Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Metabolism ; 155: 155909, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38582490

RESUMEN

BACKGROUND: Krüppel-like factor 10 (KLF10), a zinc finger transcription factor, plays a pivotal role in modulating TGF-ß-mediated cellular processes such as growth, apoptosis, and differentiation. Recent studies have implicated KLF10 in regulating lipid metabolism and glucose homeostasis. This study aimed to elucidate the precise role of hepatic KLF10 in developing metabolic dysfunction-associated steatohepatitis (MASH) in diet-induced obese mice. METHODS: We investigated hepatic KLF10 expression under metabolic stress and the effects of overexpression or ablation of hepatic KLF10 on MASH development and lipidemia. We also determined whether hepatocyte nuclear factor 4α (HNF4α) mediated the metabolic effects of KLF10. RESULTS: Hepatic KLF10 was downregulated in MASH patients and genetically or diet-induced obese mice. AAV8-mediated overexpression of KLF10 in hepatocytes prevented Western diet-induced hypercholesterolemia and steatohepatitis, whereas inactivation of hepatocyte KLF10 aggravated Western diet-induced steatohepatitis. Mechanistically, KLF10 reduced hepatic triglyceride and free fatty acid levels by inducing lipolysis and fatty acid oxidation and inhibiting lipogenesis, and reducing hepatic cholesterol levels by promoting bile acid synthesis. KLF10 highly induced HNF4α expression by directly binding to its promoter. The beneficial effect of KLF10 on MASH development was abolished in mice lacking hepatocyte HNF4α. In addition, the inactivation of KLF10 in hepatic stellate cells exacerbated Western diet-induced liver fibrosis by activating the TGF-ß/SMAD2/3 pathway. CONCLUSIONS: Our data collectively suggest that the transcription factor KLF10 plays a hepatoprotective role in MASH development by inducing HNF4α. Targeting hepatic KLF10 may offer a promising strategy for treating MASH.


Asunto(s)
Factores de Transcripción de la Respuesta de Crecimiento Precoz , Hígado Graso , Factor Nuclear 4 del Hepatocito , Factores de Transcripción de Tipo Kruppel , Animales , Factor Nuclear 4 del Hepatocito/metabolismo , Factor Nuclear 4 del Hepatocito/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Ratones , Humanos , Masculino , Factores de Transcripción de la Respuesta de Crecimiento Precoz/metabolismo , Factores de Transcripción de la Respuesta de Crecimiento Precoz/genética , Hígado Graso/metabolismo , Hígado Graso/etiología , Ratones Endogámicos C57BL , Metabolismo de los Lípidos , Hígado/metabolismo , Hepatocitos/metabolismo , Ratones Noqueados
2.
Dev Cell ; 58(21): 2326-2337.e5, 2023 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-37863040

RESUMEN

High-density lipoprotein (HDL) metabolism is regulated by complex interplay between the scavenger receptor group B type 1 (SR-BI) and multiple signaling molecules in the liver. Here, we show that lipocalin-2 (Lcn2) is a key regulator of hepatic SR-BI, HDL metabolism, and atherosclerosis. Overexpression of human Lcn2 in hepatocytes attenuates the development of atherosclerosis via SR-BI in western-diet-fed Ldlr-/- mice, whereas hepatocyte-specific ablation of Lcn2 has the opposite effect. Mechanistically, hepatocyte Lcn2 improves HDL metabolism and alleviates atherogenesis by blocking Nedd4-1-mediated SR-BI ubiquitination at K500 and K508. The Lcn2-improved HDL metabolism is abolished in mice with hepatocyte-specific Nedd4-1 or SR-BI deletion and in SR-BI (K500A/K508A) mutation mice. This study identifies a regulatory axis from Lcn2 to HDL via blocking Nedd4-1-mediated SR-BI ubiquitination and demonstrates that hepatocyte Lcn2 may be a promising target to improve HDL metabolism to treat atherosclerotic cardiovascular diseases.


