Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 4214, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38760332

ABSTRACT

The liver gene expression of the peroxisomal ß-oxidation enzyme acyl-coenzyme A oxidase 1 (ACOX1), which catabolizes very long chain fatty acids (VLCFA), increases in the context of obesity, but how this pathway impacts systemic energy metabolism remains unknown. Here, we show that hepatic ACOX1-mediated ß-oxidation regulates inter-organ communication involved in metabolic homeostasis. Liver-specific knockout of Acox1 (Acox1-LKO) protects mice from diet-induced obesity, adipose tissue inflammation, and systemic insulin resistance. Serum from Acox1-LKO mice promotes browning in cultured white adipocytes. Global serum lipidomics show increased circulating levels of several species of ω-3 VLCFAs (C24-C28) with previously uncharacterized physiological role that promote browning, mitochondrial biogenesis and Glut4 translocation through activation of the lipid sensor GPR120 in adipocytes. This work identifies hepatic peroxisomal ß-oxidation as an important regulator of metabolic homeostasis and suggests that manipulation of ACOX1 or its substrates may treat obesity-associated metabolic disorders.


Subject(s)
Acyl-CoA Oxidase , Liver , Mice, Knockout , Obesity , Animals , Liver/metabolism , Mice , Acyl-CoA Oxidase/metabolism , Acyl-CoA Oxidase/genetics , Obesity/metabolism , Obesity/genetics , Male , Insulin Resistance , Mice, Inbred C57BL , Oxidation-Reduction , Lipid Metabolism , Adipose Tissue/metabolism , Diet, High-Fat , Energy Metabolism , Fatty Acids/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics
2.
Nat Commun ; 15(1): 3213, 2024 Apr 13.
Article in English | MEDLINE | ID: mdl-38615060

ABSTRACT

Oxidative stress-induced lipid accumulation is mediated by lipid droplets (LDs) homeostasis, which sequester vulnerable unsaturated triglycerides into LDs to prevent further peroxidation. Here we identify the upregulation of lipopolysaccharide-binding protein (LBP) and its trafficking through LDs as a mechanism for modulating LD homeostasis in response to oxidative stress. Our results suggest that LBP induces lipid accumulation by controlling lipid-redox homeostasis through its lipid-capture activity, sorting unsaturated triglycerides into LDs. N-acetyl-L-cysteine treatment reduces LBP-mediated triglycerides accumulation by phospholipid/triglycerides competition and Peroxiredoxin 4, a redox state sensor of LBP that regulates the shuttle of LBP from LDs. Furthermore, chronic stress upregulates LBP expression, leading to insulin resistance and obesity. Our findings contribute to the understanding of the role of LBP in regulating LD homeostasis and against cellular peroxidative injury. These insights could inform the development of redox-based therapies for alleviating oxidative stress-induced metabolic dysfunction.


Subject(s)
Acute-Phase Proteins , Lipid Droplets , Membrane Glycoproteins , Acute-Phase Proteins/metabolism , Carrier Proteins/metabolism , Homeostasis , Lipid Droplets/metabolism , Lipopolysaccharides/metabolism , Membrane Glycoproteins/metabolism , Oxidative Stress/genetics , Oxidative Stress/physiology , Triglycerides
3.
Mol Metab ; 82: 101913, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38458567

ABSTRACT

OBJECTIVE: Adipose tissue mass is maintained by a balance between lipolysis and lipid storage. The contribution of adipose tissue lipogenesis to fat mass, especially in the setting of high-fat feeding, is considered minor. Here we investigated the effect of adipose-specific inactivation of the peroxisomal lipid synthetic protein PexRAP on fatty acid synthase (FASN)-mediated lipogenesis and its impact on adiposity and metabolic homeostasis. METHODS: To explore the role of PexRAP in adipose tissue, we metabolically phenotyped mice with adipose-specific knockout of PexRAP. Bulk RNA sequencing was used to determine transcriptomic responses to PexRAP deletion and 14C-malonyl CoA allowed us to measure de novo lipogenic activity in adipose tissue of these mice. In vitro cell culture models were used to elucidate the mechanism of cellular responses to PexRAP deletion. RESULTS: Adipose-specific PexRAP deletion promoted diet-induced obesity and insulin resistance through activation of de novo lipogenesis. Mechanistically, PexRAP inactivation inhibited the flux of carbons to ethanolamine plasmalogens. This increased the nuclear PC/PE ratio and promoted cholesterol mislocalization, resulting in activation of liver X receptor (LXR), a nuclear receptor known to be activated by increased intracellular cholesterol. LXR activation led to increased expression of the phospholipid remodeling enzyme LPCAT3 and induced FASN-mediated lipogenesis, which promoted diet-induced obesity and insulin resistance. CONCLUSIONS: These studies reveal an unexpected role for peroxisome-derived lipids in regulating LXR-dependent lipogenesis and suggest that activation of lipogenesis, combined with dietary lipid overload, exacerbates obesity and metabolic dysregulation.


