Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
1.
J Agric Food Chem ; 72(31): 17392-17404, 2024 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-39056217

RESUMEN

Ketosis in dairy cows is often accompanied by the dysregulation of lipid homeostasis in the liver. Acetyl-coenzyme A acetyltransferase 2 (ACAT2) is specifically expressed in the liver and is important for regulating lipid homeostasis in ketotic cows. Lentinan (LNT) has a wide range of pharmacological activities, and this study investigates the protective effects of LNT on ß-hydroxybutyrate (BHBA)-induced lipid metabolism disorder in bovine hepatocytes (BHECs) and elucidates the underlying mechanisms. BHECs were first pretreated with LNT to investigate the effect of LNT on BHBA-induced lipid metabolism disorder in BHECs. ACAT2 was then silenced or overexpressed to investigate whether this mediated the protective action of LNT against BHBA-induced lipid metabolism disorder in BHECs. Finally, BHECs were treated with LNT after silencing ACAT2 to investigate the interaction between LNT and ACAT2. LNT pretreatment effectively enhanced the synthesis and absorption of cholesterol, inhibited the synthesis of triglycerides, increased the expression of ACAT2, and elevated the contents of very low-density lipoprotein and low-density lipoprotein cholesterol, thereby ameliorating BHBA-induced lipid metabolism disorder in BHECs. The overexpression of ACAT2 achieved a comparable effect to LNT pretreatment, whereas the silencing of ACAT2 aggravated the effect of BHBA on inducing disorder in lipid metabolism in BHECs. Moreover, the protective effect of LNT against lipid metabolism disorder in BHBA-induced BHECs was abrogated upon silencing of ACAT2. Thus, LNT, as a natural protective agent, can enhance the regulatory capacity of BHECs in maintaining lipid homeostasis by upregulating ACAT2 expression, thereby ameliorating the BHBA-induced lipid metabolism disorder.


Asunto(s)
Ácido 3-Hidroxibutírico , Acetil-CoA C-Acetiltransferasa , Hepatocitos , Metabolismo de los Lípidos , Regulación hacia Arriba , Animales , Bovinos , Hepatocitos/metabolismo , Hepatocitos/efectos de los fármacos , Ácido 3-Hidroxibutírico/metabolismo , Ácido 3-Hidroxibutírico/farmacología , Metabolismo de los Lípidos/efectos de los fármacos , Acetil-CoA C-Acetiltransferasa/genética , Acetil-CoA C-Acetiltransferasa/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Trastornos del Metabolismo de los Lípidos/metabolismo , Trastornos del Metabolismo de los Lípidos/genética , Trastornos del Metabolismo de los Lípidos/tratamiento farmacológico , Trastornos del Metabolismo de los Lípidos/inducido químicamente , Triglicéridos/metabolismo , Enfermedades de los Bovinos/metabolismo , Enfermedades de los Bovinos/genética , Enfermedades de los Bovinos/tratamiento farmacológico , Cetosis/metabolismo , Cetosis/genética , Cetosis/inducido químicamente
2.
Aquat Toxicol ; 273: 107015, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38996482

RESUMEN

Nitrite, a highly toxic environmental contaminant, induces various physiological toxicities in aquatic animals. Herein, we investigate the in vivo effects of nitrite exposure at concentrations of 0, 0.2, 2, and 20 mg/L on glucose and lipid metabolism in zebrafish. Our results showed that exposure to nitrite induced mitochondrial oxidative stress in zebrafish liver and ZFL cells, which were evidenced by increased levels of malondialdehyde (MDA) and reactive oxygen species (ROS) as well as decreased mitochondrial membrane potential (MMP) and adenosine triphosphate (ATP). Changes in these oxidative stress markers were accompanied by alterations in the expression levels of genes involved in HIF-1α pathway (hif1α and phd), which subsequently led to the upregulation of glycolysis and gluconeogenesis-related genes (gk, pklr, pdk1, pepck, g6pca, ppp1r3cb, pgm1, gys1 and gys2), resulting in disrupted glucose metabolism. Moreover, nitrite exposure activated ERs (Endoplasmic Reticulum stress) responses through upregulating of genes (atf6, ern1 and xbp1s), leading to increased expression of lipolysis genes (pparα, cpt1aa and atgl) and decreased expression of lipid synthesis genes (srebf1, srebf2, fasn, acaca, scd, hmgcra and hmgcs1). These results were also in consistent with the observed changes in glycogen, lactate and decreased total triglyceride (TG) and total cholesterol (TC) in the liver of zebrafish. Our in vitro results showed that co-treatment with Mito-TEMPO and nitrite attenuated nitrite-induced oxidative stress and improved mitochondrial function, which were indicated by the restorations of ROS, MMP, ATP production, and glucose-related gene expression recovered. Co-treatment of TUDCA and nitrite prevented nitrite-induced ERs response and which was proved by the levels of TG and TC ameliorated as well as the expression levels of lipid metabolism-related genes. In conclusion, our study suggested that nitrite exposure disrupted hepatic glucose and lipid metabolism through mitochondrial dysfunction and ERs responses. These findings contribute to the understanding of the potential hepatotoxicity for aquatic animals in the presence of ambient nitrite.