Asunto(s)
Aterosclerosis , Lipoproteínas HDL , Ratones , Humanos , Animales , Lipoproteínas HDL/metabolismo , Lipocalina 2/genética , Lipocalina 2/metabolismo , Hepatocitos/metabolismo , Aterosclerosis/genética , Aterosclerosis/metabolismo , Hígado/metabolismo , Antígenos CD36/metabolismo
3.
Cells ; 12(15)2023 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-37566087

RESUMEN

Histone deacetylase Sirtuin 6 (SIRT6) regulates many biological processes. SIRT6 is known to regulate hepatic lipid metabolism and inhibit the development of nonalcoholic fatty liver disease (NAFLD). We aimed to investigate the role of hepatocyte SIRT6 in the development of atherosclerosis and further characterize the mechanism underlying SIRT6's effect on NAFLD. Ldlr-/- mice overexpressing or lacking hepatocyte SIRT6 were fed a Western diet for 16 weeks. The role of hepatic SIRT6 in the development of nonalcoholic steatohepatitis (NASH), atherosclerosis, and obesity was investigated. We also investigated whether p53 participates in the pathogenesis of NAFLD in mice overexpressing hepatic SIRT6. Our data show that loss of hepatocyte SIRT6 aggravated the development of NAFLD, atherosclerosis, and obesity in Ldlr-/- mice, whereas adeno-associated virus (AAV)-mediated overexpression of human SIRT6 in the liver had opposite effects. Mechanistically, hepatocyte SIRT6 likely inhibited the development of NAFLD by inhibiting lipogenesis, lipid droplet formation, and p53 signaling. Hepatocyte SIRT6 also likely inhibited the development of atherosclerosis by inhibiting intestinal lipid absorption and hepatic VLDL secretion. Hepatic SIRT6 also increased energy expenditure. In conclusion, our data indicate that hepatocyte SIRT6 protects against atherosclerosis, NAFLD, and obesity by regulating lipid metabolism in the liver and intestine.


Asunto(s)
Aterosclerosis , Enfermedad del Hígado Graso no Alcohólico , Sirtuinas , Humanos , Animales , Ratones , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Hepatocitos/metabolismo , Obesidad/complicaciones , Sirtuinas/genética , Sirtuinas/metabolismo , Lípidos , Homeostasis , Aterosclerosis/metabolismo
4.
J Agric Food Chem ; 71(28): 10629-10637, 2023 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-37418694

RESUMEN

4-Hydroxy-2-nonenal (4-HNE) is a secondary cytotoxic product generated from lipid peroxidation of polyunsaturated fatty acids (PUFAs). The accumulation of 4-HNE can covalently modify biomolecules, such as DNA and proteins, leading to various pathological conditions. Apple phloretin has been shown to be able to trap 4-HNE in vitro, but the trapping mechanisms of 4-HNE by phloretin are not fully understood. Moreover, whether the in vitro trapping efficacy of phloretin toward 4-HNE could be transferred into in vivo environments has never been investigated. In the present study, we observed the formation of 4-HNE conjugates of phloretin increased as phloretin decreased during the in vitro incubation. We then purified and characterized three mono-4-HNE-conjugates of phloretin using NMR and LC-MS/MS techniques. We thereafter demonstrated that apple phloretin could scavenge in vivo 4-HNE via the formation of at least three mono-4-HNE-conjugates of phloretin in a dose-dependent manner in mice after oral administration of three doses of phloretin (25, 100, and 400 mg/kg). The findings from this study pave the way to understanding how dihydrochalcones could act as effective scavengers of 4-HNE by working as sacrificial nucleophiles in vivo, thereby preventing or reducing the risk of 4-HNE-associated chronic diseases.


Asunto(s)
Malus , Floretina , Ratones , Animales , Peroxidación de Lípido , Malus/metabolismo , Cromatografía Liquida , Espectrometría de Masas en Tándem , Aldehídos/química
5.
Mol Nutr Food Res ; 67(8): e2200709, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36823693

RESUMEN

SCOPE: Methylglyoxal (MGO), a harmful reactive dicarbonyl, is involved in the pathogenesis and development of diabetes and diabetic complications. The goal of this study is to determine whether bioactive phenolamides in barley, p-coumaroylagmatine (pCAA) and feruloylagmatine (FAA), which share a similar guanidine group to diabetic drug metformin, have the capacity to detoxify MGO. METHODS AND RESULTS: In this study, the MGO-trapping abilities of these two phenolamides both in vitro and in mice are evaluated. It is found that in vitro anti-MGO capacities of pCAA and FAA are comparable to that of metformin, and both phenolamides could rapidly scavenge MGO via forming mono- and di-MGO adducts validated by in-house synthesized standards and interpretation of respective LC-MSn (n = 2-3) data. Furthermore, mono-MGO conjugates of phenolamides are detected from feces and urine of mice after oral administration of the corresponding phenolamides. CONCLUSION: These findings suggest that barley phenolamides may have the potentials to be developed as alternative therapeutics to prevent the development of MGO-associated diabetes and diabetic complications.


