Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Br J Nutr ; 131(4): 553-566, 2024 02 28.
Article in English | MEDLINE | ID: mdl-37699661

ABSTRACT

Sterol regulatory element-binding protein 2 (SREBP2) is considered to be a major regulator to control cholesterol homoeostasis in mammals. However, the role of SREBP2 in teleost remains poorly understand. Here, we explored the molecular characterisation of SREBP2 and identified SREBP2 as a key modulator for 3-hydroxy-3-methylglutaryl-coenzyme A reductase and 7-dehydrocholesterol reductase, which were rate-limiting enzymes of cholesterol biosynthesis. Moreover, dietary palm oil in vivo or palmitic acid (PA) treatment in vitro elevated cholesterol content through triggering SREBP2-mediated cholesterol biosynthesis in large yellow croaker. Furthermore, our results also found that PA-induced activation of SREBP2 was dependent on the stimulating of endoplasmic reticulum stress (ERS) in croaker myocytes and inhibition of ERS by 4-Phenylbutyric acid alleviated PA-induced SREBP2 activation and cholesterol biosynthesis. In summary, our findings reveal a novel insight for understanding the role of SREBP2 in the regulation of cholesterol metabolism in fish and may deepen the link between dietary fatty acid and cholesterol biosynthesis.


Subject(s)
Dietary Fats, Unsaturated , Perciformes , Animals , Cholesterol/metabolism , Endoplasmic Reticulum Stress , Muscles/metabolism , Palm Oil/pharmacology , Perciformes/metabolism , Sterol Regulatory Element Binding Protein 2/genetics , Sterol Regulatory Element Binding Protein 2/metabolism
2.
Fish Shellfish Immunol ; 141: 109031, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37640122

ABSTRACT

Glycerol monolaurate (GML) is a potential candidate for regulating metabolic syndrome and inflammatory response. However, the role of GML in modulating intestinal health in fish has not been well determined. In this study, a 70-d feeding trial was conducted to evaluate the effect of GML on intestinal barrier, antioxidant capacity, inflammatory response and microbiota community of large yellow croaker (13.05 ± 0.09 g) fed with high level soybean oil (SO) diets. Two basic diets with fish oil (FO) or SO were formulated. Based on the SO group diet, three different levels of GML 0.02% (SO0.02), 0.04% (SO0.04) and 0.08% (SO0.08) were supplemented respectively. Results showed that intestinal villus height and perimeter ratio were increased in SO0.04 treatment compared with the SO group. The mRNA expressions of intestinal physical barrier-related gene odc and claudin-11 were significantly up-regulated in different addition of GML treatments compared with the SO group. Fish fed SO diet with 0.04% GML addition showed higher activities of acid phosphatase and lysozyme compared with the SO group. The content of malonaldehyde was significantly decreased and activities of catalase and superoxide dismutase were significantly increased in 0.02% and 0.04% GML groups compared with those in the SO group. The mRNA transcriptional levels of inflammatory response-related genes (il-1ß, il-6, tnf-α and cox-2) in 0.04% GML treatment were notably lower than those in the SO group. Meanwhile, sequencing analysis of bacterial 16S rRNA V4-V5 region showed that GML addition changed gut microbiota structure and increased alpha diversity of large yellow croaker fed diets with a high level of SO. The correlation analysis results indicated that the change of intestinal microbiota relative abundance strongly correlated with intestinal health indexes. In conclusion, these results demonstrated that 0.02%-0.04% GML addition could improve intestinal morphology, physical barrier, antioxidant capacity, inflammatory response and microbiota dysbiosis of large yellow croaker fed diets with a high percentage of SO.


Subject(s)
Microbiota , Perciformes , Animals , Antioxidants/metabolism , Soybean Oil/metabolism , Dysbiosis , RNA, Ribosomal, 16S , Diet/veterinary , Perciformes/genetics , RNA, Messenger/metabolism , Animal Feed/analysis
3.
Article in English | MEDLINE | ID: mdl-36990141

