 (PHD3 LKO) or prolyl hydroxylase-deficient knockin mice (PHD3 KI) show attenuated fasting gluconeogenic genes, glycemia, and hepatic capacity to produce glucose during fasting or fed with high-fat, high-sucrose diet. Importantly, Pro615 hydroxylation of CRTC2 by PHD3 is increased in livers of fasted mice, diet-induced insulin resistance or genetically obese ob/ob mice, and humans with diabetes. These findings increase our understanding of molecular mechanisms linking protein hydroxylation to gluconeogenesis and may offer therapeutic potential for treating excessive gluconeogenesis, hyperglycemia, and type 2 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2 , Glucosa , Humanos , Ratones , Animales , Glucosa/metabolismo , Prolina/metabolismo , Hidroxilación , Diabetes Mellitus Tipo 2/metabolismo , Hígado/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Gluconeogénesis/fisiología , Prolil Hidroxilasas/metabolismo , Hepatocitos/metabolismo , Ratones Endogámicos C57BL
3.
Hepatology ; 78(5): 1492-1505, 2023 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-36680394

RESUMEN

BACKGROUND AND AIMS: NASH has emerged as a leading cause of chronic liver disease. However, the mechanisms that govern NASH fibrosis remain largely unknown. CREBZF is a CREB/ATF bZIP transcription factor that causes hepatic steatosis and metabolic defects in obesity. APPROACH AND RESULTS: Here, we show that CREBZF is a key mechanism of liver fibrosis checkpoint that promotes hepatocyte injury and exacerbates diet-induced NASH in mice. CREBZF deficiency attenuated liver injury, fibrosis, and inflammation in diet-induced mouse models of NASH. CREBZF increases HSC activation and fibrosis in a hepatocyte-autonomous manner by stimulating an extracellular matrix protein osteopontin, a key regulator of fibrosis. The inhibition of miR-6964-3p mediates CREBZF-induced production and secretion of osteopontin in hepatocytes. Adeno-associated virus -mediated rescue of osteopontin restored HSC activation, liver fibrosis, and NASH progression in CREBZF-deficient mice. Importantly, expression levels of CREBZF are increased in livers of diet-induced NASH mouse models and humans with NASH. CONCLUSIONS: Osteopontin signaling by CREBZF represents a previously unrecognized intrahepatic mechanism that triggers liver fibrosis and contributes to the severity of NASH.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico , Osteopontina , Animales , Humanos , Ratones , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Modelos Animales de Enfermedad , Hígado Graso/genética , Hígado Graso/metabolismo , Fibrosis , Hepatocitos/metabolismo , Hepatocitos/patología , Hígado/metabolismo , Hígado/patología , Cirrosis Hepática/patología , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , Osteopontina/genética , Osteopontina/metabolismo
4.
Cell Mol Gastroenterol Hepatol ; 12(3): 857-871, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33989817

RESUMEN

BACKGROUND AND AIMS: Butyric acid is an intestinal microbiota-produced short-chain fatty acid, which exerts salutary effects on alleviating nonalcoholic fatty liver disease (NAFLD). However, the underlying mechanism of butyrate on regulating hepatic lipid metabolism is largely unexplored. METHODS: A mouse model of NAFLD was induced with high-fat diet feeding, and sodium butyrate (NaB) intervention was initiated at the eighth week and lasted for 8 weeks. Hepatic steatosis was evaluated and metabolic pathways concerning lipid homeostasis were analyzed. RESULTS: Here, we report that administration of NaB by gavage once daily for 8 weeks causes an augmentation of insulin-induced gene (Insig) activity and inhibition of lipogenic gene in mice fed with high-fat diet. Mechanistically, NaB is sufficient to enhance the interaction between Insig and its upstream kinase AMP-activated protein kinase (AMPK). The stimulatory effects of NaB on Insig-1 activity are abolished in AMPKα1/α2 double knockout (AMPK-/-) mouse primary hepatocytes. Moreover, AMPK activation by NaB is mediated by LKB1, as evidenced by the observations showing NaB-mediated induction of phosphorylation of AMPK, and its downstream target acetyl-CoA carboxylase is diminished in LKB1-/- mouse embryonic fibroblasts. CONCLUSIONS: These studies indicate that NaB serves as a negative regulator of hepatic lipogenesis in NAFLD and that NaB attenuates hepatic steatosis and improves lipid profile and liver function largely through the activation of LKB1-AMPK-Insig signaling pathway. Therefore, NaB has therapeutic potential for treating NAFLD and related metabolic diseases.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Ácido Butírico/farmacología , Suplementos Dietéticos , Regulación de la Expresión Génica , Insulina/metabolismo , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Animales , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Insulina/farmacología , Metabolismo de los Lípidos/efectos de los fármacos , Lipogénesis/efectos de los fármacos , Lipogénesis/genética , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratones , Modelos Biológicos , Enfermedad del Hígado Graso no Alcohólico/patología , Fosforilación
5.
EBioMedicine ; 57: 102849, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32580141

