Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Toxicol Lett ; 219(1): 1-7, 2013 May 10.
Article in English | MEDLINE | ID: mdl-23458897

ABSTRACT

Exposure of mice to perfluorooctanoate (PFOA) evokes pronounced hepatomegaly along with significant alterations in both the histological structure and immune status of the liver. The present study was designed to evaluate the effects of this perfluorochemical on immune-mediated liver damage. In this connection, the influence of both sub-acute (10 days), moderate-dose (0.002% w/w=3±0.7mg/kg body weight/day) and short-term (28 days), low-dose (0.00005% w/w=70±2µg/kg body weight/day) dietary pretreatment with PFOA on the development of concanavalin A (Con A)-induced liver damage in mice was examined. With sub-acute, moderate, but not short-term, low-dose exposure, PFOA aggravated the acute liver damage caused by Con A, i.e., elevated serum levels of transaminases and led to more pronounced damage of hepatic tissue. This aggravation was associated with significantly enhanced hepatic level of interleukin-6 (IL-6), but unaltered hepatic levels of tumor necrosis factor-alpha (TNF-α), interferon-gamma (IFN-γ) and interleukin-4 (IL-4). Moreover, hepatic DNA fragmentation was not changed by sub-acute exposure to the moderate-dose. Our findings imply that exposure to PFOA may sensitize hepatic parenchymal cells to other toxicants that activate the hepatic immune system and thereby aggravate liver injury during acute inflammation.


Subject(s)
Caprylates/toxicity , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/immunology , Concanavalin A/toxicity , DNA Fragmentation/drug effects , Fluorocarbons/toxicity , Liver/drug effects , Administration, Oral , Animals , Chemical and Drug Induced Liver Injury/blood , Chemical and Drug Induced Liver Injury/pathology , Cytokines/blood , Dose-Response Relationship, Drug , Drug Synergism , Liver/immunology , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Time Factors , Transaminases/blood
2.
Toxicol Lett ; 217(1): 67-74, 2013 Feb 13.
Article in English | MEDLINE | ID: mdl-23237827

ABSTRACT

Exposure of rodents to perfluorooctane sulfonate (PFOS) induces pronounced hepatomegaly associated with significant alterations in hepatic histophysiology and immune status. The present investigation was designed to evaluate the effects of this perfluorochemical on immune-mediated liver damage. Accordingly, the influence of both sub-acute (10 days), moderate-dose (0.004%, w/w=6±1.3 mg/kg body weight/day) or short-term (28 days), low-dose (0.0001%, w/w=144±4 µg/kg body weight/day) dietary pretreatment with PFOS on the development of concanavalin A (Con A)-induced liver damage in mice was examined. With either regimen of exposure, PFOS exacerbated the acute liver damage caused by Con A, i.e., elevated serum levels of transaminases and led to more pronounced damage of hepatic tissue. This exacerbation was associated with either reduced (moderate dose) or unaltered (low dose) hepatic levels of the pro-inflammatory cytokines tumor necrosis factor alpha (TNF-α) and interferon gamma (IFN-γ). Moreover, hepatic DNA fragmentation was enhanced, particularly following short-term exposure to a low-dose. Our findings suggest that exposure to PFOS may sensitize hepatic parenchymal cells to other insults that activate the hepatic immune system and thereby exacerbate liver damage during acute inflammation.


Subject(s)
Alkanesulfonic Acids/administration & dosage , Chemical and Drug Induced Liver Injury/physiopathology , Disease Models, Animal , Environmental Pollutants/administration & dosage , Fluorocarbons/administration & dosage , Immunologic Factors/administration & dosage , Liver/drug effects , Alkanesulfonic Acids/toxicity , Animals , Apoptosis/drug effects , Chemical and Drug Induced Liver Injury/immunology , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Concanavalin A , Cytokines/metabolism , DNA Fragmentation/drug effects , Disease Progression , Dose-Response Relationship, Drug , Environmental Pollutants/toxicity , Fluorocarbons/toxicity , Hepatomegaly/etiology , Immunologic Factors/toxicity , Liver/immunology , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Random Allocation , Time Factors , Transaminases/blood
3.
Food Chem Toxicol ; 50(9): 2955-63, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22732150