Asunto(s)
Complicaciones de la Diabetes , Diabetes Mellitus , Hordeum , Animales , Ratones , Piruvaldehído , Productos Finales de Glicación Avanzada
6.
Cells ; 11(20)2022 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-36291054

RESUMEN

Retinoic acid signaling plays an important role in regulating lipid metabolism and inflammation. However, the role of retinoic acid receptor alpha (RARα) in atherosclerosis remains to be determined. In the current study, we investigated the role of macrophage RARα in the development of atherosclerosis. Macrophages isolated from myeloid-specific Rarα-/- (RarαMac-/-) mice showed increased lipid accumulation and inflammation and reduced cholesterol efflux compared to Rarαfl/fl (control) mice. All-trans retinoic acid (AtRA) induced ATP-binding cassette subfamily A member 1 (Abca1) and Abcg1 expression and cholesterol efflux in both RarαMac-/- mice and Rarαfl/fl mice. In Ldlr-/- mice, myeloid ablation of RARα significantly reduced macrophage Abca1 and Abcg1 expression and cholesterol efflux, induced inflammatory genes, and aggravated Western diet-induced atherosclerosis. Our data demonstrate that macrophage RARα protects against atherosclerosis, likely via inducing cholesterol efflux and inhibiting inflammation.


Asunto(s)
Aterosclerosis , Colesterol , Dieta Occidental , Macrófagos , Receptor alfa de Ácido Retinoico , Animales , Ratones , Aterosclerosis/etiología , Aterosclerosis/genética , Aterosclerosis/metabolismo , Colesterol/metabolismo , Inflamación/genética , Macrófagos/metabolismo , Receptor alfa de Ácido Retinoico/genética , Receptor alfa de Ácido Retinoico/metabolismo , Tretinoina/farmacología , Tretinoina/metabolismo , Dieta Occidental/efectos adversos , Transportador 1 de Casete de Unión a ATP/genética , Transportador 1 de Casete de Unión a ATP/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1/genética , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1/metabolismo , Ratones Noqueados
7.
J Agric Food Chem ; 70(31): 9674-9683, 2022 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-35916113

RESUMEN

6-Gingerol and 6-shogaol are the most abundant gingerols and shogaols in ginger root and have been shown to reduce the asthmatic phenotype in murine models of asthma. Several studies have described the pharmacokinetics of gingerols and shogaols in humans following the oral ingestion of ginger, while little was known about the metabolism of these components in humans, particularly in patients with asthma. In this study, a dietary supplement of 1.0 g of ginger root extract was administered to asthma patients twice daily for 56 days and serum samples were drawn at 0.5-8 h on days 0, 28, and 56. The metabolic profiles of gingerols and shogaols in human plasma and the kinetic changes of gingerols, shogaols, and their metabolites in asthma patients collected on the three different visits were analyzed using liquid chromatography-mass spectrometry (LC-MS). Ketone reduction was the major metabolic pathway of both gingerols and shogaols. Gingerdiols were identified as the major metabolites of 6-, 8-, and 10-gingerols. M11 and M9 were identified as the double-bond reduction and both the double-bond and ketone reduction metabolites of 6-shogaol, respectively. Cysteine conjugation was another major metabolic pathway of 6-shogaol in asthma patients, and two cysteine-conjugated 6-shogaol, M1 and M2, were identified as the major metabolites of 6-shogaol. Furthermore, gingerols, shogaols, and their metabolites were quantitated in the human serum collected at different time points during each of the three visits using a very sensitive high-resolution LC-MS method. The results showed that one-third of 6-gingerol was metabolized to produce its reduction metabolites, 6-gingerdiols, and more than 90% of 6-shogaol was metabolized to its phase I and cysteine-conjugated metabolites, suggesting the importance of considering the contribution of these metabolites to the bioavailability and health beneficial effects of gingerols and shogaols. All gingerols, shogaols, and their metabolites reached their peak concentrations in less than 2 h, and their half-lives (t1/2) were from 0.6 to 2.4 h. Furthermore, long-term treatment of ginger supplements, especially after 56 days of treatment, increases the absorption of ginger compounds and their metabolites in asthma patients.