Asunto(s)
Estrés del Retículo Endoplásmico , Glucosa , Metabolismo de los Lípidos , Hígado , Nitritos , Estrés Oxidativo , Contaminantes Químicos del Agua , Pez Cebra , Animales , Glucosa/metabolismo , Nitritos/toxicidad , Hígado/efectos de los fármacos , Hígado/metabolismo , Estrés del Retículo Endoplásmico/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Contaminantes Químicos del Agua/toxicidad , Metabolismo de los Lípidos/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Trastornos del Metabolismo de los Lípidos/inducido químicamente , Trastornos del Metabolismo de los Lípidos/genética
3.
Artículo en Chino | MEDLINE | ID: mdl-38964904

RESUMEN

Objective: To investigate the mechanism of Sulfo-N-succinimidyloleate (SSO) regulating lipid metabolism disorder induced by silicon dioxide (SiO(2)) . Methods: In March 2023, Rat alveolar macrophages NR8383 were cultured in vitro and randomly divided into control group (C), SSO exposure group (SSO), SiO(2) exposure group (SiO(2)) and SiO(2)+SSO exposure group (SiO(2)+SSO). NR8383 cells were exposure separately or jointly by SSO and SiO(2) for 36 h to construct cell models. Immunofluorescence and BODIPY 493/ 503 staining were used to detect cluster of differentiation (CD36) and intracellular lipid levels, the protein expression levels of CD36, liver X receptors (LXR), P-mammalian target of rapamycin (P-mTOR) and cholinephosphotransferase 1 (CHPT1) were detected by Western blot, respectively, and lipid metabolomics was used to screen for different lipid metabolites and enrichment pathways. Single-factor ANOVA was used for multi-group comparison, and LSD test was used for pair-to-group comparison. Results: SiO(2) caused the expression of CD36 and P-mTOR to increase (P=0.012, 0.020), the expression of LXR to decrease (P=0.005), and the intracellular lipid level to increase. After SSO treatment, CD36 expression decreased (P=0.023) and LXR expression increased (P=0.000) in SiO(2)+SSO exposure group compared with SiO(2) exposure group. Metabolomics identified 87 different metabolites in the C group and SiO(2) exposure group, 19 different metabolites in the SiO(2) exposure group and SiO(2)+SSO group, and 5 overlaps of different metabolites in the two comparison groups, they are PS (22∶1/14∶0), DG (O-16∶0/18∶0/0∶0), PGP (i-13∶0/i-20∶0), PC (18∶3/16∶0), and Sphinganine. In addition, the differential metabolites of the two comparison groups were mainly concentrated in the glycerophospholipid metabolism and sphingolipid metabolism pathways. The differential gene CHPT1 in glycerophospholipid metabolic pathway was verified, and the expression of CHPT1 decreased after SiO(2) exposure. Conclusion: SSO may improve SiO(2)-induced lipid metabolism disorders by regulating PS (22∶1/14∶0), DG (O-16∶0/18∶0/0∶0), PGP (i-13∶0/i-20∶0), PC (18∶3/16∶0), SPA, glycerophospholipid metabolism and sphingolipid metabolism pathways.


Asunto(s)
Antígenos CD36 , Metabolismo de los Lípidos , Dióxido de Silicio , Animales , Ratas , Dióxido de Silicio/toxicidad , Metabolismo de los Lípidos/efectos de los fármacos , Antígenos CD36/metabolismo , Metabolómica , Trastornos del Metabolismo de los Lípidos/metabolismo , Trastornos del Metabolismo de los Lípidos/inducido químicamente , Macrófagos/metabolismo , Macrófagos/efectos de los fármacos , Receptores X del Hígado/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Lípidos
4.
Environ Pollut ; 358: 124458, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-38942276

RESUMEN

Cyanobacterial blooms have emerged as a significant environmental issue worldwide in recent decades. However, the toxic effects of microcystin-LR (MC-LR) on aquatic organisms, such as frogs, have remained poorly understood. In this study, frogs (Pelophylax nigromaculatus) were exposed to environmentally relevant concentrations of MC-LR (0, 1, and 10 µg/L) for 21 days. Subsequently, we assessed the impact of MC-LR on the histomorphology of the frogs' livers and conducted a global MS-based nontarget metabolomics analysis, followed by the determination of substances involved in lipid metabolism. Results showed that MC-LR significantly induced histological alterations in the frogs' hepatopancreas. Over 200 differentially expressed metabolites were identified, primarily enriched in lipid metabolism. Biochemical analysis further confirmed that MC-LR exposure led to a disorder in lipid metabolism in the frogs. This study laid the groundwork for a mechanistic understanding of MC-LR toxicity in frogs and potentially other aquatic organisms.


Asunto(s)
Metabolismo de los Lípidos , Toxinas Marinas , Metabolómica , Microcistinas , Contaminantes Químicos del Agua , Microcistinas/toxicidad , Microcistinas/metabolismo , Animales , Metabolismo de los Lípidos/efectos de los fármacos , Contaminantes Químicos del Agua/toxicidad , Contaminantes Químicos del Agua/metabolismo , Ranidae/metabolismo , Trastornos del Metabolismo de los Lípidos/metabolismo , Trastornos del Metabolismo de los Lípidos/inducido químicamente , Hepatopáncreas/metabolismo , Hepatopáncreas/efectos de los fármacos , Hígado/metabolismo , Hígado/efectos de los fármacos
5.
ACS Appl Mater Interfaces ; 16(27): 34524-34537, 2024 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-38926154

RESUMEN

In recent years, the study of microplastics (MPs) and nanoplastics (NPs) and their effects on human health has gained significant attention. The impacts of NPs on lipid metabolism and the specific mechanisms involved remain poorly understood. To address this, we utilized high-throughput sequencing and molecular biology techniques to investigate how endoplasmic reticulum (ER) stress might affect hepatic lipid metabolism in the presence of polystyrene nanoplastics (PS-NPs). Our findings suggest that PS-NPs activate the PERK-ATF4 signaling pathway, which in turn upregulates the expression of genes related to lipid synthesis via the ATF4-PPARγ/SREBP-1 pathway. This activation leads to an abnormal accumulation of lipid droplets in the liver. 4-PBA, a known ER stress inhibitor, was found to mitigate the PS-NPs-induced lipid metabolism disorder. These results demonstrate the hepatotoxic effects of PS-NPs and clarify the mechanisms of abnormal lipid metabolism induced by PS-NPs.