ABSTRACT

It is well established that exposure of mice to perfluorooctanoate (PFOA) or perfluorooctane sulfonate (PFOS) exerts adverse effects on the thymus and spleen. Here, we characterize the effects of a 10-day dietary treatment with these compounds (0.001-0.02%, w/w) on the bone marrow (BM) of mice. At a dose of 0.02%, both compounds reduced food consumption and caused atrophy of the thymus and spleen. At this same dose, histopathological and flow cytometric analysis revealed that (i) the total numbers of BM as well as the numbers of myeloid, pro/pre B, immature B and early mature B cells were all reduced significantly; and (ii) these adverse effects were reversed either partially or completely 10days after withdrawal of these compounds. At the lower dose of 0.002%, only PFOA reduced the B-lymphoid cell population. Finally, mice fed an amount of diet equivalent to that consumed by the animals exposed to 0.02% PFOA also exhibited atrophy of the thymus and spleen, and a reduction in the number of B-lymphoid population, without affecting myeloid cells. Thus, in mice, immunotoxic doses of PFOA or PFOS induce adverse effects on the myeloid and B-lymphoid cells in the BM, in part as a consequence of reduced food consumption.


Subject(s)
Alkanesulfonic Acids/toxicity , B-Lymphocytes/drug effects , Bone Marrow Cells/drug effects , Caprylates/toxicity , Feeding Behavior , Fluorocarbons/toxicity , Alkanesulfonic Acids/administration & dosage , Animals , Body Weight/drug effects , Caprylates/administration & dosage , Dose-Response Relationship, Drug , Flow Cytometry , Fluorocarbons/administration & dosage , Male , Mice , Mice, Inbred C57BL
4.
Biochem Biophys Res Commun ; 411(3): 648-53, 2011 Aug 05.
Article in English | MEDLINE | ID: mdl-21782795

ABSTRACT

We earlier reported the formation of a unique nuclear NF1/Smad complex in serum-restricted fibroblasts that acts as an NF1-dependent repressor of the human adenine nucleotide translocase-2 gene (ANT2) [K. Luciakova, G. Kollarovic, P. Barath, B.D. Nelson, Growth-dependent repression of human adenine nucleotide translocator-2 (ANT2) transcription: evidence for the participation of Smad and Sp family proteins in the NF1-dependent repressor complex, Biochem. J. 412 (2008) 123-130]. In the present study, we show that TGF-ß, like serum-restriction: (a) induces the formation of NF1/Smad repressor complexes, (b) increases binding of the complexes to the repressor elements (Go elements) in the ANT2 promoter, and (c) inhibits ANT2 expression. Repression of ANT2 by TGF-ß is eliminated by mutating the NF1 binding sites in the Go repressor elements. All of the above responses to TGF-ß are prevented by inhibitors of TGF-ß RI and MAPK p38. These inhibitors also prevent NF1/Smad4 repressor complex formation and repression of ANT2 expression in serum-restricted cells, suggesting that similar signaling pathways are initiated by TGF-ß and serum-restriction. The present finding that NF1/Smad4 repressor complexes are formed through TGF-ß signaling pathways suggests a new, but much broader, role for these complexes in the initiation or maintenance of the growth-inhibited state.