Asunto(s)
Asma , Zingiber officinale , Animales , Asma/tratamiento farmacológico , Catecoles/química , Cisteína/metabolismo , Alcoholes Grasos/química , Zingiber officinale/química , Humanos , Cetonas/metabolismo , Ratones , Extractos Vegetales/química
8.
Hepatol Commun ; 6(10): 2665-2675, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35852305

RESUMEN

All-trans retinoic acid (AtRA) is an active metabolite of vitamin A that influences many biological processes in development, differentiation, and metabolism. AtRA functions through activation of retinoid acid receptors (RARs). AtRA is shown to ameliorate hepatic steatosis, but the underlying mechanism is not well understood. In this study, we investigated the role of hepatocyte RAR alpha (RARα) in mediating the effect of AtRA on hepatosteatosis in mice. Hepatocyte-specific Rarα-/- (L-Rarα-/- ) mice and their control mice were fed a chow diet, high-fat diet (HFD), or a high-fat/cholesterol/fructose (HFCF) diet. Some of the mice were also treated with AtRA. Loss of hepatocyte RARα-induced hepatosteatosis in chow-fed aged mice and HFD-fed mice. AtRA prevented and reversed HFCF diet-induced obesity and hepatosteatosis in the control mice but not in L-Rarα-/- mice. Furthermore, AtRA reduced hepatocyte fatty acid uptake and lipid droplet formation, dependent on hepatocyte RARα. Our data suggest that hepatocyte RARα plays an important role in preventing hepatosteatosis and mediates AtRA's effects on diet-induced hepatosteatosis.


Asunto(s)
Receptores de Ácido Retinoico , Vitamina A , Animales , Dieta , Ácidos Grasos , Fructosa , Ratones , Receptores de Ácido Retinoico/genética , Receptor alfa de Ácido Retinoico/genética , Tretinoina/farmacología
9.
Mol Nutr Food Res ; 66(9): e2101107, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35194934

RESUMEN

SCOPE: Acrolein (ACR), a lipid peroxidation product, pathologically participates in various chronic diseases. In vitro evidence suggestes that dietary dihydrochalcones (DHCs) potentiate safe and alternative therapeutics to synthetic pharmaceuticals for ACR scavenging. Here, to investigate whether ingested DHCs could trap ACR and thereof result in reductions in endogenous ACR in mice is aimed. METHODS AND RESULTS: Three doses of phloretin (25, 100, and 400 mg kg-1 ), a major dietary DHC, are orally administrated to mice and 24 h urine and fecal samples are collected, respectively. High-resolution MS-based targeted metabolomics reveal for the first time that phloretin and its oxidized metabolite are able to trap endogenous ACR via formation of ACR conjugates. Quantification further demonstrate that a) more than 13% of ingested phloretin can dose-dependently trap 0.77-9.92 nmol of ACR within 24 h; b) phloretin ingestion leads to marked reductions in both free ACR and ACR metabolites in mouse urine compared to control; and c) trapping reactions by phloretin can account for up to 20.1% of the total decreases in endogenous ACR, depending on the administration doses. CONCLUSION: Findings from this study indicate that regular consumption of DHCs-rich diets holds great promise to alleviate the development of ACR-associated chronic diseases.


Asunto(s)
Acroleína , Floretina , Acroleína/metabolismo , Acroleína/farmacología , Animales , Chalconas , Ingestión de Alimentos , Peroxidación de Lípido , Ratones , Floretina/farmacología
10.
Am J Physiol Lung Cell Mol Physiol ; 321(5): L912-L924, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34549600

RESUMEN

Asthma affects millions of people worldwide and its prevalence is increasing. It is characterized by chronic airway inflammation, airway remodeling, and pathologic bronchoconstriction, and it poses a continuous treatment challenge with very few new therapeutics available. Thus, many asthmatics turn to plant-based complementary products, including ginger, for better symptom control, indicating an unmet need for novel therapies. Previously, we demonstrated that 6-shogaol (6S), the primary bioactive component of ginger, relaxes human airway smooth muscle (hASM) likely by inhibition of phosphodiesterases (PDEs) in the ß-adrenergic (cyclic nucleotide PDEs), and muscarinic (phospholipase C, PLC) receptor pathways. However, oral 6S is extensively metabolized and it is unknown if the resulting metabolites remain bioactive. Here, we screened all the known human metabolites of 6S and several metabolite-based synthetic derivatives to better understand their mechanism of action and structure-function relationships. We demonstrate that several metabolites and metabolite-based synthetic derivatives are able to prevent Gq-coupled stimulation of intracellular calcium [Ca2+]i and inositol trisphosphate (IP3) synthesis by inhibiting PLC, similar to the parent compound 6S. We also show that these compounds prevent recontraction of ASM after ß-agonist relaxation likely by inhibiting PDEs. Furthermore, they potentiate isoproterenol-induced relaxation. Importantly, moving beyond cell-based assays, metabolites also retain the functional ability to relax Gq-coupled-contractions in upper (human) and lower (murine) airways. The current study indicates that, although oral ginger may be metabolized rapidly, it retains physiological activity through its metabolites. Moreover, we are able to use naturally occurring metabolites as inspiration to develop novel therapeutics for brochoconstrictive diseases.