Asunto(s)
Factor de Transcripción Activador 4 , Poliestirenos , Transducción de Señal , eIF-2 Quinasa , Poliestirenos/química , Poliestirenos/toxicidad , Poliestirenos/farmacología , Factor de Transcripción Activador 4/metabolismo , Factor de Transcripción Activador 4/genética , Animales , Ratones , Transducción de Señal/efectos de los fármacos , eIF-2 Quinasa/metabolismo , eIF-2 Quinasa/genética , Trastornos del Metabolismo de los Lípidos/metabolismo , Trastornos del Metabolismo de los Lípidos/inducido químicamente , Trastornos del Metabolismo de los Lípidos/tratamiento farmacológico , Nanopartículas/química , Nanopartículas/toxicidad , Microplásticos/toxicidad , Estrés del Retículo Endoplásmico/efectos de los fármacos , Metabolismo de los Lípidos/efectos de los fármacos , Masculino , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Ratones Endogámicos C57BL
6.
Acta Pharmacol Sin ; 45(9): 1898-1911, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38760545

RESUMEN

Tacrolimus, one of the macrolide calcineurin inhibitors, is the most frequently used immunosuppressant after transplantation. Long-term administration of tacrolimus leads to dyslipidemia and affects liver lipid metabolism. In this study, we investigated the mode of action and underlying mechanisms of this adverse reaction. Mice were administered tacrolimus (2.5 mg·kg-1·d-1, i.g.) for 10 weeks, then euthanized; the blood samples and liver tissues were collected for analyses. We showed that tacrolimus administration induced significant dyslipidemia and lipid deposition in mouse liver. Dyslipidemia was also observed in heart or kidney transplantation patients treated with tacrolimus. We demonstrated that tacrolimus did not directly induce de novo synthesis of fatty acids, but markedly decreased fatty acid oxidation (FAO) in AML12 cells. Furthermore, we showed that tacrolimus dramatically decreased the expression of HMGCS2, the rate-limiting enzyme of ketogenesis, with decreased ketogenesis in AML12 cells, which was responsible for lipid deposition in normal hepatocytes. Moreover, we revealed that tacrolimus inhibited forkhead box protein O1 (FoxO1) nuclear translocation by promoting FKBP51-FoxO1 complex formation, thus reducing FoxO1 binding to the HMGCS2 promoter and its transcription ability in AML12 cells. The loss of HMGCS2 induced by tacrolimus caused decreased ketogenesis and increased acetyl-CoA accumulation, which promoted mitochondrial protein acetylation, thereby resulting in FAO function inhibition. Liver-specific HMGCS2 overexpression via tail intravenous injection of AAV8-TBG-HMGCS2 construct reversed tacrolimus-induced mitochondrial protein acetylation and FAO inhibition, thus removing the lipid deposition in hepatocytes. Collectively, this study demonstrates a novel mechanism of liver lipid deposition and hyperlipidemia induced by long-term administration of tacrolimus, resulted from the loss of HMGCS2-mediated ketogenesis and subsequent FAO inhibition, providing an alternative target for reversing tacrolimus-induced adverse reaction.


Asunto(s)
Hidroximetilglutaril-CoA Sintasa , Hígado , Ratones Endogámicos C57BL , Tacrolimus , Animales , Tacrolimus/farmacología , Ratones , Masculino , Hidroximetilglutaril-CoA Sintasa/metabolismo , Hidroximetilglutaril-CoA Sintasa/genética , Humanos , Hígado/metabolismo , Hígado/efectos de los fármacos , Metabolismo de los Lípidos/efectos de los fármacos , Proteína Forkhead Box O1/metabolismo , Inmunosupresores/farmacología , Trastornos del Metabolismo de los Lípidos/metabolismo , Trastornos del Metabolismo de los Lípidos/inducido químicamente , Trastornos del Metabolismo de los Lípidos/tratamiento farmacológico , Línea Celular
7.
Biomed Pharmacother ; 174: 116557, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38583337

RESUMEN

Myricanol (MY) is one of the main active components from bark of Myrica Rubra. It is demonstrated that MY rescues dexamethasone (DEX)-induced muscle dysfunction via activating silent information regulator 1 (SIRT1) and increasing adenosine 5'-monophosphate-activated protein kinase (AMPK) phosphorylation. Since SIRT1 and AMPK are widely involved in the metabolism of nutrients, we speculated that MY may exert beneficial effects on DEX-induced metabolic disorders. This study for the first time applied widely targeted metabolomics to investigate the beneficial effects of MY on glucose, lipids, and protein metabolism in DEX-induced metabolic abnormality in mice. The results showed that MY significantly reversed DEX-induced soleus and gastrocnemius muscle weight loss, muscle fiber damage, and muscle strength loss. MY alleviated DEX-induced metabolic disorders by increasing SIRT1 and glucose transporter type 4 (GLUT4) expressions. Additionally, myricanol prevented muscle cell apoptosis and atrophy by inhibiting caspase 3 cleavages and muscle ring-finger protein-1 (MuRF1) expression. Metabolomics showed that MY treatment reversed the serum content of carnitine ph-C1, palmitoleic acid, PS (16:0_17:0), PC (14:0_20:5), PE (P-18:1_16:1), Cer (t18:2/38:1(2OH)), four amino acids and their metabolites, and 16 glycerolipids in DEX mice. Kyoto encyclopedia of genes and genomes (KEGG) and metabolic set enrichment analysis (MSEA) analysis revealed that MY mainly affected metabolic pathways, glycerolipid metabolism, lipolysis, fat digestion and absorption, lipid and atherosclerosis, and cholesterol metabolism pathways through regulation of metabolites involved in glutathione, butanoate, vitamin B6, glycine, serine and threonine, arachidonic acid, and riboflavin metabolism. Collectively, MY can be used as an attractive therapeutic agent for DEX-induced metabolic abnormalities.