ABSTRACT

3-hydroxyacyl-CoA dehydratases 1 (Hacd1) is a critical enzyme in long-chain polyunsaturated fatty acids (LC-PUFA) biosynthesis. The difference in expression of hacd1 might account for the stronger capacity of LC-PUFA biosynthesis in freshwater fish than in marine fish, but little is known about fish hacd1. Therefore, this study compared the responses of large yellow croaker and rainbow trout hacd1 to different oil sources or fatty acids, and also examined transcriptional regulation of this gene. In this study, hacd1 was highly expressed in the liver of large yellow croaker and rainbow trout, which is the main organ for LC-PUFA biosynthesis. Therefore, we cloned the hacd1 coding sequence, with a phylogenetic analysis showing that this gene is evolutionarily conserved. Its localization to the endoplasmic reticulum (ER), likely also indicates a conserved structure and function. The expression of hacd1 in the liver was significantly decreased after the substitution of soybean oil (SO) for fish oil but was not significantly affected after palm oil (PO) substitution. Linoleic acid (LA) incubation significantly promoted hacd1 expression in primary hepatocytes of large yellow croaker and eicosapentaenoic acid (EPA) incubation significantly promoted hacd1 expression in primary hepatocytes of rainbow trout. Transcription factors STAT4, C/EBPα, C/EBPß, HNF1, HSF3 and FOXP3 were identified in both large yellow croaker and rainbow trout. HNF1 had a stronger activation effect in rainbow trout than in large yellow croaker. FOXP3 inhibited hacd1 promoter activity in large yellow croaker but had no effect in rainbow trout. Therefore, the differences between HNF1 and FOXP3 affected the expression of hacd1 in the liver thus being responsible for the high capacity of LC-PUFA biosynthesis in rainbow trout.


Subject(s)
Oncorhynchus mykiss , Perciformes , Animals , Oncorhynchus mykiss/genetics , Oncorhynchus mykiss/metabolism , Phylogeny , Fatty Acids/metabolism , Perciformes/genetics , Perciformes/metabolism , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism
4.
Br J Nutr ; 129(1): 29-40, 2023 01 14.
Article in English | MEDLINE | ID: mdl-35473947

ABSTRACT

Dietary l-carnitine (LC) is a nutritional factor that reduces liver lipid content. However, whether dietary LC can improve lipid metabolism via simultaneous activation of mitochondrial fatty acid (FA) ß-oxidation and suppression of endoplasmic reticulum (ER) stress is still unknown. Large yellow croaker were fed with a high-fat diet (HFD) supplemented with dietary LC at 0, 1·2 or 2·4 ‰ for 10 weeks. The results indicated that a HFD supplemented with LC reduced the liver total lipid and TAG content and improved serum lipid profiles. LC supplementation administered to this fish increased the liver antioxidant capacity by decreasing serum and liver malondialdehyde levels and enhancing the liver antioxidant capacity, which then relieved the liver damage. Dietary LC increased the ATP dynamic process and mitochondrial number, decreased mitochondrial DNA damage and enhanced the protein expression of mitochondrial ß-oxidation, biogenesis and mitophagy. Furthermore, dietary LC supplementation increased the expression of genes and proteins related to peroxisomal ß-oxidation and biogenesis. Interestingly, feeding fish with LC-enriched diets decreased the protein levels indicative of ER stress, such as glucose-regulated protein 78, p-eukaryotic translational initiation factor 2a and activating transcription factor 6. Dietary LC supplementation downregulated mRNA expression relative to FA synthesis, reduced liver lipid and relieved liver damage through regulating ß-oxidation and biogenesis of mitochondria and peroxisomes, as well as the ER stress pathway in fish fed with HFD. The present study provides the first evidence that dietary LC can improve lipid metabolism via simultaneously promoting FA ß-oxidation capability and suppressing the ER stress pathway in fish.


Subject(s)
Lipid Metabolism , Perciformes , Animals , Diet, High-Fat/adverse effects , Antioxidants/metabolism , Carnitine/metabolism , Liver/metabolism , Fatty Acids/metabolism , Perciformes/genetics , Endoplasmic Reticulum Stress , Lipids
5.
Antioxidants (Basel) ; 11(12)2022 Nov 28.
Article in English | MEDLINE | ID: mdl-36552565

ABSTRACT

Dietary intake of omega-3 fatty acids found in fish has been reported to reduce the risk of Alzheimer's Disease (AD). Stearidonic acid (SDA), a plant-based omega-3 fatty acid, has been targeted as a potential surrogate for fish-based fatty acids. However, its role in neuronal degeneration is unknown. This study was designed to evaluate effects of SDA on Amyloid-ß(A-ß)-induced neurotoxicity in rat hippocampal cells. Results showed that SDA effectively converted to eicosapentaenoic acid (EPA) in hippocampal cells. Aß-induced apoptosis in H19-7 cells was protected by SDA pretreatment as evidenced by its regulation on the expression of relevant pro- and anti-apoptotic genes, as well as the inhibition on caspase activation. SDA also protected H19-7 cells from Aß-induced oxidative stress by regulating the expression of relevant pro- and anti-oxidative genes, as well as the improvement in activity of catalase. As for Aß/LPS-induced neuronal inflammation, SDA pretreatment reduced the release of IL-1ß and TNFα. Further, we found that the anti-Aß effect of SDA involves its inhibition on the expression of amyloid precursor protein and the regulation on MAPK signaling. These results demonstrated that SDAs have neuroprotective effect in Aß-induced H19-7 hippocampal cells. This beneficial effect of SDA was attributed to its antiapoptotic, antioxidant, and anti-inflammatory properties.