Asunto(s)
Calcio/metabolismo , Relajación Muscular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Zingiber officinale , Animales , Asma/inducido químicamente , Asma/metabolismo , Broncoconstricción/efectos de los fármacos , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/metabolismo , Zingiber officinale/metabolismo , Humanos , Isoproterenol/farmacología , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Ratones Endogámicos C57BL , Relajación Muscular/fisiología , Músculo Liso/metabolismo , Miocitos del Músculo Liso/metabolismo
11.
Diabetes ; 70(11): 2506-2517, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34475098

RESUMEN

Activating transcription factor 3 (ATF3) has been shown to play an important role in HDL metabolism; yet, the role of hepatocytic ATF3 in the development of steatohepatitis remains elusive. Here we show that adenoassociated virus-mediated overexpression of human ATF3 in hepatocytes prevents diet-induced steatohepatitis in C57BL/6 mice and reverses steatohepatitis in db/db mice. Conversely, global or hepatocyte-specific loss of ATF3 aggravates diet-induced steatohepatitis. Mechanistically, hepatocytic ATF3 induces hepatic lipolysis and fatty acid oxidation and inhibits inflammation and apoptosis. We further show that hepatocyte nuclear factor 4α (HNF4α) is required for ATF3 to improve steatohepatitis. Thus, the current study indicates that ATF3 protects against steatohepatitis through, at least in part, hepatic HNF4α. Targeting hepatic ATF3 may be useful for treatment of steatohepatitis.


Asunto(s)
Factor de Transcripción Activador 3/metabolismo , Factor Nuclear 4 del Hepatocito/metabolismo , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Factor de Transcripción Activador 3/genética , Animales , Dieta Occidental , Regulación de la Expresión Génica/fisiología , Células Estrelladas Hepáticas , Factor Nuclear 4 del Hepatocito/genética , Hepatocitos/metabolismo , Humanos , Macrófagos del Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , MicroARNs/genética , MicroARNs/metabolismo , Enfermedad del Hígado Graso no Alcohólico/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo
12.
Mol Metab ; 51: 101244, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33930596

RESUMEN

OBJECTIVE: Hepatic miR-34a expression is elevated in diet-induced or genetically obese mice and patients with non-alcoholic steatohepatitis (NASH), yet hepatocyte miR-34a's role in the progression of non-alcoholic fatty liver disease (NAFLD) from non-alcoholic fatty liver (NAFL) to NASH remains to be elucidated. METHODS: Mice overexpressing or deficient in hepatocyte miR-34a and control mice were fed a diet enriched in fats, cholesterol, and fructose (HFCF) to induce NASH. C57BL/6 mice with NASH were treated with an miR-34a inhibitor or a scramble control oligo. The effect of miR-34a on the development, progression, and reversal of NAFLD was determined. RESULTS: The hepatocyte-specific expression of miR-34a aggravated HFCF diet-induced NAFLD. In contrast, germline or adult-onset deletion of hepatocyte miR-34a attenuated the development and progression of NAFLD. In addition, pharmacological inhibition of miR-34a reversed HFCF diet-induced steatohepatitis. Mechanistically, hepatocyte miR-34a regulated the development and progression of NAFLD by inducing lipid absorption, lipogenesis, inflammation, and apoptosis but inhibiting fatty acid oxidation. CONCLUSIONS: Hepatocyte miR-34a is an important regulator in the development and progression of NAFLD. MiR-34a may be a useful target for treating NAFLD.


Asunto(s)
Hepatocitos/metabolismo , Lipogénesis/genética , Hígado/patología , MicroARNs/metabolismo , Enfermedad del Hígado Graso no Alcohólico/genética , Animales , Apoptosis/genética , Colesterol/administración & dosificación , Colesterol/efectos adversos , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Exosomas/metabolismo , Fructosa/administración & dosificación , Fructosa/efectos adversos , Regulación de la Expresión Génica/efectos de los fármacos , Hepatocitos/citología , Hepatocitos/efectos de los fármacos , Humanos , Lipogénesis/efectos de los fármacos , Hígado/citología , Masculino , Ratones , Ratones Transgénicos , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/patología
13.
Nat Metab ; 3(1): 59-74, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33462514