Subject(s)
Adenine Nucleotide Translocator 2/genetics , Fibroblasts/metabolism , Gene Expression Regulation , Neurofibromin 1/metabolism , Repressor Proteins/metabolism , Smad4 Protein/metabolism , Transforming Growth Factor beta/metabolism , Animals , Diploidy , Humans , Mice , Transcription, Genetic , Transforming Growth Factor beta/antagonists & inhibitors
5.
Int Immunopharmacol ; 10(11): 1420-7, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20816993

ABSTRACT

It is well established that exposure of mice to perfluorooctanoate (PFOA) or perfluorooctane sulfonate (PFOS) induces hepatomegaly and, concurrently, immunotoxicity. However, the effects of these perfluorochemicals on the histology and immune status of the liver have not been yet investigated and we have examined these issues here. Dietary treatment of male C57BL/6 mice with 0.002% (w/w) PFOA or 0.005% (w/w) PFOS for 10 days resulted in significant reductions in serum levels of cholesterol and triglycerides, a moderate increase in the serum activity of alkaline phosphatase (ALP) and hepatomegaly, without affecting other immune organs. This hepatomegaly was associated with marked hypertrophy of the centrilobular hepatocytes, with elevated numbers of cytoplasmic acidophilic granules and occasional mitosis. Furthermore, dietary exposure to PFOA or PFOS altered the hepatic immune status: whereas exposure to PFOA enhanced the numbers of total, as well as of phenotypically distinct subpopulations of intrahepatic immune cells (IHIC), and in particular the presumptive erythrocyte progenitor cells, treatment with PFOS enhanced only the numbers of hepatic cells that appear immunophenotypically to be erythrocyte progenitors, without affecting other types of IHIC. In addition, exposure to these compounds attenuated hepatic levels of tumor necrosis factor-alpha (TNF-α), interferon-gamma (IFN-γ) and interleukin-4 (IL-4). Furthermore, the exposed animals exhibited a significant increase in hepatic levels of erythropoietin, a hormone required for erythropoiesis. Thus, in mice, PFOA- and PFOS-induced hepatomegaly is associated with significant alterations in hepatic histophysiology and immune status, as well as induction of hepatic erythropoiesis.


Subject(s)
Alkanesulfonic Acids/toxicity , Caprylates/toxicity , Environmental Pollutants/toxicity , Fluorocarbons/toxicity , Hepatomegaly/chemically induced , Liver/drug effects , Alkaline Phosphatase/blood , Alkanesulfonic Acids/administration & dosage , Animals , Caprylates/administration & dosage , Cholesterol/blood , Diet/adverse effects , Environmental Pollutants/administration & dosage , Erythropoietin/analysis , Fluorocarbons/administration & dosage , Hepatomegaly/immunology , Interferon-gamma/analysis , Interleukin-4/analysis , Liver/immunology , Male , Mice , Mice, Inbred C57BL , Mitosis/drug effects , Mitosis/immunology , Triglycerides/blood , Tumor Necrosis Factor-alpha/analysis
6.
Toxicology ; 267(1-3): 132-9, 2010 Jan 12.
Article in English | MEDLINE | ID: mdl-19900501

ABSTRACT

Short-term exposure of mice to high doses of perfluorooctanesulfonate (PFOS), an ubiquitous and highly persistent environmental contaminant, induces various metabolic changes and toxic effects, including immunotoxicity. However, extrapolation of these findings to the long-term, low-dose exposures to which humans are subject is highly problematic. In this connection, recent studies have concluded that sub-chronic (28-day) exposure of mice by oral gavage to doses of PFOS that result in serum levels comparable to those found in general human populations suppress adaptive immunity. Because of the potential impact of these findings on environmental research and monitoring, we have examined here whether sub-chronic dietary exposure (a major route of human exposure) to a similarly low-dose of PFOS also suppress adaptive immune responses. Dietary treatment of male B6C3F1 mice for 28 days with a dose of PFOS that resulted in a serum concentration of 11mug/ml (ppm) significantly reduced body weight gain and increased liver mass. However, this treatment did not alter the cellular compositions of the thymus and spleen; the number of splenic cells secreting IgM antibodies against sheep red blood cell (SRBC); serum levels of IgM and IgG antibodies specifically towards SRBC; or circulating levels of IgM antibodies against the T-cell-independent antigen trinitrophenyl conjugated to lipopolysaccharide (TNP-LPS). These findings indicate that such sub-chronic dietary exposure of mice to PFOS resulting in serum levels approximately 8-85-fold greater than those observed in occupationally exposed individuals does not exert adverse effects on adaptive immunity.