Subject(s)
Insulin Resistance , Lipogenesis , Animals , Mice , 1-Acylglycerophosphocholine O-Acyltransferase/metabolism , Adipose Tissue/metabolism , Cholesterol/metabolism , Dietary Fats/metabolism , Lipogenesis/genetics , Liver X Receptors/metabolism , Mice, Knockout , Obesity/metabolism
4.
Biochem Biophys Res Commun ; 702: 149655, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38340654

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) is one of the most common causes of liver disease worldwide. MTARC1, encoded by the MTARC1 gene, is a mitochondrial outer membrane-anchored enzyme. Interestingly, the MTARC1 p.A165T (rs2642438) variant is associated with a decreased risk of NAFLD, indicating that MTARC1 might be an effective target. It has been reported that the rs2642438 variant does not have altered enzymatic activity so we reasoned that this variation may affect MTARC1 stability. In this study, MTARC1 mutants were generated and stability was assessed using a protein stability reporter system both in vitro and in vivo. We found that the MTARC1 p.A165T variant has dramatically reduced the stability of MTARC1, as assessed in several cell lines. In mice, the MTARC1 A168T mutant, the equivalent of human MTARC1 A165T, had diminished stability in mouse liver. Additionally, several MTARC1 A165 mutants, including A165S, A165 N, A165V, A165G, and A165D, had dramatically decreased stability as well, suggesting that the alanine residue of MTARC1 165 site is essential for MTARC1 protein stability. Collectively, our data indicates that the MTARC1 p.A165T variant (rs2642438) leads to reduced stability of MTARC1. Given that carriers of rs2642438 show a decreased risk of NAFLD, the findings herein support the notion that MTARC1 inhibition may be a therapeutic target to combat NAFLD.


Subject(s)
Non-alcoholic Fatty Liver Disease , Animals , Humans , Mice , Lipid Metabolism , Liver/metabolism , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Protein Stability
5.
Biochem Biophys Res Commun ; 677: 63-69, 2023 Oct 15.
Article in English | MEDLINE | ID: mdl-37549603

ABSTRACT

The mammalian cell cycle is divided into four sequential phases, namely G1 (Gap 1), S (synthesis), G2 (Gap 2), and M (mitosis). Wee1, whose turnover is tightly and finely regulated, is a well-known kinase serving as a gatekeeper for the G2/M transition. However, the mechanism underlying the turnover of Wee1 is not fully understood. Autophagy, a highly conserved cellular process, maintains cellular homeostasis by eliminating intracellular aggregations, damaged organelles, and individual proteins. In the present study, we found autophagy deficiency in mouse liver caused G2/M arrest in two mouse models, namely Fip200 and Atg7 liver-specific knockout mice. To uncover the link between autophagy deficiency and G2/M transition, we combined transcriptomic and proteomic analysis for liver samples from control and Atg7 liver-specific knockout mice. The data suggest that the inhibition of autophagy increases the protein level of Wee1 without any alteration of its mRNA abundance. Serum starvation, an autophagy stimulus, downregulates the protein level of Wee1 in vitro. In addition, the half-life of Wee1 is extended by the addition of chloroquine, an autophagy inhibitor. LC3, a central autophagic protein functioning in autophagy substrate selection and autophagosome biogenesis, interacts with Wee1 as assessed by co-immunoprecipitation assay. Furthermore, overexpression of Wee1 leads to G2/M arrest both in vitro and in vivo. Collectively, our data indicate that autophagy could degrade Wee1-a gatekeeper of the G2/M transition, whereas the inhibition of autophagy leads to the accumulation of Wee1 and causes G2/M arrest in mouse liver.