RESUMEN

BACKGROUND: Nonalcoholic fatty liver disease (NAFLD) is associated with altered production of secreted proteins. Increased understanding of secreted proteins could lead to improved prediction and treatment of NAFLD. Here, we aimed to discover novel secreted proteins in humans that are associated with hepatic fat content using unbiased proteomic profiling strategy, and how the identified Thbs1 modulates lipid metabolism and hepatic steatosis. METHOD: NAFLD patients were enrolled and treated with lifestyle intervention. Patients who underwent liver biopsy were enrolled for analyzing the correlation between circulating Thbs1 and liver steatosis. Mice were fed on high-fat, high-sucrose diet and treated with recombinant Thbs1. Primary hepatocytes isolated from CD36 knockout (CD36-/-) mice and their wild-type littermates (controls) were treated with glucose plus insulin for 24 h together with or without recombinant Thbs1. FINDING: Serum Thbs1 levels are increased in participants with NAFLD and positively associated with liver steatosis grades. Improvement of liver steatosis after lifestyle intervention was accompanied with significant reduction of serum Thbs1 levels. Pharmacological administration of recombinant human Thbs1 attenuates hepatic steatosis in diet-induced obese mice. Treatment with Thbs1 protein or stably overexpression of Thbs1 causes a significant reduction of lipid accumulation in primary hepatocytes or HepG2 cells exposed to high glucose plus insulin, suggesting that Thbs1 regulates lipid metabolism in a hepatocyte-autonomous manner. Mechanistically, Thbs1 inhibits cleavage and processing of SREBP-1, leading to a reduction of target lipogenic gene expression and hepatic steatosis. Inhibitory effects of Thbs1 on lipogenesis and triglyceride accumulation are abrogated in CD36 deficient primary hepatocytes exposed to high glucose plus insulin. Interestingly, beneficial effects of Thbs1 on lipid accumulation are observed in primary hepatocytes treated with a Thbs1 nonapeptide mimetic ABT-526. INTERPRETATION: Thbs1 is a biomarker for NAFLD in humans, and pharmacological and genetic approaches for the modulation of Thbs1 activity may have the therapeutic potential for treating hepatic steatosis. FUND: A full list of funding bodies that contributed to this study can be found in the Funding Sources section.


Asunto(s)
Hígado Graso/genética , Metabolismo de los Lípidos/genética , Enfermedad del Hígado Graso no Alcohólico/genética , Proteómica , Trombospondina 1/genética , Animales , Antígenos CD36/genética , Dieta Alta en Grasa/efectos adversos , Hígado Graso/dietoterapia , Hígado Graso/metabolismo , Hígado Graso/patología , Células Hep G2 , Hepatocitos/metabolismo , Humanos , Insulina/genética , Insulina/metabolismo , Resistencia a la Insulina/genética , Lipogénesis/genética , Hígado/metabolismo , Hígado/patología , Ratones , Ratones Noqueados , Enfermedad del Hígado Graso no Alcohólico/dietoterapia , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , Fragmentos de Péptidos/farmacología , Trombospondina 1/farmacología , Triglicéridos/sangre
6.
Diabetes ; 69(8): 1611-1623, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32354858