Asunto(s)
Dexametasona , Animales , Dexametasona/farmacología , Ratones , Masculino , Metabolismo de los Lípidos/efectos de los fármacos , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Sirtuina 1/metabolismo , Metaboloma/efectos de los fármacos , Trastornos del Metabolismo de los Lípidos/tratamiento farmacológico , Trastornos del Metabolismo de los Lípidos/metabolismo , Trastornos del Metabolismo de los Lípidos/inducido químicamente , Apoptosis/efectos de los fármacos , Ratones Endogámicos C57BL , Metabolómica/métodos
8.
Sci Total Environ ; 931: 172795, 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38677429

RESUMEN

The ubiquitous presence of micro-and nanoplastics (MNPs) in the environment and everyday products has attracted attention due to their hazardous risks. However, the effects of MNPs on reproduction and the underlying mechanisms remain unclear. The present study investigated the impact of polystyrene (PS) nanoplastics of 80, 200 and 500 nm diameters on zebrafish reproduction at an environmentally relevant concentration of 0.5 mg/L. Exposure to PS delayed spermatogenesis and caused aberrant follicular growth, resulting in dysgenesis in F0 adults and impacting F1 embryo development. Notably, the reproductive toxicity exhibited size-dependency, with the 500 nm PS being the most detrimental. Combined analyses of transcriptomics and metabolomics in ovary tissue revealed that treatment with 500 nm PS affected the peroxisome proliferator-activated receptor (PPAR) signaling pathway, dysregulated lipid transport, binding and activity processes, and led to dysgenesis in zebrafish. Specifically, the ovulatory dysfunction induced by PS exposure resembled clinical manifestations of polycystic ovary syndrome (PCOS) and can be attributed to lipid metabolism disorder involving glycerophospholipid, sphingolipid, arachidonic acid, and alpha-linolenic acid. Collectively, our results provide new evidence revealing the molecular mechanisms of PS-induced reproductive toxicity, highlighting that MNPs may pose a risk to female reproductive health.


Asunto(s)
Trastornos del Metabolismo de los Lípidos , Receptores Activados del Proliferador del Peroxisoma , Poliestirenos , Reproducción , Contaminantes Químicos del Agua , Pez Cebra , Animales , Poliestirenos/toxicidad , Receptores Activados del Proliferador del Peroxisoma/metabolismo , Reproducción/efectos de los fármacos , Trastornos del Metabolismo de los Lípidos/inducido químicamente , Contaminantes Químicos del Agua/toxicidad , Femenino , Metabolismo de los Lípidos/efectos de los fármacos , Masculino , Microplásticos/toxicidad
9.
J Appl Toxicol ; 44(6): 919-932, 2024 06.
Artículo en Inglés | MEDLINE | ID: mdl-38400677

RESUMEN

Isobavachalcone (IBC) is a flavonoid component derived from Psoraleae Fructus that can increase skin pigmentation and treat vitiligo. However, IBC has been reported to be hepatotoxic. Current studies on IBC hepatotoxicity are mostly on normal organisms but lack studies on hepatotoxicity in patients. This study established the depigmented zebrafish model by using phenylthiourea (PTU) and investigated the difference in hepatotoxicity between normal and depigmented zebrafish caused by IBC and the underlying mechanism. Morphological, histological, and ultrastructural examination and RT-qPCR verification were used to evaluate the effects of IBC on the livers of zebrafish larvae. IBC significantly decreased liver volume, altered lipid metabolism, and induced pathological and ultrastructural changes in the livers of zebrafish with depigmentation compared with normal zebrafish. The RNA-sequencing and RT-qPCR results showed that the difference in hepatotoxicity between normal and depigmented zebrafish caused by IBC was closely related to the calcium signaling pathway, lipid decomposition and metabolism, and oxidative stress. This work delved into the mechanism of the enhanced IBC-induced hepatotoxicity in depigmented zebrafish and provided a new insight into the hepatotoxicity of IBC.


Asunto(s)
Señalización del Calcio , Chalconas , Enfermedad Hepática Inducida por Sustancias y Drogas , Pez Cebra , Animales , Chalconas/toxicidad , Señalización del Calcio/efectos de los fármacos , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Trastornos del Metabolismo de los Lípidos/inducido químicamente , Trastornos del Metabolismo de los Lípidos/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Metabolismo de los Lípidos/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos
10.
Chem Res Toxicol ; 36(5): 725-733, 2023 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-37093692

RESUMEN

Our previous studies found that di (2-ethylhexyl) phthalate (DEHP) could disorder lipid metabolism in adolescents but the mechanisms underlying this association remained unclear. This study was undertaken to clarify the mediating effect of JAK3/STAT5/PPARγ on disorder lipid levels induced by DEHP in adolescents. We recruited 478 adolescent students (median age 18.1 years). The mRNA expression and DNA methylation levels of JAK3/STAT5/PPARγ were detected by real-time PCR and the MethylTarget, respectively. We used multiple linear regression to analyze the association between DEHP metabolites (MEHP, MEOHP, MEHHP, MECPP, MCMHP, and ΣDEHP) levels, mRNA expression, and DNA methylation levels. The mediating effect of JAK3/STAT5/PPARγ mRNA expression levels was examined by mediation analysis. We found that all DEHP metabolite levels were positively correlated with TC/HDL-C and LDL-C/HDL-C (P < 0.05). The MEOHP level was negatively associated with DNA methylation levels and positively associated with mRNA levels of PPARγ and STAT5b (P < 0.05). The MEHP level was negatively associated with the DNA methylation level and positively associated with the mRNA level of JAK3 (P < 0.05). Higher MEOHP was associated with a higher level of TC/HDL-C, the mediation analysis showed the mediation effect was 17.18% for the JAK3 level, 10.76% for the STAT5b level, and 11% for the PPARγ level. Higher MEHP was associated with a higher level of LDL-C/HDL-C, the mediation effect was 14.49% for the JAK3 level. In conclusion, DEHP metabolites decreased the DNA methylation levels, inducing the increase of the mRNA levels of JAK3/STAT5/PPARγ. In addition, the mRNA levels mediated the association between DEHP exposure and disorder lipid levels.