6.
Fish Shellfish Immunol ; 128: 50-59, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35843522

ABSTRACT

A 70-day feeding trial was conducted to investigate effects of dietary lysolecithin on growth performance, serum biochemical indexes, antioxidant capacity, lipid metabolism and inflammation-related genes expression of juvenile large yellow croaker (Larimichthys crocea) with initial weight of 6.04 ± 0.08 g. A formulated diet containing approximately 42% crude protein and 12.5% crude lipid was used as the control diet (CON). The other three experimental diets were formulated with supplementation of 0.2%, 0.4% and 0.6% lysolecithin based on the control diet, respectively. Results showed that weight gain rate (WGR) and specific growth rate (SGR) significantly increased in fish fed diets with lysolecithin compared with those in the control diet (P < 0.05). Fish fed diets with 0.4% and 0.6% lysolecithin had notably higher lipid content in muscle than that in the control diet (P < 0.05). When fish were fed diets with lysolecithin, serum high-density lipoprotein cholesterol (HDL-c) content was notably higher than that in the control diet (P < 0.05), while fish fed the diet with 0.6% lysolecithin had a significant lower serum low-density lipoprotein cholesterol (LDL-c) content than that in the control diet (P < 0.05). Meanwhile, serum aspartate transaminase (AST) and alanine transaminase (ALT) activities in fish fed diets with lysolecithin were remarkably lower than those in the control diet (P < 0.05). With the increase of dietary lysolecithin from 0.2% to 0.6%, mRNA expression of stearoyl-coenzyme A desaturase 1 (scd1), diacylglycerol acyltransferase 2 (dgat2) and sterol-regulatory element binding protein 1 (srebp1) showed decreasing trends. Furthermore, mRNA expression of carnitine palmitoyl transferase 1 (cpt1) and lipoprotein lipase (lpl) among each dietary lysolecithin treatment were significantly higher than those in the control diet (P < 0.05). In terms of inflammation, mRNA expression of tumor necrosis factor α (tnf-α) and interleukin-1 ß (il-1ß) were significantly down-regulated in fish fed diets with lysolecithin compared with those in the control diet (P < 0.05), while the mRNA expression of interleukin-10 (il-10) was significantly higher than that in the control diet (P < 0.05). In conclusion, dietary lysolecithin could promote the growth performance, improve hepatic lipid metabolism and regulate inflammation response in juvenile large yellow croaker, and the optimal supplement level of lysolecithin was approximately 0.4% in this study.


Subject(s)
Lipid Metabolism , Perciformes , Alanine Transaminase/metabolism , Animal Feed/analysis , Animals , Antioxidants/metabolism , Aspartate Aminotransferases/metabolism , Carnitine/metabolism , Cholesterol, LDL/metabolism , Diacylglycerol O-Acyltransferase/genetics , Diet/veterinary , Dietary Supplements , Fatty Acid Desaturases/metabolism , Inflammation/veterinary , Interleukin-10/metabolism , Interleukin-1beta/metabolism , Lipoprotein Lipase , Lipoproteins, HDL , Lysophosphatidylcholines/metabolism , Perciformes/metabolism , RNA, Messenger/metabolism , Tumor Necrosis Factor-alpha/metabolism
7.
Fish Shellfish Immunol ; 126: 12-20, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35526799

ABSTRACT

LPCAT3, a subtype of lysophosphatidylcholine acyltransferases, is a key enzyme in phosphatidylcholine remodeling pathway and plays a significant role in mediating inflammatory response in mammals. However, its inflammatory function in fish has yet to be discovered. Herein, this study aimed to investigate its role in inflammation in Larimichthys crocea. We analyzed the coding sequence of Larimichthys crocea LPCAT3 (Lc-LPCAT3) and explored the effect of Lc-LPCAT3 on palmitate (PA)-induced inflammation. We found that in macrophage cell line of Larimichthys crocea, the mRNA expression of Lc-lpcat3 was upregulated by PA with the elevated pro-inflammatory genes expression, including il1ß, il6, il8, tnfα and ifnγ. Next, the role of Lc-LPCAT3 in inflammation induced by PA was further investigated. Results showed that knockdown of Lc-LPCAT3 mitigated PA-induced pro-inflammatory genes mRNA expression, including il1ß, il8, tnfα and ifnγ, in which JNK signaling pathway was involved. In contrast, overexpression of Lc-LPCAT3 induced pro-inflammatory genes expression including il1ß, tnfα and ifnγ. Furthermore, several transcription factors with negative regulation of Lc-LPCAT3 promoter activity were discovered including LXRα, RXRα, PPARα, PPARγ, CEBPα, CEBPß, CEBPδ, SREBP1 and SREBP2, and SREBP1 had the strongest regulatory effect. In conclusion, we first discovered that fish LPCAT3 participated in PA-induced inflammation, and targeting SREBP1 might be an effective coping strategy.