RESUMEN

Activating transcription factor (ATF)3 is known to have an anti-inflammatory function, yet the role of hepatic ATF3 in lipoprotein metabolism or atherosclerosis remains unknown. Here we show that overexpression of human ATF3 in hepatocytes reduces the development of atherosclerosis in Western-diet-fed Ldlr-/- or Apoe-/- mice, whereas hepatocyte-specific ablation of Atf3 has the opposite effect. We further show that hepatic ATF3 expression is inhibited by hydrocortisone. Mechanistically, hepatocyte ATF3 enhances high-density lipoprotein (HDL) uptake, inhibits intestinal fat and cholesterol absorption and promotes macrophage reverse cholesterol transport by inducing scavenger receptor group B type 1 (SR-BI) and repressing cholesterol 12α-hydroxylase (CYP8B1) in the liver through its interaction with p53 and hepatocyte nuclear factor 4α, respectively. Our data demonstrate that hepatocyte ATF3 is a key regulator of HDL and bile acid metabolism and atherosclerosis.


Asunto(s)
Factor de Transcripción Activador 3/fisiología , Aterosclerosis/prevención & control , Ácidos y Sales Biliares/metabolismo , Hepatocitos/metabolismo , Lipoproteínas HDL/metabolismo , Animales , Apolipoproteínas E/genética , Colesterol en la Dieta/metabolismo , Grasas de la Dieta/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Factor Nuclear 4 del Hepatocito/metabolismo , Humanos , Hidrocortisona/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de LDL/genética , Receptores Depuradores de Clase B/metabolismo , Esteroide 12-alfa-Hidroxilasa/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
14.
Am J Physiol Gastrointest Liver Physiol ; 320(2): G166-G174, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33325808

RESUMEN

Human carboxylesterase 2 (CES2) has triacylglycerol hydrolase (TGH) activities and plays an important role in lipolysis. In this study, we aim to determine the role of human CES2 in the progression or reversal of steatohepatitis in diet-induced or genetically obese mice. High-fat/high-cholesterol/high-fructose (HFCF) diet-fed C57BL/6 mice or db/db mice were intravenously injected with an adeno-associated virus expressing human CES2 under the control of an albumin promoter. Human CES2 protected against HFCF diet-induced nonalcoholic fatty liver disease (NAFLD) in C57BL/6J mice and reversed steatohepatitis in db/db mice. Human CES2 also improved glucose tolerance and insulin sensitivity. Mechanistically, human CES2 reduced hepatic triglyceride (T) and free fatty acid (FFA) levels by inducing lipolysis and fatty acid oxidation and inhibiting lipogenesis via suppression of sterol regulatory element-binding protein 1. Furthermore, human CES2 overexpression improved mitochondrial respiration and glycolytic function, and inhibited gluconeogenesis, lipid peroxidation, apoptosis, and inflammation. Our data suggest that hepatocyte-specific expression of human CES2 prevents and reverses steatohepatitis. Targeting hepatic CES2 may be an attractive strategy for treatment of NAFLD.NEW & NOTEWORTHY Human CES2 attenuates high-fat/cholesterol/fructose diet-induced steatohepatitis and reverses steatohepatitis in db/db mice. Mechanistically, human CES2 induces lipolysis, fatty acid and glucose oxidation, and inhibits hepatic glucose production, inflammation, lipid oxidation, and apoptosis. Our data suggest that human CES2 may be targeted for treatment of non-alcoholic steatohepatitis (NASH).


Asunto(s)
Carboxilesterasa/metabolismo , Hepatocitos/enzimología , Enfermedad del Hígado Graso no Alcohólico/inducido químicamente , Enfermedad del Hígado Graso no Alcohólico/terapia , Ácido 3-Hidroxibutírico/sangre , Ácido 3-Hidroxibutírico/metabolismo , Alanina Transaminasa/sangre , Alanina Transaminasa/metabolismo , Animales , Apoptosis/fisiología , Aspartato Aminotransferasas/sangre , Aspartato Aminotransferasas/metabolismo , Glucemia , Carboxilesterasa/genética , Dieta/efectos adversos , Hidroxiprolina/sangre , Hidroxiprolina/metabolismo , Metabolismo de los Lípidos , Malondialdehído/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Transgénicos , Obesidad/inducido químicamente , Especies Reactivas de Oxígeno/metabolismo
15.
Hepatology ; 73(6): 2251-2265, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33098092