Subject(s)
Apoptosis , Proteomics , Mice , Animals , Protein-Tyrosine Kinases/metabolism , Nuclear Proteins/metabolism , Cell Line, Tumor , G2 Phase Cell Cycle Checkpoints , Cell Cycle/physiology , Cell Cycle Proteins/metabolism , Mitosis , Autophagy , Mice, Knockout , Mammals/metabolism
6.
Methods Mol Biol ; 2662: 25-31, 2023.
Article in English | MEDLINE | ID: mdl-37076668

ABSTRACT

Brown adipose tissue (BAT) is an important regulator of energy homeostasis. Primary brown adipocyte culture provides a powerful and physiologically relevant tool for in vitro studies related to BAT. Here, we describe a detailed procedure for isolation and differentiation of adipocyte precursors from neonatal murine interscapular BAT (iBAT).


Subject(s)
Adipogenesis , Adipose Tissue, Brown , Animals , Mice , Adipose Tissue, Brown/physiology , Adipogenesis/physiology , Adipocytes, Brown , Homeostasis , Cell Differentiation
7.
Nat Metab ; 3(12): 1648-1661, 2021 12.
Article in English | MEDLINE | ID: mdl-34903883

ABSTRACT

To liberate fatty acids (FAs) from intracellular stores, lipolysis is regulated by the activity of the lipases adipose triglyceride lipase (ATGL), hormone-sensitive lipase and monoacylglycerol lipase. Excessive FA release as a result of uncontrolled lipolysis results in lipotoxicity, which can in turn promote the progression of metabolic disorders. However, whether cells can directly sense FAs to maintain cellular lipid homeostasis is unknown. Here we report a sensing mechanism for cellular FAs based on peroxisomal degradation of FAs and coupled with reactive oxygen species (ROS) production, which in turn regulates FA release by modulating lipolysis. Changes in ROS levels are sensed by PEX2, which modulates ATGL levels through post-translational ubiquitination. We demonstrate the importance of this pathway for non-alcoholic fatty liver disease progression using genetic and pharmacological approaches to alter ROS levels in vivo, which can be utilized to increase hepatic ATGL levels and ameliorate hepatic steatosis. The discovery of this peroxisomal ß-oxidation-mediated feedback mechanism, which is conserved in multiple organs, couples the functions of peroxisomes and lipid droplets and might serve as a new way to manipulate lipolysis to treat metabolic disorders.


Subject(s)
Fatty Acids/metabolism , Lipolysis , Oxidation-Reduction , Peroxisomes/metabolism , Acyltransferases/metabolism , Disulfides , Fatty Liver/etiology , Fatty Liver/metabolism , Fatty Liver/pathology , Gene Expression Regulation , HEK293 Cells , Humans , Lipid Metabolism , Liver/metabolism , Models, Biological , Peroxins/genetics , Peroxins/metabolism , Protein Binding , Protein Stability , Reactive Oxygen Species/metabolism , Ubiquitination
8.
Trends Cell Biol ; 31(8): 656-670, 2021 08.
Article in English | MEDLINE | ID: mdl-33674166

ABSTRACT

Peroxisomes are involved in multiple metabolic processes, including fatty acid oxidation, ether lipid synthesis, and reactive oxygen species (ROS) metabolism. Recent studies suggest that peroxisomes are critical mediators of cellular responses to various forms of stress, including oxidative stress, hypoxia, starvation, cold exposure, and noise. As dynamic organelles, peroxisomes can modulate their proliferation, morphology, and movement within cells, and engage in crosstalk with other organelles in response to external cues. Although peroxisome-derived hydrogen peroxide has a key role in cellular signaling related to stress, emerging studies suggest that other products of peroxisomal metabolism, such as acetyl-CoA and ether lipids, are also important for metabolic adaptation to stress. Here, we review molecular mechanisms through which peroxisomes regulate metabolic and environmental stress.


Subject(s)
Oxidative Stress , Peroxisomes , Lipid Metabolism , Oxidation-Reduction , Peroxisomes/metabolism , Reactive Oxygen Species/metabolism
9.
Cell Rep ; 33(1): 108228, 2020 10 06.
Article in English | MEDLINE | ID: mdl-33027649

ABSTRACT

The Mediator complex relays regulatory signals from gene-specific transcription factors to the basal transcriptional machinery. However, the role of individual Mediator subunits in different tissues remains unclear. Here, we demonstrate that MED19 is essential for adipogenesis and maintenance of white adipose tissue (WAT) by mediating peroxisome proliferator-activated receptor gamma (PPARγ) transcriptional activity. MED19 knockdown blocks white adipogenesis, but not brown adipogenesis or C2C12 myoblast differentiation. Adipose-specific MED19 knockout (KO) in mice results in a striking loss of WAT, whitening of brown fat, hepatic steatosis, and insulin resistance. Inducible adipose-specific MED19 KO in adult animals also results in lipodystrophy, demonstrating its requirement for WAT maintenance. Global gene expression analysis reveals induction of genes involved in apoptosis and inflammation and impaired expression of adipose-specific genes, resulting from decreased PPARγ residency on adipocyte gene promoters and reduced association of PPARγ with RNA polymerase II. These results identify MED19 as a crucial facilitator of PPARγ-mediated gene expression in adipose tissue.


Subject(s)
Adipose Tissue, White/metabolism , Gene Expression/genetics , Mediator Complex/metabolism , PPAR gamma/metabolism , Adipocytes, Brown/metabolism , Adipogenesis , Animals , Cell Differentiation , Humans , Mice
10.
Autophagy ; 16(9): 1727-1728, 2020 09.
Article in English | MEDLINE | ID: mdl-32687428

ABSTRACT

Hepatic lipid homeostasis is controlled by a coordinated regulation of various metabolic pathways involved in de novo synthesis, uptake, storage, and catabolism of lipids. Disruption of this balance could lead to hepatic steatosis. Peroxisomes play an essential role in lipid metabolism, yet their importance is often overlooked. In a recent study, we demonstrated a role for hepatic peroxisomal ß-oxidation in autophagic degradation of lipid droplets. ACOX1 (acyl-Coenzyme A oxidase 1, palmitoyl), the rate-limiting enzyme of peroxisomal ß-oxidation, increases with fasting or high-fat diet (HFD). Liver-specific acox1 knockout (acox1-LKO) protects mice from hepatic steatosis induced by starvation or HFD via induction of lipophagy. Mechanistically, we showed that hepatic ACOX1 deficiency decreases the total cytosolic acetyl-CoA levels, which leads to reduced acetylation of RPTOR/RAPTOR, a component of MTORC1, which is a key regulator of macroautophagy/autophagy. These results identify peroxisome-derived acetyl-CoA as a critical metabolic regulator of autophagy that controls hepatic lipid homeostasis.


Subject(s)
Autophagy , Liver/metabolism , Peroxisomes/metabolism , Regulatory-Associated Protein of mTOR/metabolism , TOR Serine-Threonine Kinases/metabolism , Acetyl Coenzyme A/metabolism , Acetylation , Animals , Humans , Lipid Droplets/metabolism , Mice, Knockout , Models, Biological , Oxidation-Reduction
11.
Mol Cell ; 79(1): 30-42.e4, 2020 07 02.
Article in English | MEDLINE | ID: mdl-32473093

ABSTRACT

Autophagy is activated by prolonged fasting but cannot overcome the ensuing hepatic lipid overload, resulting in fatty liver. Here, we describe a peroxisome-lysosome metabolic link that restricts autophagic degradation of lipids. Acyl-CoA oxidase 1 (Acox1), the enzyme that catalyzes the first step in peroxisomal ß-oxidation, is enriched in liver and further increases with fasting or high-fat diet (HFD). Liver-specific Acox1 knockout (Acox1-LKO) protected mice against hepatic steatosis caused by starvation or HFD due to induction of autophagic degradation of lipid droplets. Hepatic Acox1 deficiency markedly lowered total cytosolic acetyl-CoA levels, which led to decreased Raptor acetylation and reduced lysosomal localization of mTOR, resulting in impaired activation of mTORC1, a central regulator of autophagy. Dichloroacetic acid treatment elevated acetyl-CoA levels, restored mTORC1 activation, inhibited autophagy, and increased hepatic triglycerides in Acox1-LKO mice. These results identify peroxisome-derived acetyl-CoA as a key metabolic regulator of autophagy that controls hepatic lipid homeostasis.


Subject(s)
Acetyl Coenzyme A/metabolism , Acyl-CoA Oxidase/physiology , Autophagy , Fatty Acids/chemistry , Fatty Liver/pathology , Mechanistic Target of Rapamycin Complex 1/metabolism , Peroxisomes/chemistry , Acetylation , Animals , Autophagy-Related Protein 5/physiology , Diet, High-Fat/adverse effects , Fasting , Fatty Liver/etiology , Fatty Liver/metabolism , Female , Male , Mechanistic Target of Rapamycin Complex 1/genetics , Mice , Mice, Knockout , Mitochondria/metabolism , Oxidation-Reduction , Peroxisomes/metabolism , Regulatory-Associated Protein of mTOR/genetics , Regulatory-Associated Protein of mTOR/metabolism
12.
Sci Signal ; 12(606)2019 11 05.
Article in English | MEDLINE | ID: mdl-31690633

ABSTRACT

TFEB is a basic helix-loop-helix transcription factor that confers protection against metabolic diseases such as atherosclerosis by targeting a network of genes involved in autophagy-lysosomal biogenesis and lipid catabolism. In this study, we sought to characterize the role of TFEB in adipocyte and adipose tissue physiology and evaluate the therapeutic potential of adipocyte-specific TFEB overexpression in obesity. We demonstrated that mice with adipocyte-specific TFEB overexpression (Adipo-TFEB) were protected from diet-induced obesity, insulin resistance, and metabolic sequelae. Adipo-TFEB mice were lean primarily through increased metabolic rate, suggesting a role for adipose tissue browning and enhanced nonshivering thermogenesis in fat. Transcriptional characterization revealed that TFEB targeted genes involved in adipose tissue browning rather than those involved in autophagy. One such gene encoded PGC-1α, an established target of TFEB that promotes adipocyte browning. To dissect the role of PGC-1α in mediating the downstream effects of TFEB overexpression, we generated mice with adipocyte-specific PGC-1α deficiency and TFEB overexpression. Without PGC-1α, the ability of TFEB overexpression to brown adipose tissue and to elicit beneficial metabolic effects was blunted. Overall, these data implicate TFEB as a PGC-1α-dependent regulator of adipocyte browning and suggest its therapeutic potential in treating metabolic disease.


Subject(s)
Adipocytes/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Gene Expression Regulation , Metabolic Diseases/prevention & control , Obesity/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/biosynthesis , Adipocytes/pathology , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Metabolic Diseases/genetics , Metabolic Diseases/metabolism , Metabolic Diseases/pathology , Mice , Mice, Transgenic , Obesity/chemically induced , Obesity/genetics , Obesity/pathology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics
13.
Trends Endocrinol Metab ; 30(10): 710-723, 2019 10.
Article in English | MEDLINE | ID: mdl-31422871

ABSTRACT

The global prevalence of obesity continues to increase, suggesting a need for alternative treatment approaches. Targeting brown fat function to promote energy expenditure represents one such approach. Brown adipocytes and the related beige adipocytes oxidize fatty acids and glucose to generate heat and are activated by cold exposure or consumption of high-calorie diets. Alternative, more practical means to activate thermogenic fat are needed. Here, we review emerging data suggesting new roles for lipids in activating thermogenesis that extend beyond their serving as a fuel source for heat generation. Lipids have also been implicated in mediating interorgan communication, crosstalk between organelles, and cellular signaling regulating thermogenesis. Understanding how lipids regulate thermogenesis could identify innovative therapeutic interventions for obesity.


Subject(s)
Adipose Tissue, Beige/metabolism , Thermogenesis/physiology , Animals , Humans , Mitochondria/metabolism , Obesity/metabolism , Plasmalogens/metabolism
14.
J Clin Invest ; 129(2): 694-711, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30511960

ABSTRACT

Peroxisomes perform essential functions in lipid metabolism, including fatty acid oxidation and plasmalogen synthesis. Here, we describe a role for peroxisomal lipid metabolism in mitochondrial dynamics in brown and beige adipocytes. Adipose tissue peroxisomal biogenesis was induced in response to cold exposure through activation of the thermogenic coregulator PRDM16. Adipose-specific knockout of the peroxisomal biogenesis factor Pex16 (Pex16-AKO) in mice impaired cold tolerance, decreased energy expenditure, and increased diet-induced obesity. Pex16 deficiency blocked cold-induced mitochondrial fission, decreased mitochondrial copy number, and caused mitochondrial dysfunction. Adipose-specific knockout of the peroxisomal ß-oxidation enzyme acyl-CoA oxidase 1 (Acox1-AKO) was not sufficient to affect adiposity, thermogenesis, or mitochondrial copy number, but knockdown of the plasmalogen synthetic enzyme glyceronephosphate O-acyltransferase (GNPAT) recapitulated the effects of Pex16 inactivation on mitochondrial morphology and function. Plasmalogens are present in mitochondria and decreased with Pex16 inactivation. Dietary supplementation with plasmalogens increased mitochondrial copy number, improved mitochondrial function, and rescued thermogenesis in Pex16-AKO mice. These findings support a surprising interaction between peroxisomes and mitochondria regulating mitochondrial dynamics and thermogenesis.


Subject(s)
Adipose Tissue/metabolism , Cold Temperature , Lipids/biosynthesis , Mitochondria/metabolism , Mitochondrial Dynamics , Peroxisomes/metabolism , Thermogenesis , Acyl-CoA Oxidase/genetics , Acyl-CoA Oxidase/metabolism , Animals , COS Cells , Chlorocebus aethiops , HEK293 Cells , Humans , Lipids/genetics , Mice , Mice, Knockout , Mitochondria/genetics , Peroxins/genetics , Peroxins/metabolism , Peroxisomes/genetics , Plasmalogens/pharmacology
15.
Article in English | MEDLINE | ID: mdl-30405527

ABSTRACT

Leptin is known to inhibit appetite and promote energy metabolism in vertebrates. Leptin resistance (LR) commonly occurs in diet-induced obesity (DIO) in mammals. However, the roles of leptin in the energy homeostasis in DIO animals with LR remain unclear. Here we first verified the high expression of leptin in subcutaneous adipose tissue (SCAT) as in liver in Nile tilapia. Furthermore, we produced two types of DIO Nile tilapia by using a high-carbohydrate diet (HCD) or a high-fat diet (HFD), and confirmed the existence of LR in both models. Notably, we found that HCD-DIO fish retained leptin action in the activation of lipid metabolism and showed LR in glucose metabolism regulation, while this selective leptin action between lipid and glucose metabolism was reversed in HFD-DIO fish. Fasting the fish for 1 week completely recovered leptin actions in the regulation of lipid and glucose metabolism. Therefore, leptin may retain more of its activities in animals with LR than previously believed. Evolutionally, this selective regulation of leptin in nutrients metabolism could be an adaptive mechanism in animals to store surplus calories when different types of food are abundant.

16.
Environ Pollut ; 235: 245-254, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29291524

ABSTRACT

Antibiotics have been widely used in human and veterinary medicine to both treat and prevent disease. Due to their high water solubility and low bioavailability, many antibiotic residues have been found in aquatic environments. Fish are an indispensable link between the environmental pollution and human health. However, the chronic effects of environmental concentrations of antibiotics in fish have not been thoroughly investigated. Sulfamethoxazole (SMX) and oxytetracycline (OTC) are frequently detected in aquatic environments. In this study, zebrafish were exposed to SMX (260 ng/L) and OTC (420 ng/L) for a six-week period. Results indicated that exposure to antibiotics did not influence weight gain of fish but increased the metabolic rate and caused higher mortality when treated fish were challenged with Aeromonas hydrophila. Furthermore, exposure to antibiotics in water resulted in a significant decrease in intestinal goblet cell numbers, alkaline phosphatase (AKP), acid phosphatase (ACP) activities, and the anti-oxidant response while there was a significant increase in expression of inflammatory factors. Antibiotic exposure also disturbed the intestinal microbiota in the OTC-exposed group. Our results indicated that environmental antibiotic concentrations can impair the gut health of zebrafish. The potential health risk of antibiotic residues in water should be evaluated in the future.