RESUMEN

Nonalcoholic steatohepatitis has emerged as a major cause of liver diseases with no effective therapies. Here, we evaluate the efficacies and pharmacokinetics of B1344, a long-acting polyethylene glycolylated (PEGylated) fibroblast growth factor 21 analog, in a nongenetically modified nonhuman primate species that underwent liver biopsy and demonstrate the potential for efficacies in humans. B1344 is sufficient to selectively activate signaling from the ßKlotho/FGFR1c receptor complex. In cynomolgus monkeys with nonalcoholic fatty liver disease (NAFLD), administration of B1344 via subcutaneous injection for 11 weeks caused a profound reduction of hepatic steatosis, inflammation, and fibrosis, along with amelioration of liver injury and hepatocyte death, as evidenced by liver biopsy specimen and biochemical analysis. Moreover, improvement of metabolic parameters was observed in the monkeys, including reduction of body weight and improvement of lipid profiles and glycemic control. To determine the role of B1344 in the progression of murine NAFLD independent of obesity, B1344 was administered to mice fed a methionine- and choline-deficient diet. Consistently, B1344 administration prevented the mice from lipotoxicity damage and nonalcoholic steatohepatitis in a dose-dependent manner. These results provide preclinical validation for an innovative therapeutic approach to NAFLD and support further clinical testing of B1344 for treating nonalcoholic steatohepatitis and other metabolic diseases in humans.


Asunto(s)
Factores de Crecimiento de Fibroblastos/farmacocinética , Factores de Crecimiento de Fibroblastos/uso terapéutico , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Animales , Peso Corporal/efectos de los fármacos , Línea Celular , Colina , Fibrosis/sangre , Fibrosis/tratamiento farmacológico , Inflamación/sangre , Inflamación/tratamiento farmacológico , Hígado/efectos de los fármacos , Hígado/metabolismo , Macaca fascicularis , Masculino , Metionina , Ratones , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/sangre , Primates , Ratas , Transducción de Señal/efectos de los fármacos
7.
Hepatology ; 71(4): 1421-1436, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31469186

RESUMEN

BACKGROUND AND AIMS: STAT3, a member of the signal transducer and activator of transcription (STAT) family, is strongly associated with liver injury, inflammation, regeneration, and hepatocellular carcinoma development. However, the signals that regulate STAT3 activity are not completely understood. APPROACH AND RESULTS: Here we characterize CREB/ATF bZIP transcription factor CREBZF as a critical regulator of STAT3 in the hepatocyte to repress liver regeneration. We show that CREBZF deficiency stimulates the expression of the cyclin gene family and enhances liver regeneration after partial hepatectomy. Flow cytometry analysis reveals that CREBZF regulates cell cycle progression during liver regeneration in a hepatocyte-autonomous manner. Similar results were observed in another model of liver regeneration induced by intraperitoneal injection of carbon tetrachloride (CCl4 ). Mechanistically, CREBZF potently associates with the linker domain of STAT3 and represses its dimerization and transcriptional activity in vivo and in vitro. Importantly, hepatectomy-induced hyperactivation of cyclin D1 and liver regeneration in CREBZF liver-specific knockout mice was reversed by selective STAT3 inhibitor cucurbitacin I. In contrast, adeno-associated virus-mediated overexpression of CREBZF in the liver inhibits the expression of the cyclin gene family and attenuates liver regeneration in CCl4 -treated mice. CONCLUSIONS: These results characterize CREBZF as a coregulator of STAT3 to inhibit regenerative capacity, which may represent an essential cellular signal to maintain liver mass homeostasis. Therapeutic approaches to inhibit CREBZF may benefit the compromised liver during liver transplantation.