Asunto(s)
Dietilhexil Ftalato , Trastornos del Metabolismo de los Lípidos , Adolescente , Humanos , LDL-Colesterol/metabolismo , Dietilhexil Ftalato/efectos adversos , Pueblos del Este de Asia , Janus Quinasa 3/metabolismo , Ácidos Ftálicos/efectos adversos , PPAR gamma/genética , PPAR gamma/metabolismo , Factor de Transcripción STAT5/metabolismo , Estudiantes , Trastornos del Metabolismo de los Lípidos/inducido químicamente , Trastornos del Metabolismo de los Lípidos/metabolismo
11.
Toxicol Sci ; 191(2): 276-284, 2023 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-36534932

RESUMEN

2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is widely considered as the most toxic and common carcinogen in the world. Exposure to TCDD causes liver lipid metabolism disorder and steatosis. However, the molecular mechanism of TCDD-induced liver lipid accumulation is not completely clear. Here, we found that a 5 µg/kg TCDD exposure for 3 weeks induced hepatocyte lipid deposition, increased CD36 expression, and promoted AMP-activated protein kinase (AMPK) ɑ phosphorylation in the liver of C57BL/6J mice. Furthermore, sulfo-N-succinimidyl oleate, a CD36 inhibiter, blunted TCDD-induced lipid deposition in Huh7 cells, confirming the critical role of CD36 in TCDD-induced hepatic steatosis. In terms of molecular mechanisms, we found that TCDD exposure increased reactive oxygen species (ROS) levels in Huh7 cells, which activated AMPK. Moreover, the activated AMPK upregulated CD36 expression. Therefore, we can see that the increase in CD36 expression induced by TCDD was regulated by ROS/AMPK/CD36 signaling pathway. Our results help to clarify the molecular mechanism of TCDD-induced hepatic steatosis.


Asunto(s)
Hígado Graso , Trastornos del Metabolismo de los Lípidos , Dibenzodioxinas Policloradas , Animales , Ratones , Proteínas Quinasas Activadas por AMP/metabolismo , Hígado Graso/inducido químicamente , Hígado Graso/metabolismo , Metabolismo de los Lípidos , Trastornos del Metabolismo de los Lípidos/inducido químicamente , Trastornos del Metabolismo de los Lípidos/metabolismo , Lípidos , Hígado/metabolismo , Ratones Endogámicos C57BL , Dibenzodioxinas Policloradas/toxicidad , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Antígenos CD36/metabolismo
12.
J Environ Sci (China) ; 124: 481-490, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36182156

RESUMEN

Triclosan (TCS) is a ubiquitous antimicrobial used in daily consumer products. Previous reports have shown that TCS could induce hepatotoxicity, endocrine disruption, disturbance on immune function and impaired thyroid function. Kidney is critical in the elimination of toxins, while the effects of TCS on kidney have not yet been well-characterized. The aim of the present study was to investigate the effects of TCS exposure on kidney function and the possible underlying mechanisms in mice. Male C57BL/6 mice were orally exposed to TCS with the doses of 10 and 100 mg/(kg•day) for 13 weeks. TCS was dissolved in dimethyl sulfoxide (DMSO) and diluted by corn oil for exposure. Corn oil containing DMSO was used as vehicle control. Serum and kidney tissues were collected for study. Biomarkers associated with kidney function, oxidative stress, inflammation and fibrosis were assessed. Our results showed that TCS could cause renal injury as was revealed by increased levels of renal function markers including serum creatinine, urea nitrogen and uric acid, as well as increased oxidative stress, pro-inflammatory cytokines and fibrotic markers in a dose dependent manner, which were more significantly in 100 mg/(kg•day) group. Mass spectrometry-based analysis of metabolites related with lipid metabolism demonstrated the occurrence of lipid accumulation and defective fatty acid oxidation in 100 mg/(kg•day) TCS-exposed mouse kidney. These processes might lead to lipotoxicity and energy depletion, thus resulting in kidney fibrosis and functional decline. Taken together, the present study demonstrated that TCS could induce lipid accumulation and fatty acid metabolism disturbance in mouse kidney, which might contribute to renal function impairment. The present study further widens our insights into the adverse effects of TCS.


Asunto(s)
Antiinfecciosos , Trastornos del Metabolismo de los Lípidos , Triclosán , Animales , Aceite de Maíz/metabolismo , Aceite de Maíz/farmacología , Creatinina/metabolismo , Creatinina/farmacología , Citocinas/metabolismo , Citocinas/farmacología , Dimetilsulfóxido/metabolismo , Dimetilsulfóxido/farmacología , Ácidos Grasos/metabolismo , Fibrosis , Riñón/metabolismo , Metabolismo de los Lípidos , Trastornos del Metabolismo de los Lípidos/inducido químicamente , Trastornos del Metabolismo de los Lípidos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Nitrógeno/metabolismo , Triclosán/toxicidad , Urea , Ácido Úrico/metabolismo , Ácido Úrico/farmacología
13.
Ecotoxicol Environ Saf ; 239: 113649, 2022 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-35605325

RESUMEN

Microcystin-LR (MC-LR), a potent hepatotoxin can cause liver damages. However, research on hepatic lipid metabolism caused by long-term exposure to environmental concentrations MC-LR is limited. In the current study, mice were exposed to various low concentrations of MC-LR (0, 1, 30, 60, 90, 120 µg/L in the drinking water) for 9 months. The general parameters, serum and liver lipids, liver tissue pathology, lipid metabolism-related genes and proteins of liver were investigated. The results show that chronic MC-LR exposure had increased the levels of triglyceride (TG) and total cholesterol (TC) in serum and liver. In addition, histological observation revealed that hepatic lobules were disordered with obvious inflammatory cell infiltration and lipid droplets. More importantly, the mRNA and proteins expression levels of lipid synthesis-related nuclear sterol regulatory element binding protein-1c (nSREBP-1c), SREBP-1c, cluster of differentiation 36 (CD36), acetyl-CoA-carboxylase1 (ACC1), stearoyl-CoA desaturase1 (SCD1) and fatty acid synthase (FASN) were increased in MC-LR treated groups, the expression levels of fatty acids ß-oxidation related genes peroxisomal acyl-coenzyme A oxidase 1 (ACOX1) was decreased after exposure to 60-120 µg/L MC-LR. Furthermore, the inflammatory factors interleukin 6 (IL-6) and tumor necrosis factor-α (TNF-α) were higher than that in the control group. All the findings indicated that mice were exposed to chronic low concentrations MC-LR caused liver inflammation and hepatic lipid metabolism disorder .