Subject(s)
Alkanesulfonic Acids/toxicity , Fluorocarbons/toxicity , Immunity, Humoral/drug effects , Spleen/drug effects , Thymus Gland/drug effects , Alkanesulfonic Acids/administration & dosage , Alkanesulfonic Acids/blood , Animals , Body Weight/drug effects , Corticosterone/blood , Diet , Dose-Response Relationship, Drug , Eating/drug effects , Fluorocarbons/administration & dosage , Fluorocarbons/blood , Immunoglobulin G/blood , Immunoglobulin M/blood , Male , Mice , Organ Size/drug effects , Spleen/immunology , Thymus Gland/immunology
7.
Toxicology ; 262(3): 207-14, 2009 Aug 21.
Article in English | MEDLINE | ID: mdl-19540903

ABSTRACT

Having found previously that high-dose, short-term dietary exposure of mice to perfluorooctanesulfonate (PFOS) or perfluorooctanoate (PFOA) suppresses adaptive immunity, in the present study we characterize the effects of these fluorochemicals on the innate immune system. Male C57BL/6 mice receiving 0.02% (w/w) PFOS or PFOA in their diet for 10 days exhibited a significant reduction in the numbers of total white blood cells (WBC), involving lymphopenia in both cases, but neutropenia only in response to treatment with PFOA. Moreover, both compounds also markedly reduced the number of macrophages (CD11b(+) cells) in the bone marrow, but not in the spleen or peritoneal cavity. The ex vivo production of tumor necrosis factor-alpha (TNF-alpha) and interleukin 6 (IL-6) by peritoneal macrophages isolated from animals treated with PFOA or PFOS was increased modestly. Moreover, both fluorochemicals markedly enhanced the ex vivo production of these same cytokines by peritoneal and bone marrow macrophages stimulated either in vitro or in vivo with lipopolysaccharide (LPS); whereas there was no such effect on splenic macrophages. The serum levels of these inflammatory cytokines observed in response to in vivo stimulation with LPS were elevated substantially by prior exposure to PFOA, but not by PFOS. None of these parameters of innate immunity were altered in animals receiving a dietary dose of these compounds that was 20-fold lower (0.001%, w/w). These findings reveal that in addition to suppressing adaptive immunity, high-dose, short-term exposure of mice to either PFOS or PFOA augments inflammatory responses to LPS, a potent activator of innate immunity.


Subject(s)
Alkanesulfonic Acids/toxicity , Caprylates/toxicity , Fluorocarbons/toxicity , Immunity, Innate/drug effects , Neutrophils/drug effects , Alkanesulfonic Acids/administration & dosage , Animals , Bone Marrow/drug effects , Bone Marrow/metabolism , Caprylates/administration & dosage , Dose-Response Relationship, Drug , Fluorocarbons/administration & dosage , Interleukin-6/metabolism , Leukocytes/drug effects , Leukocytes/metabolism , Lipopolysaccharides/metabolism , Lymphopenia/chemically induced , Macrophages/drug effects , Macrophages/metabolism , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Male , Mice , Mice, Inbred C57BL , Neutropenia/chemically induced , Neutrophils/metabolism , Spleen/drug effects , Spleen/metabolism , Tumor Necrosis Factor-alpha
8.
Toxicology ; 260(1-3): 68-76, 2009 Jun 16.
Article in English | MEDLINE | ID: mdl-19464571