Asunto(s)
Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Regulación de la Expresión Génica , Regeneración Hepática/genética , Hígado/fisiología , Factor de Transcripción STAT3/genética , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Tetracloruro de Carbono/toxicidad , Ciclo Celular/genética , Eliminación de Gen , Hepatocitos/efectos de los fármacos , Hepatocitos/fisiología , Hígado/efectos de los fármacos , Hígado/lesiones , Redes y Vías Metabólicas , Ratones , Ratones Noqueados
8.
Exp Mol Med ; 51(9): 1-14, 2019 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-31506421

RESUMEN

Microbial metabolites have emerged as critical components that mediate the metabolic effects of the gut microbiota. Here, we show that indole-3-propionic acid (IPA), a tryptophan metabolite produced by gut bacteria, is a potent anti-non-alcoholic steatohepatitis (NASH) microbial metabolite. Here, we demonstrate that administration of IPA modulates the microbiota composition in the gut and inhibits microbial dysbiosis in rats fed a high-fat diet. IPA induces the expression of tight junction proteins, such as ZO-1 and Occludin, and maintains intestinal epithelium homeostasis, leading to a reduction in plasma endotoxin levels. Interestingly, IPA inhibits NF-κB signaling and reduces the levels of proinflammatory cytokines, such as TNFα, IL-1ß, and IL-6, in response to endotoxin in macrophages to repress hepatic inflammation and liver injury. Moreover, IPA is sufficient to inhibit the expression of fibrogenic and collagen genes and attenuate diet-induced NASH phenotypes. The beneficial effects of IPA on the liver are likely mediated through inhibiting the production of endotoxin in the gut. These findings suggest a protective role of IPA in the control of metabolism and uncover the gut microbiome and liver cross-talk in regulating the intestinal microenvironment and liver pathology via a novel dietary nutrient metabolite. IPA may provide a new therapeutic strategy for treating NASH.


Asunto(s)
Microbioma Gastrointestinal/efectos de los fármacos , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Ocludina/genética , Propionatos/farmacología , Proteína de la Zonula Occludens-1/genética , Animales , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Disbiosis/tratamiento farmacológico , Disbiosis/genética , Disbiosis/metabolismo , Disbiosis/microbiología , Endotoxinas/metabolismo , Microbioma Gastrointestinal/genética , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Indoles/farmacología , Interleucina-1beta , Interleucina-6/genética , Hígado/efectos de los fármacos , Hígado/lesiones , Hígado/patología , Macrófagos/efectos de los fármacos , FN-kappa B/genética , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/microbiología , Ratas , Triptófano/metabolismo , Factor de Necrosis Tumoral alfa/genética
9.
Diabetes ; 68(6): 1130-1142, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30862681

RESUMEN

Obesity and related inflammation are critical for the pathogenesis of insulin resistance, but the underlying mechanisms are not fully understood. Formyl peptide receptor 2 (FPR2) plays important roles in host immune responses and inflammation-related diseases. We found that Fpr2 expression was elevated in the white adipose tissue of high-fat diet (HFD)-induced obese mice and db/db mice. The systemic deletion of Fpr2 alleviated HFD-induced obesity, insulin resistance, hyperglycemia, hyperlipidemia, and hepatic steatosis. Furthermore, Fpr2 deletion in HFD-fed mice elevated body temperature, reduced fat mass, and inhibited inflammation by reducing macrophage infiltration and M1 polarization in metabolic tissues. Bone marrow transplantations between wild-type and Fpr2-/- mice and myeloid-specific Fpr2 deletion demonstrated that Fpr2-expressing myeloid cells exacerbated HFD-induced obesity, insulin resistance, glucose/lipid metabolic disturbances, and inflammation. Mechanistic studies revealed that Fpr2 deletion in HFD-fed mice enhanced energy expenditure probably through increasing thermogenesis in skeletal muscle; serum amyloid A3 and other factors secreted by adipocytes induced macrophage chemotaxis via Fpr2; and Fpr2 deletion suppressed macrophage chemotaxis and lipopolysaccharide-, palmitate-, and interferon-γ-induced macrophage M1 polarization through blocking their signals. Altogether, our studies demonstrate that myeloid Fpr2 plays critical roles in obesity and related metabolic disorders via regulating muscle energy expenditure, macrophage chemotaxis, and M1 polarization.