Asunto(s)
Trastornos del Metabolismo de los Lípidos , Metabolismo de los Lípidos , Microcistinas , Animales , Trastornos del Metabolismo de los Lípidos/inducido químicamente , Trastornos del Metabolismo de los Lípidos/patología , Hígado/metabolismo , Hígado/fisiopatología , Toxinas Marinas/toxicidad , Ratones , Microcistinas/toxicidad , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética
14.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 47(1): 18-25, 2022 Jan 28.
Artículo en Inglés, Chino | MEDLINE | ID: mdl-35545359

RESUMEN

OBJECTIVES: Perfluorooctanoic acid (PFOA) can cause lipid metabolism disorders in animal body and affect the lipolysis and synthesis of fatty acids. Peroxisome proliferators-activated receptor (PPAR) plays an extremely important role in this process. This study aims to explore the effects of PFOA on liver lipid metabolism disorders in Sprague Dewley (SD) rats and the expression of PPAR. METHODS: A total of 40 male SD rats were randomly divided into 4 groups (n=10 in each group): a control group (ddH2O), a low-dose PFOA group [PFOA 1.25 mg/(kg·d)], a middle-dose PFOA group [PFOA 5.00 mg/(kg·d)], and a high-dose PFOA group [PFOA 20.00 mg/(kg·d)]. The rats were fed with normal diet, and PFOA exposure were performed by oral gavage for 14 days, and the rats were observed, weighted and recorded every day during the exposure. After the exposure, the blood was collected, and the livers were quickly stripped after the rats were killed. Part of the liver tissues were fixed in 4% paraformaldehyde for periodic acid-schiff (PAS) staining; the contents of HDLC, LDLC, TG, TC in serum and liver tissues, as well as the activities of their related enzymes were assayed; The expression levels of cyclic adenosine monophosphate-response element binding protein (Cbp), general control of amino acid synthesis 5-like 2 (Gcn5L2), peroxidation peroxisome proliferation factor activated receptor γ (PPAR), silent information regulator 1 (Sirt1) and human retinoid X receptor alpha 2 (Rxrα2) ) were detected by Western blotting. RESULTS: After 14 days of PFOA exposure, the PAS staining positive particles in the cytoplasm and nucleus of SD rats in the medium and high dose groups were significantly reduced compared with the control group. The serum levels of LDLC and TC in the low-dose and middle-dose groups were significantly reduced compared with the control group (all P<0.05), while the high-dose group showed an increasing tendency, without siginificant difference (P>0.05), there was no significant difference in HDLC and TG (both P>0.05). The activities of alkaline phosphatase (AKP) and alanine aminotransferase (ALT) were increased significantly (both P<0.05) compared with control group; the ratio of ALT/aspartate aminotransferase (AST) in the high-dose group was increased significantly (P<0.05), there was no significant difference in LDH and TG (both P>0.05); the HDLC content in the liver tissues in the high-dose group was significantly reduced, compared with the control group (P<0.05); the TC contents in the liver tissues in the low, medium and high-dose groups were significantly increased (all P<0.05), there was no significant difference in LDLC and TG (both P>0.05); the AKP activity in the livers in the medium and high-dose groups was significantly increased (both P<0.05), there was no siginificant difference in LDH, ALT, and the ratio of ALT/AST (all P>0.05); the protein expression levels of Ppar γ, Cbp and Rxrα2 in the liver in the high dose groups were significantly down-regulated compared with the control group (all P<0.05), while the protein expression levels of Sirt1 were significantly up-regulated (all P<0.05). CONCLUSIONS: PFOA exposure can cause lipid metabolism disorder and glycogen reduction in SD rat livers, which may be related to the activation of Sirt1 and inhibition of Ppar γ expression, leading to affecting the normal metabolism of fatty acids and promoting glycolysis.


Asunto(s)
Trastornos del Metabolismo de los Lípidos , Metabolismo de los Lípidos , Animales , Caprilatos , Ácidos Grasos/metabolismo , Ácidos Grasos/farmacología , Fluorocarburos , Trastornos del Metabolismo de los Lípidos/inducido químicamente , Trastornos del Metabolismo de los Lípidos/metabolismo , Hígado/metabolismo , Masculino , PPAR gamma , Ratas , Ratas Sprague-Dawley , Sirtuina 1/metabolismo
15.
Food Chem Toxicol ; 165: 113119, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35537648

RESUMEN

Di-2-ethylhexyl phthalate (DEHP) has been widely used in many fields (agricultural products, medical instruments, and food packing). As an environmental contaminant, DEHP has a negative impact on human and animal health, and thus toxicity caused by DEHP is increasingly serious health concern. Nevertheless, DEHP-induced liver damage in quail remains unclear. To investigate the mechanism of liver damage caused by DEHP, male quail were treated with DEHP (250, 500, and 750 mg/kg) by gavage. Notably, DEHP exposure results in increased blood lipids and the accumulation of triglycerides (TG), total cholesterol (TC), and non-esterified fatty acid (NEFA) in the liver. Histopathological analysis showed that steatosis and inflammatory cell infiltration were observed in the liver tissue of quails exposed to DEHP. The results of Oil Red O staining displayed that DEHP induced lipid storage in the liver. Moreover, DEHP induced lipid metabolism disorders by activating the LXR/SREBP-1c and PPARα/γ signaling pathway. DEHP exposure obviously caused the up-regulation of pro-inflammatory cytokines (NF-κB, IL-6, IL-8, IL-1ß, and TNF-a). This study showed that DEHP could induce lipid metabolism disorders and inflammatory response via LXR/SREBP-1c/PPARα/γ and NF-κB signaling pathways.