Subject(s)
1-Acylglycerophosphocholine O-Acyltransferase , Perciformes , 1-Acylglycerophosphocholine O-Acyltransferase/genetics , 1-Acylglycerophosphocholine O-Acyltransferase/metabolism , Animals , Fish Proteins/genetics , Fish Proteins/metabolism , Inflammation/chemically induced , Inflammation/genetics , Inflammation/veterinary , Interleukin-8 , Macrophages/metabolism , Mammals/genetics , Mammals/metabolism , Palmitates/metabolism , Perciformes/genetics , Perciformes/metabolism , RNA, Messenger/metabolism , Tumor Necrosis Factor-alpha/metabolism
8.
Int J Biol Macromol ; 194: 153-162, 2022 Jan 01.
Article in English | MEDLINE | ID: mdl-34863827

ABSTRACT

Interferon gamma (IFN-γ) is a widely expressed cytokine that has potent antiviral and immunomodulatory effects. The expression and bioactivity of IFN-γ have been reported in several fish species. However, the molecular mechanism mediated by IFN-γ in fish macrophages has not been completely elucidated. This study used the macrophage cell line to investigate the functional activities and regulation mechanism of large yellow croaker IFN-γ (LcIFN-γ). Herein, the mRNA expression of Lcifn-γ was significantly upregulated in macrophages after LPS and poly(I:C) treatment. Recombinant LcIFN-γ protein (rLcIFN-γ) significantly enhanced the phagocytic ability and respiratory burst activity of macrophages. Meanwhile, rLcIFN-γ induced M1 phenotype polarization of macrophages with the upregulated expressions of pro-inflammatory gene. Moreover, rLcIFN-γ upregulated the IFN-stimulated genes (ISGs) expression and activated JAK (Janus tyrosine kinases)-STAT (signal transducer and activator of transcription) signaling pathway by causing the phosphorylation of JAK1 and STAT1Tyr701. Furthermore, the promoter activity of IFN-regulatory factor 1 (IRF1) was significantly upregulated by the phosphorylated transcription factor STAT1 through binding to its promoter region. In addition to the classical JAK-STAT pathway, rLcIFN-γ also activated multiple distinct signaling cascades such as mitogen-activated protein kinase (MAPK) and protein kinase B (AKT) pathways. Overall, this study indicated the powerful effects of LcIFN-γ on macrophage activation of large yellow croaker and its molecular mechanism.


Subject(s)
Interferon-gamma/metabolism , Macrophages/immunology , Macrophages/metabolism , Perciformes/physiology , Animals , Gene Expression , Interferon-gamma/genetics , Janus Kinases/metabolism , Lipopolysaccharides/immunology , Macrophage Activation/immunology , Phagocytosis , Protein Binding , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , STAT Transcription Factors/metabolism
9.
J Hazard Mater ; 419: 126454, 2021 10 05.
Article in English | MEDLINE | ID: mdl-34198221

ABSTRACT

Nanoplastics (NPs) cause various adverse effects on marine fish. However, effects of dietary NPs exposure on liver lipid metabolism and muscle nutritional quality of carnivorous marine fish are not fully understood. In this study, a 21-day feeding test was conducted to simulate the food chain transfer of polystyrene nanoplastics (PS NPs) and then evaluate effects of different dietary PS NPs levels on the survival, growth performance, liver lipid metabolism, and muscle nutritional quality of large yellow croaker Larimichthys crocea. Results indicated that the survival and growth of large yellow croaker decreased with the increase of PS NPs levels. Moreover, PS NPs induced excessive liver lipid accumulation by down-regulating the expression of lipolysis-related genes and inhibiting the AMPK-PPARα signaling pathway. In vitro, PS NPs could be accumulated in hepatocytes, reduce cell viability, and disrupt lipid metabolism of hepatocytes. Also, we found for the first time that PS NPs altered fatty acid composition and texture of fish muscle by enhancing oxidative stress and disrupting lipid metabolism. Overall, this study indicated that PS NPs induced liver lipid deposition by inhibiting lipolysis, and demonstrated that PS NPs altered the nutritional quality of fish, which might cause potential health effects for human consumers.


Subject(s)
Lipid Metabolism , Perciformes , Animals , Fatty Acids/metabolism , Humans , Liver/metabolism , Microplastics , Muscles/metabolism , Nutritive Value , Polystyrenes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...