Asunto(s)
Quimiotaxis/genética , Dieta Alta en Grasa , Resistencia a la Insulina/genética , Macrófagos/inmunología , Receptores de Formil Péptido/genética , Animales , Temperatura Corporal/genética , Metabolismo Energético/genética , Hígado Graso/genética , Hígado Graso/inmunología , Hiperglucemia/genética , Hiperglucemia/inmunología , Hiperlipidemias/genética , Hiperlipidemias/inmunología , Inflamación/genética , Inflamación/inmunología , Resistencia a la Insulina/inmunología , Ratones , Ratones Noqueados , Ratones Obesos , Proteína Amiloide A Sérica/metabolismo , Termogénesis/genética
10.
Nat Commun ; 10(1): 623, 2019 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-30733434

RESUMEN

Insulin-induced gene (Insig) negatively regulates SREBP-mediated de novo fatty acid synthesis in the liver. However, the upstream regulation of Insig is incompletely understood. Here we report that AMPK interacts with and mediates phosphorylation of Insig. Thr222 phosphorylation following AMPK activation is required for protein stabilization of Insig-1, inhibition of cleavage and processing of SREBP-1, and lipogenic gene expression in response to metformin or A769662. AMPK-dependent phosphorylation ablates Insig's interaction with E3 ubiquitin ligase gp78 and represses its ubiquitination and degradation, whereas AMPK deficiency shows opposite effects. Interestingly, activation of AMPK by metformin causes an augmentation of Insig stability and reduction of lipogenic gene expression, and leads to the attenuation of hepatic steatosis in HFHS diet-fed mice. Moreover, hepatic overexpression of Insig-1 rescues hepatic steatosis in liver-specific AMPKα2 knockout mice fed with HFHS diet. These findings uncover a novel effector of AMPK. Targeting Insig may have the therapeutic potential for treating fatty liver disease and related disorders.


Asunto(s)
Regulación de la Expresión Génica , Lipogénesis/genética , Animales , Compuestos de Bifenilo , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Lipogénesis/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Metformina/farmacología , Ratones , Ratones Noqueados , Fosforilación/efectos de los fármacos , Pironas/farmacología , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Tiofenos/farmacología , Ubiquitinación/efectos de los fármacos
11.
J Biol Chem ; 294(3): 772-782, 2019 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-30429217

RESUMEN

Autophagy is of key importance for eliminating aggregated proteins during the maintenance of cellular proteostasis in response to endoplasmic reticulum (ER) stress. However, the upstream signaling that mediates autophagy activation in response to ER stress is incompletely understood. In this study, in vivo and in vitro approaches were utilized that include gain- and loss-of-function assays and mouse livers and human cell lines with tunicamycin-induced pharmacological ER stress. We report that calreticulin, a quality control chaperone that binds to misfolded glycoproteins for refolding in the ER, is induced under ER stress. Calreticulin overexpression stimulated the formation of autophagosomes and increased autophagic flux. Interestingly, calreticulin was sufficient for attenuating ER stress in tunicamycin- or thapsigargin-treated HeLa cells, whereas lentivirus-mediated shRNA calreticulin knockdown exacerbated ER stress. Mechanistically, we noted that calreticulin induces autophagy by interacting with microtubule-associated protein 1A/1B-light chain 3 (LC3). Confocal microscopy revealed that the colocalization of calreticulin and LC3 at the autophagosome was enhanced under ER stress conditions. Importantly, a conserved LC3-interacting region was necessary for calreticulin-mediated stimulation of autophagy and for reducing ER stress. These findings indicate a calreticulin-based mechanism that couples ER stress to autophagy activation, which, in turn, attenuates cellular stress, likely by alleviating the formation of aberrantly folded proteins. Pharmacological or genetic approaches that activate calreticulin-autophagy signaling may have potential for managing ER stress and related cellular disorders.