Asunto(s)
Dietilhexil Ftalato , Trastornos del Metabolismo de los Lípidos , Animales , Dietilhexil Ftalato/metabolismo , Dietilhexil Ftalato/toxicidad , Metabolismo de los Lípidos , Trastornos del Metabolismo de los Lípidos/inducido químicamente , Trastornos del Metabolismo de los Lípidos/metabolismo , Trastornos del Metabolismo de los Lípidos/patología , Hígado/metabolismo , Masculino , FN-kappa B/genética , FN-kappa B/metabolismo , PPAR alfa/genética , PPAR alfa/metabolismo , Ácidos Ftálicos , Transducción de Señal , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo
16.
Life Sci ; 294: 120379, 2022 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-35134438

RESUMEN

AIMS: Fluorene-9-bisphenol (BHPF), as a substitute for bisphenol A, is used in many industries in daily life. Many studies have clarified its effects as an endocrine disruptor on organisms, but its effect on lipid metabolism of zebrafish larvae is not clear. Patients with non-alcoholic fatty liver disease (NAFLD) are more susceptible to external pollutants. It is not clear how BHPF perturbs lipid metabolism or promotes NAFLD progression. MAIN METHODS: We explored the biological effects of BHPF on locomotor activity, inflammatory response, endoplasmic reticulum (ER) stress and lipid metabolism in zebrafish, especially in the mechanism of lipid homeostasis disorder. In addition, the role of BHPF in the progression of non-alcoholic fatty liver disease (NAFLD) was further explored. KEY FINDINGS: We found that high concentration (100 nmol/L) BHPF caused retarded growth, mild lipid accumulation and reduced the locomotive activity of zebrafish larvae, accompanied by a decrease in endogenous cortisol level. At the same time, it caused the full activation of inflammation and ER stress. Rescue experiments by 25(OH)D3 demonstrated that high concentration of BHPF caused defects in 1,25(OH)2D3 metabolic pathway through downregulation of cyp2r1, which further damaged pgc1a-mediated fatty acid oxidation and mitochondrial function, resulting in lipid accumulation. In summary, exposure to BHPF could damage lipid homeostasis and worsen the diet-induced NAFLD. SIGNIFICANCE: Our findings provide new insights into the role of BHPF in development of overweight and obesity and also improve understanding of its toxicological mechanism. Our results play a warning role in the administration of environmental pollutants.


Asunto(s)
Compuestos de Bencidrilo/toxicidad , Ácidos Grasos/química , Fluorenos/toxicidad , Larva/efectos de los fármacos , Trastornos del Metabolismo de los Lípidos/patología , Locomoción/efectos de los fármacos , Fenoles/toxicidad , Animales , Larva/metabolismo , Trastornos del Metabolismo de los Lípidos/inducido químicamente , Trastornos del Metabolismo de los Lípidos/metabolismo , Pez Cebra
17.
Clin Nutr ; 41(1): 21-32, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34864452

RESUMEN

BACKGROUND & AIMS: Folic acid supplementation is widely accepted during pregnancy, as it exerts a protective effect on neural tube defects. However, the long-term underlying effects of folic acid supplementation during pregnancy (FASDP) on offspring remain unclear. METHODS: Thirty pregnant female rats were randomly divided into normal control group, folic acid appropriate supplementation group (2.5 × FA group) and folic acid oversupplementation group (5 × FA group) and fed with corresponding folic acid concentration AIN93G diet. UPLC-Q-TOF-MS, UPLC-TQ-MS and GC-MS were performed to detect the serum metabolites profiles in adult male offspring and explore the effects of FASDP. Moreover, molecular biology technologies were used to clarify the underlying mechanism. RESULTS: We demonstrate that 2.5-folds folic acid leads to dyslipidemic-diabetic slightly in male offspring, while 5-folds folic acid aggravates the disorder and prominent hepatic lipid accumulations. Using untargeted and targeted metabolomics, total 63 differential metabolites and 12 significantly differential KEGG pathways are identified. Of note, arginine biosynthesis, arginine and proline metabolism are the two most significant pathways. Mechanistic investigations reveal that the increased levels of arginase-1 (Arg1) causes the lipid metabolism disorder by regulating nitric oxide synthase-3 (NOS3)-adenosine monophosphate activated protein kinase-α (AMPKα) pathway, resulting in lipid accumulation in hepatocytes. CONCLUSIONS: Our data suggest that maternal folic acid oversupplementation during pregnancy contributes to lipid metabolism disorder in male offspring by regulating Arg1-NOS3-AMPKα pathway.


Asunto(s)
Arginasa/metabolismo , Suplementos Dietéticos/efectos adversos , Ácido Fólico/efectos adversos , Trastornos del Metabolismo de los Lípidos/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Dieta/métodos , Femenino , Ácido Fólico/administración & dosificación , Cromatografía de Gases y Espectrometría de Masas , Hepatocitos/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Hígado/metabolismo , Masculino , Metabolómica , Óxido Nítrico Sintasa de Tipo III/metabolismo , Embarazo , Ratas , Transducción de Señal/efectos de los fármacos
18.
Biol Trace Elem Res ; 200(6): 2767-2776, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-34392477