RESUMEN

BACKGROUND AND AIMS: Hepatocyte nuclear factor 4α (HNF4α) is highly enriched in the liver, but its role in the progression of nonalcoholic liver steatosis (NAFL) to NASH has not been elucidated. In this study, we investigated the effect of gain or loss of HNF4α function on the development and progression of NAFLD in mice. APPROACH AND RESULTS: Overexpression of human HNF4α protected against high-fat/cholesterol/fructose (HFCF) diet-induced steatohepatitis, whereas loss of Hnf4α had opposite effects. HNF4α prevented hepatic triglyceride accumulation by promoting hepatic triglyceride lipolysis, fatty acid oxidation, and VLDL secretion. Furthermore, HNF4α suppressed the progression of NAFL to NASH. Overexpression of human HNF4α inhibited HFCF diet-induced steatohepatitis in control mice but not in hepatocyte-specific p53-/- mice. In HFCF diet-fed mice lacking hepatic Hnf4α, recapitulation of hepatic expression of HNF4α targets cholesterol 7α-hydroxylase and sterol 12α-hydroxylase and normalized hepatic triglyceride levels and attenuated steatohepatitis. CONCLUSIONS: The current study indicates that HNF4α protects against diet-induced development and progression of NAFLD by coordinating the regulation of lipolytic, p53, and bile acid signaling pathways. Targeting hepatic HNF4α may be useful for treatment of NASH.


Asunto(s)
Ácidos y Sales Biliares/metabolismo , Dieta Alta en Grasa , Factor Nuclear 4 del Hepatocito/metabolismo , Hepatocitos/metabolismo , Hígado/metabolismo , Enfermedad del Hígado Graso no Alcohólico/prevención & control , Proteína p53 Supresora de Tumor/metabolismo , Animales , Ácido Quenodesoxicólico/análogos & derivados , Ácido Quenodesoxicólico/farmacología , Colesterol 7-alfa-Hidroxilasa/metabolismo , Factor Nuclear 4 del Hepatocito/genética , Hepatocitos/patología , Hígado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Transducción de Señal , Esteroide 12-alfa-Hidroxilasa/metabolismo , Triglicéridos/sangre , Proteína p53 Supresora de Tumor/genética
16.
Mol Nutr Food Res ; 64(22): e2000615, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32997396

RESUMEN

SCOPE: In this work, an integrated strategy is developed for rapid discovery, precise identification, and automated quantification for the biomarkers of food intake (BFIs) for specific food exposure using an ultra-high-pressure liquid chromatography-high-resolution mass spectrometry (MS) based targeted metabolomics approach. METHODS AND RESULTS: Using whole grain (WG) wheat intake as an example, the combination of paired mass distance networking and parallel reaction monitoring analysis is applied to selectively extract and identify WG metabolites in human urine samples. As a result, a total of 76 wheat phytochemical-derived metabolites, including 17 alkylresorcinol metabolites, 20 benzoxazinoid derivatives, and 39 phenolic acid metabolites are identified. Subsequently, a MS spectral database consisting of the identified metabolites is created by mzVault. The characteristics of identified metabolites from the database are incorporated into the TraceFinder software to establish a quantification platform. Using a standardized urine sample, the authors are able to simultaneously quantify both free and conjugated (sulfate and glucuronide) WG wheat metabolites in real samples without further enzymatic hydrolysis, which is validated by using authentic standards to quantify these metabolites. CONCLUSION: This novel strategy opens the window to study the biomarkers of specific food intake and make it feasible to validate the BFIs in large-scale human studies.


Asunto(s)
Biomarcadores/orina , Ingestión de Alimentos , Metabolómica/métodos , Triticum , Benzoxazinas/orina , Cromatografía Líquida de Alta Presión/métodos , Bases de Datos Factuales , Humanos , Espectrometría de Masas , Prueba de Estudio Conceptual , Urinálisis/métodos , Urinálisis/normas , Granos Enteros
17.
J Agric Food Chem ; 68(28): 7416-7424, 2020 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-32573222

RESUMEN

Our previous study has found that dietary genistein could ameliorate high-fat diet (HFD)-induced obesity and especially lower methylglyoxal (MGO) and advanced glycation end product (AGE) accumulation in healthy mice exposed to genistein and HFD. However, it is still unclear whether dietary genistein intervention has a similar beneficial effect in obese mice. In this study, the mice were induced with obesity after being fed a HFD for nine weeks before being administered with two doses of genistein, 0.1% (G 0.1) and 0.2% (G 0.2), in the HFD for additional 19 weeks. After 19 week treatment, genistein supplementation reduced body and liver weights, plasma and liver MGO levels, and kidney AGE levels in mice. Mechanistically, genistein upregulated the expressions of glyoxalase I and II and aldose reductase to detoxify MGO, and genistein and its microbial metabolites, dihydrogenistein and 6'-hydroxy-O-demethylangolensin, were able to trap endogenous MGO via formation of MGO conjugates. Taken together, our results provide novel insights into the antiobesity and antiglycation roles of dietary genistein in obese subjects.