Asunto(s)
Autofagosomas/metabolismo , Autofagia , Calreticulina/metabolismo , Estrés del Retículo Endoplásmico , Retículo Endoplásmico/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Calreticulina/genética , Retículo Endoplásmico/genética , Células HEK293 , Células HeLa , Humanos , Proteínas Asociadas a Microtúbulos/genética
12.
Autophagy ; 14(10): 1779-1795, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30044707

RESUMEN

Normal metabolism requires a controlled balance between anabolism and catabolism. It is not completely known how this balance can be retained when the level of nutrient supply changes in the long term. We found that in murine liver anabolism, as represented by the phosphorylation of RPS6KB (ribosomal protein S6 kinase), was soon elevated while catabolism, as represented by TFEB (transcription factor EB)-directed gene transcription and lysosomal activities, was downregulated after the administration of a high-fat diet (HFD). Surprisingly, neither the alteration in RPS6KB phosphorylation nor that in TFEB functions was static over the long course of HFD feeding. Instead, the 2 signals exhibited dynamic alterations in opposite directions, which could be explained by the dependence of MTORC1 (MTOR complex 1) activation on TFEB-supported lysosome function and the feedback suppression of TFEB by MTORC1. Disruption of the dynamics by enforced expression of TFEB in HFD-fed mice at the peaks of MTORC1 activation restored lysosome function. Consistently, interference of MTORC1 activation with rapamycin or with a constitutively activated RRAGA mutant at the peak or nadir of MTORC1 oscillation enhanced or reduced the lysosome function, respectively. These treatments also improved or exacerbated hepatic steatosis and liver injury, respectively. Finally, there was a significant inverse correlation between TFEB activation and steatosis severity in the livers of patients with non-alcohol fatty liver diseases, supporting the clinical relevance of TFEB-regulated events. Thus, maintaining catabolic function through feedback mechanisms during enhanced anabolism, which is caused by nutrient oversupply, is important for reducing liver pathology.


Asunto(s)
Autofagia , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Hígado Graso/patología , Retroalimentación Fisiológica , Hígado/patología , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Nutrientes , Transducción de Señal , Animales , Núcleo Celular/metabolismo , Dieta Alta en Grasa , Hígado Graso/metabolismo , Lípidos/química , Lisosomas/metabolismo , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación/genética , Enfermedad del Hígado Graso no Alcohólico/patología , Factores de Tiempo
13.
Hepatology ; 68(4): 1361-1375, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29637572

RESUMEN

Insulin is critical for the regulation of de novo fatty acid synthesis, which converts glucose to lipid in the liver. However, how insulin signals are transduced into the cell and then regulate lipogenesis remains to be fully understood. Here, we identified CREB/ATF bZIP transcription factor (CREBZF) of the activating transcription factor/cAMP response element-binding protein (ATF/CREB) gene family as a key regulator for lipogenesis through insulin-Akt signaling. Insulin-induced gene 2a (Insig-2a) decreases during refeeding, allowing sterol regulatory element binding protein 1c to be processed to promote lipogenesis; but the mechanism of reduction is unknown. We show that Insig-2a inhibition is mediated by insulin-induced CREBZF. CREBZF directly inhibits transcription of Insig-2a through association with activating transcription factor 4. Liver-specific knockout of CREBZF causes an induction of Insig-2a and Insig-1 and resulted in repressed lipogenic program in the liver of mice during refeeding or upon treatment with streptozotocin and insulin. Moreover, hepatic CREBZF deficiency attenuates hepatic steatosis in high-fat, high-sucrose diet-fed mice. Importantly, expression levels of CREBZF are increased in livers of diet-induced insulin resistance or genetically obese ob/ob mice and humans with hepatic steatosis, which may underscore the potential role of CREBZF in the development of sustained lipogenesis in the liver under selective insulin resistance conditions. CONCLUSION: These findings uncover an unexpected mechanism that couples changes in extracellular hormonal signals to hepatic lipid homeostasis; disrupting CREBZF function may have the therapeutic potential for treating fatty liver disease and insulin resistance. (Hepatology 2018).