RESUMEN

Estrogen exerts essential role in liver metabolism, and its deficiency is frequently accompanied by a series of metabolic disorder diseases. To investigate the role of estrogen deficiency in fluorine ions (F-) induced liver injury, the ovariectomy (OVX) rat models were performed by surgically removing the ovaries, and the rats from OVX and non-OVX models were exposed to differential dose of F- (0, 25, 50 and 100 mg/L) in drinking water for 90 days. The liver morphological structure was evaluated by hematoxylin-eosin staining. Proliferation ability of hepatocytes was evaluated by 5-bromo-2-deoxyuridine (BrdU) assay. And distribution of lipid droplets in liver tissue was observed via oil red O staining. In addition, the liver function and lipid metabolism parameters in serum were detected by commercial kits. Results showed that F- induced hepatocytes morphological damage and inhibited the proliferation ability of hepatocytes; estrogen deficiency exacerbated these changes. The deposition of lipid droplets in the liver tissue was multiplicative with increased F- dose, especially after estrogen deficiency. In addition, F- exposure increased (P < 0.05 or P < 0.01) serum aminotransferase (ALT), aminotransferase (AST), alkaline phosphatase (ALP), and γ-glutamyl transpeptidase (γ-GT) activities and total bilirubin (T-bil) level; meanwhile, serum triglyceride (TG) and cholesterol (TC) levels were also elevated (P < 0.05 or P < 0.01). F--induced liver function and lipid metabolism indexes were further increased (P < 0.05 or P < 0.01) in the state of estrogen deficiency. In conclusion, estrogen deficiency aggravated F--induced liver damage and lipid metabolism disorder.


Asunto(s)
Trastornos del Metabolismo de los Lípidos , Metabolismo de los Lípidos , Animales , Estrógenos/metabolismo , Femenino , Fluoruros/metabolismo , Trastornos del Metabolismo de los Lípidos/inducido químicamente , Trastornos del Metabolismo de los Lípidos/metabolismo , Hígado/metabolismo , Ratas , Ratas Sprague-Dawley , Transaminasas/metabolismo
19.
Food Funct ; 12(11): 4825-4841, 2021 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-33949580

RESUMEN

Large yellow croaker roe phospholipids (LYCRPLs) have great nutritional value because they are rich in docosahexaenoic acid (DHA), which is an n-3 polyunsaturated fatty acid (n-3 PUFA). In previous research, we studied the effect of LYCRPLs on the inhibition of triglyceride accumulation at the cellular level. However, its lipid regulation effect in rats on a high-fat diet and its influence on the gut microbiota has not yet been clarified. In this study, a high-fat diet was used to induce the lipid metabolism disorder in SD rats, and simvastatin, low-dose, medium-dose and high-dose LYCRPLs were given by intragastric administration for 8 weeks. The rats' body weight, food intake, organ index, blood biochemical indicators, epididymal fat tissue and liver histopathology were compared and analyzed. High-throughput 16S rRNA gene sequencing technology and bioinformatics analysis technology were also used to analyze the diversity of gut microbiota in rats. We found that LYCRPLs can significantly regulate lipid metabolism, and improve the gut microbiota disorder induced in rats by a high-fat diet. These results can lay a foundation for the study of the regulation mechanism of LYCRPLs lipid metabolism, and also provide a theoretical basis for the development of LYCRPLs as functional food additives and excipients with hypolipidemic effects.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Ácidos Docosahexaenoicos/farmacología , Microbioma Gastrointestinal/efectos de los fármacos , Trastornos del Metabolismo de los Lípidos/tratamiento farmacológico , Perciformes/metabolismo , Fosfolípidos/farmacología , Animales , Peso Corporal , Aditivos Alimentarios/metabolismo , Alimentos Funcionales , Metabolismo de los Lípidos/efectos de los fármacos , Trastornos del Metabolismo de los Lípidos/inducido químicamente , Hígado/metabolismo , Masculino , ARN Ribosómico 16S/genética , Ratas , Ratas Sprague-Dawley , Triglicéridos/metabolismo
20.
J Agric Food Chem ; 68(41): 11468-11479, 2020 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-32962341

RESUMEN

Di(2-ethylhexyl) phthalate (DEHP) is a widespread pollutant that badly affects animals and human health. Lycopene (LYC) has been used as a dietary supplement that has effective antioxidant and antiobesity functions. The present goal was to understand the molecular mechanisms of LYC preventing DEHP-induced lipid metabolism of the liver. The mice were intragastrically administered with LYC (5 mg/kg) and/or DEHP (500 mg/kg or 1000 mg/kg). Here, we found that LYC attenuated DEHP-caused hepatic histopathological lesions including steatosis. Hematological and biochemical analyses revealed that LYC ameliorated DEHP-caused liver function and lipid metabolism disorders. DEHP caused lipid metabolism disorders via activating the peroxisome proliferator activated receptor α/γ (PPARα/γ) signal transducer and Farnesoid X receptor (FXR)/liver X receptor (LXR) signaling pathway. As a major regulator of lipid metabolism, hypoxia-inducible factor-1α (HIF-1α) system was elevated with increased fatty degeneration under DEHP exposure. However, LYC could decrease the levels of HIF-1α/PPARα/PPARγ/FXR/LXR signaling pathway-related factors. Our research indicated that LYC could prevent DEHP-induced lipid metabolism disorders via inhibiting the HIF-1α-mediated PPARα/PPARγ/FXR/LXR system. This study may provide a possible molecular mechanism for fatty liver induced by DEHP.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Trastornos del Metabolismo de los Lípidos/tratamiento farmacológico , Hepatopatías/tratamiento farmacológico , Receptores X del Hígado/metabolismo , Licopeno/administración & dosificación , PPAR alfa/metabolismo , PPAR gamma/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Antioxidantes , Dietilhexil Ftalato/efectos adversos , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Metabolismo de los Lípidos/efectos de los fármacos , Trastornos del Metabolismo de los Lípidos/inducido químicamente , Trastornos del Metabolismo de los Lípidos/genética , Trastornos del Metabolismo de los Lípidos/metabolismo , Hepatopatías/etiología , Hepatopatías/genética , Hepatopatías/metabolismo , Receptores X del Hígado/genética , Masculino , Ratones , Ratones Endogámicos ICR , PPAR alfa/genética , PPAR gamma/genética , Receptores Citoplasmáticos y Nucleares/genética , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...