Asunto(s)
Genisteína/metabolismo , Productos Finales de Glicación Avanzada/metabolismo , Obesidad/dietoterapia , Piruvaldehído/metabolismo , Aldehído Reductasa/metabolismo , Animales , Dieta Alta en Grasa/efectos adversos , Humanos , Lactoilglutatión Liasa/metabolismo , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Obesidad/etiología , Obesidad/metabolismo , Extractos Vegetales/metabolismo , Piruvaldehído/efectos adversos
18.
J Agric Food Chem ; 68(46): 12769-12772, 2020 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-32433871

RESUMEN

This special issue of the Journal of Agricultural and Food Chemistry (JAFC) is a highlight of the Agricultural and Food Chemistry Division (AGFD) technical program at the 258th National Meeting of the American Chemical Society (ACS) in San Diego, CA, U.S.A., on August 25-29, 2019. At the conference, AGFD had 44 oral sessions at 19 symposia and 100 poster presentations with more than 400 abstract submissions. The technical program covered a broad range of current research and development topics in agricultural and food chemistry, including bioactive food components, diet and human nutrition, utilization of agricultural materials in food systems, food packaging, nanotechnology, and food safety, as well as several special award symposia. This is the first JAFC special issue that highlights an ACS national meeting program with joint efforts from AGFD.


Asunto(s)
Química Agrícola , Análisis de los Alimentos , Agricultura , Dieta , Manipulación de Alimentos , Humanos , Valor Nutritivo
19.
Hepatol Commun ; 4(4): 527-539, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32258948

RESUMEN

Rodents have at least five carboxylesterase 1 (Ces1) genes, whereas there is only one CES1 gene in humans, raising the question as to whether human CES1 and mouse Ces1 genes share the same functions. In this study, we investigate the role of human CES1 in the development of steatohepatitis or dyslipidemia in C57BL/6 mice. Hepatocyte-specific expression of human CES1 prevented Western diet or alcohol-induced steatohepatitis and hyperlipidemia. Mechanistically, human CES1 induced lipolysis and fatty acid oxidation, leading to a reduction in hepatic triglyceride and free fatty acid levels. Human CES1 also reduced hepatic-free cholesterol levels and induced low-density lipoprotein receptor. In addition, human CES1 induced hepatic lipoprotein lipase and apolipoprotein C-II expression. Conclusion: Hepatocyte-specific overexpression of human CES1 attenuates diet-induced steatohepatitis and hyperlipidemia.

20.
Mol Ther ; 28(1): 202-216, 2020 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-31604677

RESUMEN

Macrophages play a crucial role in the pathogenesis of atherosclerosis, but the molecular mechanisms remain poorly understood. Here we show that microRNA-34a (miR-34a) is a key regulator of macrophage cholesterol efflux and reverse cholesterol transport by modulating ATP-binding cassette transporters ATP-binding cassette subfamily A member 1 (ABCA1) and ATP-binding cassette subfamily G member 1 (ABCG1). miR-34a also regulates M1 and M2 macrophage polarization via liver X receptor α. Furthermore, global loss of miR-34a reduces intestinal cholesterol or fat absorption by inhibiting cytochrome P450 enzymes CYP7A1 and sterol 12α-hydroxylase (CYP8B1). Consistent with these findings, macrophage-selective or global ablation of miR-34a markedly inhibits the development of atherosclerosis. Finally, therapeutic inhibition of miR-34a promotes atherosclerosis regression and reverses diet-induced metabolic disorders. Our studies outline a central role of miR-34a in regulating macrophage cholesterol efflux, inflammation, and atherosclerosis, suggesting that miR-34a is a promising target for treatment of cardiometabolic diseases.


Asunto(s)
Aterosclerosis/metabolismo , Colesterol/metabolismo , Macrófagos/metabolismo , MicroARNs/metabolismo , Transportador 1 de Casete de Unión a ATP/genética , Transportador 1 de Casete de Unión a ATP/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1/genética , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1/metabolismo , Animales , Polaridad Celular/genética , Modelos Animales de Enfermedad , Células Hep G2 , Humanos , Receptores X del Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE , MicroARNs/genética , Células RAW 264.7 , Células THP-1 , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...