Asunto(s)
Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Hígado Graso/patología , Regulación de la Expresión Génica , Resistencia a la Insulina/genética , Lipogénesis/genética , Análisis de Varianza , Animales , Biopsia con Aguja , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Hígado Graso/genética , Hepatocitos/metabolismo , Hepatocitos/patología , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Distribución Aleatoria , Transducción de Señal
14.
Br J Pharmacol ; 175(2): 374-387, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29065221

RESUMEN

BACKGROUND AND PURPOSE: Berberine, a compound from rhizome coptidis, is traditionally used to treat gastrointestinal infections, such as bacterial diarrhoea. Recently, berberine was shown to have hypoglycaemic and hypolipidaemic effects. We investigated the mechanisms by which berberine regulates hepatic lipid metabolism and energy expenditure in mice. EXPERIMENTAL APPROACH: Liver-specific SIRT1 knockout mice and their wild-type littermates were fed a high-fat, high-sucrose (HFHS) diet and treated with berberine by i.p. injection for five weeks. Mouse primary hepatocytes and human HepG2 cells were treated with berberine and then subjected to immunoblotting analysis and Oil Red O staining. KEY RESULTS: Berberine attenuated hepatic steatosis and controlled energy balance in mice by inducing autophagy and FGF21. These beneficial effects of berberine on autophagy and hepatic steatosis were abolished by a deficiency of the nutrient sensor SIRT1 in the liver of HFHS diet-fed obese mice and in mouse primary hepatocytes. SIRT1 is essential for berberine to potentiate autophagy and inhibit lipid storage in mouse livers in response to fasting. Mechanistically, the berberine stimulates SIRT1 deacetylation activity and induces autophagy in an autophagy protein 5-dependent manner. Moreover, the administration of berberine was shown to promote hepatic gene expression and circulating levels of FGF21 and ketone bodies in mice in a SIRT1-dependent manner. CONCLUSIONS AND IMPLICATIONS: Berberine acts in the liver to regulate lipid utilization and maintain whole-body energy metabolism by mediating autophagy and FGF21 activation. Hence, it has therapeutic potential for treating metabolic defects under nutritional overload, such as fatty liver diseases, type 2 diabetes and obesity.


Asunto(s)
Autofagia/efectos de los fármacos , Berberina/farmacología , Berberina/uso terapéutico , Metabolismo Energético/efectos de los fármacos , Hígado Graso/tratamiento farmacológico , Factores de Crecimiento de Fibroblastos/biosíntesis , Sirtuina 1/fisiología , Animales , Autofagia/fisiología , Dieta de Carga de Carbohidratos , Dieta Alta en Grasa , Hígado Graso/fisiopatología , Factores de Crecimiento de Fibroblastos/sangre , Expresión Génica/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Cuerpos Cetónicos/sangre , Masculino , Ratones , Ratones Noqueados , Sirtuina 1/genética
15.
J Agric Food Chem ; 64(17): 3422-9, 2016 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-27064900

RESUMEN

Modification of chitosan (CS) through grafting with caffeic acid (CA, CA-g-CS) and ferulic acid (FA, FA-g-CS) significantly improved its solubility under neutral and alkaline environments. Spherical and physicochemically stable nanocomplexes assembled from the phenolic acid grafting CS and caseinophosphopeptide (CPP) were obtained with particle size <300 nm and zeta potential of <+30 mV. The net polymer nanocomplexes composed with the phenolic acid grafting CS and CPP showed strong antioxidant activity. The encapsulation efficiencies of epigallocatechin-3-gallate (EGCG) in the CA-g-CS-CPP nanocomplexes and FA-g-CS-CPP nanocomplexes were 88.1 ± 6.7 and 90.4 ± 4.2%, respectively. Improved delivery properties of EGCG were achieved after loading with the antioxidant nanocomplexes, including controlling release of EGCG under simulated gastric environments and preventing its degradation under neutral and alkaline environments.


Asunto(s)
Antioxidantes/administración & dosificación , Catequina/análogos & derivados , Nanopartículas/química , Antioxidantes/química , Catequina/administración & dosificación , Microscopía Electrónica de Transmisión , Espectrofotometría Ultravioleta , Espectroscopía Infrarroja por Transformada de Fourier
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...