ABSTRACT

We have previously shown that short-term, high-dose exposure of mice to the environmentally persistent perfluorooctanoate (PFOA) results in thymic and splenic atrophy and the attenuation of specific humoral immune responses. Here we characterize the effects of a 10-day treatment with different dietary doses (1-0.001%, w/w) of perfluorooctanesulfonate (PFOS), a similar fluorochemical, on the immune system of male C57BL/6 mice. At doses greater than 0.02%, PFOS induced clinical signs of toxicity in the animals, whereas at the concentration of 0.02%, this compound caused weight loss, hepatomegaly and atrophy of the thymus, spleen and adipose tissue without toxicity. With this latter dose, histopathological and flow-cytometric analysis revealed that (i) the thymic cortex was virtually depleted of cells; (ii) the total numbers of thymocytes and splenocytes were reduced by 84 and 43%, respectively; (iii) although all populations of thymocytes and splenocytes were smaller, the thymic CD4(+)CD8(+) cells and the splenic B-lymphocytes were most decreased. These alterations resembled those evoked by analogous exposure to PFOA, but were less pronounced. At lower doses (less than 0.02%), PFOS induced hepatomegaly without affecting the thymus or spleen. Finally, comparison of male wild-type 129/Sv mice and the corresponding knock-outs lacking peroxisome proliferator-activated receptor-alpha (PPARalpha) indicated that these effects of PFOS are not strain-dependent. More importantly, hepatomegaly is independent of PPARalpha, the thymic changes are partially dependent on this receptor, and splenic responses are largely eliminated in its absence. Thus, immunomodulation caused by PFOS is a high-dose phenomenon partially dependent on PPARalpha.


Subject(s)
Alkanesulfonic Acids/toxicity , Fluorocarbons/toxicity , PPAR alpha/deficiency , Spleen/drug effects , Spleen/pathology , Thymus Gland/drug effects , Thymus Gland/pathology , Alkanesulfonic Acids/blood , Animals , Atrophy/chemically induced , Atrophy/immunology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Body Weight/drug effects , Caprylates/toxicity , Cell Survival/drug effects , Dose-Response Relationship, Drug , Dose-Response Relationship, Immunologic , Flow Cytometry , Fluorocarbons/blood , Immunophenotyping , Liver/anatomy & histology , Liver/drug effects , Liver/immunology , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Organ Size/drug effects , PPAR alpha/immunology , Spleen/anatomy & histology , Spleen/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Thymus Gland/anatomy & histology , Thymus Gland/immunology
9.
Biochem J ; 412(1): 123-30, 2008 May 15.
Article in English | MEDLINE | ID: mdl-18215124

ABSTRACT

NF1 (nuclear factor 1) binds to two upstream elements of the human ANT2 (adenine nucleotide translocator-2) promoter and actively represses expression of the gene in growth-arrested diploid skin fibroblasts [Luciakova, Barath, Poliakova, Persson and Nelson (2003) J. Biol. Chem. 278, 30624-30633]. ChIP (chromatin immunoprecipitation) and co-immunoprecipitation analyses of nuclear extracts from growth-arrested and growth-activated diploid cells demonstrate that NF1, when acting as a repressor, is part of a multimeric complex that also includes Smad and Sp-family proteins. This complex appears to be anchored to both the upstream NF1-repressor elements and the proximal promoter, Sp1-dependent activation elements in growth-arrested cells. In growth-activated cells, the repressor complex dissociates and NF1 leaves the promoter. As revealed by co-immunoprecipitation experiments, NF1-Smad4-Sp3 complexes are present in nuclear extracts only from growth-inhibited cells, suggesting that the growth-state-dependent formation of these complexes is not an ANT2 promoter-specific event. Consistent with the role of Smad proteins in the repression complex, TGF-beta (transforming growth factor-beta) can fully repress ANT2 transcription in normally growing fibroblasts. Finally, pull-down experiments of in vitro transcribed/translated NF1 isoforms by GST (glutathione transferase)-Smad and GST-Smad MH fusion proteins indicate direct physical interactions between members of the two families. These findings suggest a possible functional relationship between the NF1 and Smad proteins that has not been previously observed.


Subject(s)
Adenine Nucleotide Translocator 2/genetics , Cell Proliferation , Down-Regulation , