Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Biochem Mol Toxicol ; 29(1): 1-9, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25155036

ABSTRACT

Arsenic, a ubiquitous environmental toxicant, can affect lipid metabolism through mechanisms that are not well understood. We studied the effect of arsenic on serum lipids, lipid-regulating genes, and transcriptional regulator sterol regulatory element binding protein 1c (SREBP-1c). C57BL/6 mice were administered 0 or 100 ppb sodium arsenite in drinking water for 5 weeks. Arsenic exposure was associated with decreased liver weight but no change in body weight. Serum triglycerides level fell in arsenic-exposed animals, but not in fed animals, after short-term fasting. Hepatic expression of SREBP-1c was reduced in arsenic-exposed fed animals, with a 16-fold change in reduction. Similar effects were seen for SREBP-1c in white adipose tissue. However, fasting resulted in dissociation of the expression of SREBP-1c and its targets, and SREBP-1c protein content could not be shown to correlate with its mRNA expression. We conclude that arsenic modulates hepatic expression of genes involved in lipid regulation through mechanisms that are independent of SREBP-1c expression.


Subject(s)
Adipose Tissue, White/metabolism , Arsenites/pharmacology , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Lipogenesis/drug effects , Liver/metabolism , Sodium Compounds/pharmacology , Sterol Regulatory Element Binding Protein 1/biosynthesis , Animals , Arsenic/pharmacology , Male , Mice , Triglycerides/biosynthesis
2.
PLoS One ; 7(5): e38249, 2012.
Article in English | MEDLINE | ID: mdl-22693606

ABSTRACT

BACKGROUND: Arsenic (As) exposure is a significant worldwide environmental health concern. Chronic exposure via contaminated drinking water has been associated with an increased incidence of a number of diseases, including reproductive and developmental effects. The goal of this study was to identify adverse outcomes in a mouse model of early life exposure to low-dose drinking water As (10 ppb, current U.S. EPA Maximum Contaminant Level). METHODOLOGY AND FINDINGS: C57B6/J pups were exposed to 10 ppb As, via the dam in her drinking water, either in utero and/or during the postnatal period. Birth outcomes, the growth of the F1 offspring, and health of the dams were assessed by a variety of measurements. Birth outcomes including litter weight, number of pups, and gestational length were unaffected. However, exposure during the in utero and postnatal period resulted in significant growth deficits in the offspring after birth, which was principally a result of decreased nutrients in the dam's breast milk. Cross-fostering of the pups reversed the growth deficit. Arsenic exposed dams displayed altered liver and breast milk triglyceride levels and serum profiles during pregnancy and lactation. The growth deficits in the F1 offspring resolved following separation from the dam and cessation of exposure in male mice, but did not resolve in female mice up to six weeks of age. CONCLUSIONS/SIGNIFICANCE: Exposure to As at the current U.S. drinking water standard during critical windows of development induces a number of adverse health outcomes for both the dam and offspring. Such effects may contribute to the increased disease risks observed in human populations.


Subject(s)
Arsenic/pharmacology , Drinking Water/chemistry , Fetus/drug effects , Growth and Development/drug effects , Water Pollutants, Chemical/pharmacology , Animal Husbandry , Animals , Arsenic/metabolism , Dose-Response Relationship, Drug , Female , Lactation/drug effects , Male , Mice , Mice, Inbred C57BL , Milk, Human/drug effects , Milk, Human/metabolism , Pregnancy , Triglycerides/metabolism
3.
Biochim Biophys Acta ; 1771(8): 972-82, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17631413

ABSTRACT

Peroxisome proliferator-activated receptor (PPAR)alpha is a nuclear receptor activated by natural ligands such as fatty acids as well as by synthetic ligands such as fibrates currently used to treat dyslipidemia. PPARalpha regulates the expression of genes encoding proteins that are involved in lipid metabolism, fatty acid oxidation, and glucose homeostasis, thereby improving markers for atherosclerosis and insulin resistance. In addition, PPARalpha exerts anti-inflammatory effects both in the vascular wall and the liver. Here we provide an overview of the mechanisms through which PPARalpha affects the initiation and progression of atherosclerosis, with emphasis on the modulation of atherosclerosis-associated inflammatory responses. PPARalpha activation interferes with early steps in atherosclerosis by reducing leukocyte adhesion to activated endothelial cells of the arterial vessel wall and inhibiting subsequent transendothelial leukocyte migration. In later stages of atherosclerosis, evidence suggests activation of PPARalpha inhibits the formation of macrophage foam cells by regulating expression of genes involved in reverse cholesterol transport, formation of reactive oxygen species (ROS), and associated lipoprotein oxidative modification among others. Furthermore, PPARalpha may increase the stability of atherosclerotic plaques and limit plaque thrombogenicity. These various effects may be linked to the generation of PPARalpha ligands by endogenous mechanisms of lipoprotein metabolism. In spite of this dataset, other reports implicate PPARalpha in responses such as hypertension and diabetic cardiomyopathy. Although some clinical trials data with fibrates suggest that fibrates may decrease cardiovascular events, other studies have been less clear, in terms of benefit. Independent of the clinical effects of currently used drugs purported to achieve PPARalpha, extensive data establish the importance of PPARalpha in the transcriptional regulation of lipid metabolism, atherosclerosis, and inflammation.


Subject(s)
Atherosclerosis/physiopathology , Inflammation/physiopathology , Liver/physiopathology , PPAR alpha/physiology , Atherosclerosis/immunology , Cell Adhesion , Cytokines/physiology , Endothelium, Vascular/physiopathology , Humans , Inflammation/immunology , Leukocytes/physiology , PPAR alpha/immunology
4.
J Biol Chem ; 281(2): 934-44, 2006 Jan 13.
Article in English | MEDLINE | ID: mdl-16272564

ABSTRACT

Proteins secreted from adipose tissue are increasingly recognized to play an important role in the regulation of glucose metabolism. However, much less is known about their effect on lipid metabolism. The fasting-induced adipose factor (FIAF/angiopoietin-like protein 4/peroxisome proliferator-activated receptor gamma angiopoietin-related protein) was previously identified as a target of hypolipidemic fibrate drugs and insulin-sensitizing thiazolidinediones. Using transgenic mice that mildly overexpress FIAF in peripheral tissues we show that FIAF is an extremely powerful regulator of lipid metabolism and adiposity. FIAF overexpression caused a 50% reduction in adipose tissue weight, partly by stimulating fatty acid oxidation and uncoupling in fat. In addition, FIAF overexpression increased plasma levels of triglycerides, free fatty acids, glycerol, total cholesterol, and high density lipoprotein (HDL)-cholesterol. Functional tests indicated that FIAF overexpression severely impaired plasma triglyceride clearance but had no effect on very low density lipoprotein production. The effects of FIAF overexpression were amplified by a high fat diet, resulting in markedly elevated plasma and liver triglycerides, plasma free fatty acids, and plasma glycerol levels, and impaired glucose tolerance in FIAF transgenic mice fed a high fat diet. Remarkably, in mice the full-length form of FIAF was physically associated with HDL, whereas truncated FIAF was associated with low density lipoprotein. In human both full-length and truncated FIAF were associated with HDL. The composite data suggest that via physical association with plasma lipoproteins, FIAF acts as a powerful signal from fat and other tissues to prevent fat storage and stimulate fat mobilization. Our data indicate that disturbances in FIAF signaling might be involved in dyslipidemia.


Subject(s)
Adipose Tissue/metabolism , Blood Proteins/genetics , Blood Proteins/physiology , Lipoproteins/chemistry , Angiopoietin-Like Protein 4 , Angiopoietins , Animals , Blood Proteins/chemistry , Body Weight , Cholesterol/metabolism , Fats/chemistry , Gene Expression , Glucose/metabolism , Glucose Tolerance Test , Humans , Hypercholesterolemia/metabolism , Immunoblotting , Insulin/metabolism , Lipase/metabolism , Lipids/chemistry , Lipoproteins, HDL/chemistry , Lipoproteins, HDL/metabolism , Lipoproteins, LDL/chemistry , Lipoproteins, VLDL/chemistry , Liver/enzymology , Male , Mice , Mice, Transgenic , Models, Genetic , Oligonucleotide Array Sequence Analysis , Protein Binding , RNA, Messenger/metabolism , Signal Transduction , Time Factors , Triglycerides/blood , Triglycerides/metabolism , Up-Regulation
5.
Biochem J ; 392(Pt 2): 313-24, 2005 Dec 01.
Article in English | MEDLINE | ID: mdl-16086669

ABSTRACT

PPARs (peroxisome-proliferator-activated receptors) alpha, beta/delta and gamma are a group of transcription factors that are involved in numerous processes, including lipid metabolism and adipogenesis. By comparing liver mRNAs of wild-type and PPARalpha-null mice using microarrays, a novel putative target gene of PPARalpha, G0S2 (G0/G1 switch gene 2), was identified. Hepatic expression of G0S2 was up-regulated by fasting and by the PPARalpha agonist Wy14643 in a PPARalpha-dependent manner. Surprisingly, the G0S2 mRNA level was highest in brown and white adipose tissue and was greatly up-regulated during mouse 3T3-L1 and human SGBS (Simpson-Golabi-Behmel syndrome) adipogenesis. Transactivation, gel shift and chromatin immunoprecipitation assays indicated that G0S2 is a direct PPARgamma and probable PPARalpha target gene with a functional PPRE (PPAR-responsive element) in its promoter. Up-regulation of G0S2 mRNA seemed to be specific for adipogenesis, and was not observed during osteogenesis or myogenesis. In 3T3-L1 fibroblasts, expression of G0S2 was associated with growth arrest, which is required for 3T3-L1 adipogenesis. Together, these data indicate that G0S2 is a novel target gene of PPARs that may be involved in adipocyte differentiation.


Subject(s)
Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , PPAR alpha/metabolism , Adipocytes/cytology , Adipocytes/metabolism , Adipogenesis , Animals , Base Sequence , Cell Line , Endoplasmic Reticulum/metabolism , Gene Deletion , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Liver/cytology , Male , Mice , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , PPAR alpha/genetics , Promoter Regions, Genetic/genetics , Protein Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Response Elements/genetics , Sequence Homology, Nucleic Acid , Substrate Specificity , Up-Regulation
6.
J Biol Chem ; 279(33): 34411-20, 2004 Aug 13.
Article in English | MEDLINE | ID: mdl-15190076

ABSTRACT

The fasting-induced adipose factor (FIAF, ANGPTL4, PGAR, HFARP) was previously identified as a novel adipocytokine that was up-regulated by fasting, by peroxisome proliferator-activated receptor agonists, and by hypoxia. To further characterize FIAF, we studied regulation of FIAF mRNA and protein in liver and adipose cell lines as well as in human and mouse plasma. Expression of FIAF mRNA was up-regulated by peroxisome proliferator-activated receptor alpha (PPARalpha) and PPARbeta/delta agonists in rat and human hepatoma cell lines and by PPARgamma and PPARbeta/delta agonists in mouse and human adipocytes. Transactivation, chromatin immunoprecipitation, and gel shift experiments identified a functional PPAR response element within intron 3 of the FIAF gene. At the protein level, in human and mouse blood plasma, FIAF was found to be present both as the native protein and in a truncated form. Differentiation of mouse 3T3-L1 adipocytes was associated with the production of truncated FIAF, whereas in human white adipose tissue and SGBS adipocytes, only native FIAF could be detected. Interestingly, truncated FIAF was produced by human liver. Treatment with fenofibrate, a potent PPARalpha agonist, markedly increased plasma levels of truncated FIAF, but not native FIAF, in humans. Levels of both truncated and native FIAF showed marked interindividual variation but were not associated with body mass index and were not influenced by prolonged semistarvation. Together, these data suggest that FIAF, similar to other adipocytokines such as adiponectin, may partially exert its function via a truncated form.


Subject(s)
Fenofibrate/pharmacology , Intercellular Signaling Peptides and Proteins/blood , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism , 3T3-L1 Cells , Adipocytes/metabolism , Adipokines/metabolism , Adiponectin , Adipose Tissue/metabolism , Angiopoietin-Like Protein 4 , Angiopoietins , Animals , Base Sequence , Blotting, Western , Cell Differentiation , Cell Line , Cell Line, Tumor , Cells, Cultured , Chromatin/metabolism , Cytokines/metabolism , Fenofibrate/metabolism , Humans , Introns , Liver/metabolism , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Precipitin Tests , Proteins/metabolism , RNA/metabolism , RNA, Messenger/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic , Transcriptional Activation , Up-Regulation
7.
J Gen Virol ; 83(Pt 1): 257-265, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11752723

ABSTRACT

White spot syndrome virus (WSSV) infects penaeid shrimp and other crustaceans. The WSSV virion consists of an enveloped rod-shaped nucleocapsid enclosing a large circular double-stranded DNA genome of 293 kbp. The virion envelope contains two major proteins of 28 (VP28) and 19 kDa (VP19) and the nucleocapsid consists of three major proteins of 26 (VP26), 24 (VP24) and 15 kDa (VP15). Study on the morphogenesis of the WSSV particle requires the genomic identification and chemical characterization of these WSSV virion proteins. An internal amino acid sequence of envelope protein VP19 was obtained by amino acid sequencing and used to locate the VP19 open reading frame of this protein on the genome, as WSSV ORF182. VP19 contained two putative transmembrane domains, which may anchor this protein in the WSSV envelope. Similarly, the gene for VP15 was located on the WSSV genome as ORF109. N-terminal amino acid sequencing on VP15 suggested that this protein was expressed from the second ATG of its ORF and the first methionine is lost by N-terminal protein processing. The 15 kDa protein is very basic and is a candidate DNA-binding protein in the WSSV nucleocapsid. None of the five major structural WSSV proteins appear to be glycosylated, which is an unusual feature among enveloped animal viruses.


Subject(s)
DNA Viruses/metabolism , Decapoda/virology , Nucleocapsid Proteins/metabolism , Viral Envelope Proteins/metabolism , Amino Acid Sequence , Animals , Astacoidea , Base Sequence , Cell Line , DNA Viruses/genetics , DNA, Viral , Genes, Viral , Genome, Viral , Glycosylation , Molecular Sequence Data , Nucleocapsid Proteins/genetics , Viral Envelope Proteins/genetics , Virion
SELECTION OF CITATIONS
SEARCH DETAIL
...