Subject(s)
Anti-Bacterial Agents/toxicity , Gastrointestinal Microbiome/drug effects , Water Pollutants, Chemical/toxicity , Zebrafish/physiology , Animals , Humans , Oxytetracycline/toxicity , Sulfamethoxazole/toxicity , Zebrafish/metabolism
17.
Cell Rep ; 20(12): 2766-2774, 2017 Sep 19.
Article in English | MEDLINE | ID: mdl-28930673

ABSTRACT

How the nuclear receptor PPARγ regulates the development of two functionally distinct types of adipose tissue, brown and white fat, as well as the browning of white fat, remains unclear. Our previous studies suggest that PexRAP, a peroxisomal lipid synthetic enzyme, regulates PPARγ signaling and white adipogenesis. Here, we show that PexRAP is an inhibitor of brown adipocyte gene expression. PexRAP inactivation promoted adipocyte browning, increased energy expenditure, and decreased adiposity. Identification of PexRAP-interacting proteins suggests that PexRAP function extends beyond its role as a lipid synthetic enzyme. Notably, PexRAP interacts with importin-ß1, a nuclear import factor, and knockdown of PexRAP in adipocytes reduced the levels of nuclear phospholipids. PexRAP also interacts with PPARγ, as well as PRDM16, a critical transcriptional regulator of thermogenesis, and disrupts the PRDM16-PPARγ complex, providing a potential mechanism for PexRAP-mediated inhibition of adipocyte browning. These results identify PexRAP as an important regulator of adipose tissue remodeling.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression Regulation , Sugar Alcohol Dehydrogenases/metabolism , Thermogenesis/genetics , Transcription Factors/metabolism , Adipocytes/metabolism , Adipose Tissue, Brown/metabolism , Animals , Base Sequence , Cell Nucleus/metabolism , Gene Expression Profiling , Isotope Labeling , Mice, Inbred C57BL , Mice, Knockout , PPAR gamma/metabolism , Protein Binding , Protein Transport , Subcutaneous Fat/metabolism , Sugar Alcohol Dehydrogenases/genetics , Transcription, Genetic
18.
Sci Rep ; 7: 41706, 2017 01 31.
Article in English | MEDLINE | ID: mdl-28139735

ABSTRACT

Peroxisome proliferation activated receptor α (PPARα) is an important transcriptional regulator of lipid metabolism and is activated by high-fat diet (HFD) and fibrates in mammals. However, whether nutritional background affects PPARα activation and the hypolipidemic effects of PPARα ligands have not been investigated in fish. In the present two-phase study of Nile tilapia (Oreochromis niloticus), fish were first fed a HFD (13% fat) or low-fat diet (LFD; 1% fat) diet for 10 weeks, and then fish from the first phase were fed the HFD or LFD supplemented with 200 mg/kg body weight fenofibrate for 4 weeks. The results indicated that the HFD did not activate PPARα or other lipid catabolism-related genes. Hepatic fatty acid ß-oxidation increased significantly in the HFD and LFD groups after the fenofibrate treatment, when exogenous substrates were sufficiently provided. Only in the HFD group, fenofibrate significantly increased hepatic PPARα mRNA and protein expression, and decreased liver and plasma triglyceride concentrations. This is the first study to show that body fat deposition and dietary lipid content affects PPARα activation and the hypolipidemic effects of fenofibrate in fish, and this could be due to differences in substrate availability for lipid catabolism in fish fed with different diets.


Subject(s)
Cichlids/physiology , Fenofibrate/pharmacology , Hypolipidemic Agents/pharmacology , Nutritional Physiological Phenomena , Animals , Diet, High-Fat , Fatty Acids/metabolism , Gene Expression Regulation/drug effects , Lipid Metabolism/drug effects , Organ Specificity/genetics , Oxidation-Reduction , PPAR alpha/genetics , PPAR alpha/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
19.
Biochim Biophys Acta ; 1861(9 Pt A): 1036-1048, 2016 09.
Article in English | MEDLINE | ID: mdl-27320014

ABSTRACT

Although the key metabolic regulatory functions of mammalian peroxisome proliferator-activated receptor α (PPARα) have been thoroughly studied, the molecular mechanisms and metabolic regulation of PPARα activation in fish are less known. In the first part of the present study, Nile tilapia (Nt)PPARα was cloned and identified, and high mRNA expression levels were detected in the brain, liver, and heart. NtPPARα was activated by an agonist (fenofibrate) and by fasting and was verified in primary hepatocytes and living fish by decreased phosphorylation of NtPPARα and/or increased NtPPARα mRNA and protein expression. In the second part of the present work, fenofibrate was fed to fish or fish were fasted for 4weeks to investigate the metabolic regulatory effects of NtPPARα. A transcriptomic study was also performed. The results indicated that fenofibrate decreased hepatic triglyceride and 18C-series fatty acid contents but increased the catabolic rate of intraperitoneally injected [1-(14)C] palmitate in vivo, hepatic mitochondrial ß-oxidation efficiency, the quantity of cytochrome b DNA, and carnitine palmitoyltransferase-1a mRNA expression. Fenofibrate also increased serum glucose, insulin, and lactate concentrations. Fasting had stronger hypolipidemic and gene regulatory effects than those of fenofibrate. Taken together, we conclude that: 1) liver is one of the main target tissues of the metabolic regulation of NtPPARα activation; 2) dephosphorylation is the basal NtPPARα activation mechanism rather than enhanced mRNA and protein expression; 3) activated NtPPARα has a hypolipidemic effect by increasing activity and the number of hepatic mitochondria; and 4) PPARα activation affects carbohydrate metabolism by altering energy homeostasis among nutrients.


Subject(s)
Hepatocytes/metabolism , Liver/metabolism , PPAR alpha/biosynthesis , Tilapia/genetics , Animals , Fatty Acids/metabolism , Gene Expression Regulation , PPAR alpha/metabolism , RNA, Messenger/biosynthesis , Triglycerides/metabolism
20.
Fish Shellfish Immunol ; 50: 160-7, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26820103

ABSTRACT

Suppressor of cytokine signaling (SOCS) proteins are inverse feedback regulators of cytokine and hormone signaling mediated by the Janus kinase/signal transducer and activator of transcription (JAK/STAT) signaling pathway that are involved in immunity, growth and development of organisms. In the present study, three SOCS genes, SOCS-1, SOCS-2 and SOCS-3, were identified in an economically important fish, Nile tilapia (Oreochromis niloticus) referred to as NtSOCS-1, NtSOCS-2 and NtSOCS-3. Multiple alignments showed that, the three SOCS molecules share highly conserved functional domains, including the SRC homology 2 (SH2) domain, the extended SH2 subdomain (ESS) and the SOCS box with others vertebrate counterparts. Phylogenetic analysis indicated that NtSOCS-1, 2 and 3 belong to the SOCS type II subfamily. Whereas NtSOCS-1 and 3 showed close evolutionary relationship with Perciformes, NtSOCS-2 was more related to Salmoniformes. Tissue specific expression results showed that, NtSOCS-1, 2 and 3 were constitutively expressed in all nine tissues examined. NtSOCS-1 and 3 were highly expressed in immune-related tissues, such as gills, foregut and head kidney. However, NtSOCS-2 was superlatively expressed in liver, brain and heart. In vivo, NtSOCS-1 and 3 mRNA levels were up-regulated after lipopolysaccharide (LPS) challenge while NtSOCS-2 was down-regulated. In vitro, LPS stimulation increased NtSOCS-3 mRNA expression, however it inhibited the transcription of NtSOCS-1 and 2. Collectively, our findings suggest that, the NtSOCS-1 and 3 might play significant role(s) in innate immune response, while NtSOCS-2 may be more involved in metabolic regulation.


Subject(s)
Cichlids/genetics , Fish Proteins/genetics , Immunity, Innate , Suppressor of Cytokine Signaling Proteins/genetics , Amino Acid Sequence , Animals , Cichlids/metabolism , Cloning, Molecular , DNA, Complementary/genetics , DNA, Complementary/metabolism , Escherichia coli/chemistry , Fish Proteins/chemistry , Fish Proteins/metabolism , Immunity, Innate/drug effects , Lipopolysaccharides/pharmacology , Molecular Conformation , Organ Specificity , Phylogeny , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Alignment/veterinary , Suppressor of Cytokine Signaling Proteins/chemistry , Suppressor of Cytokine Signaling Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...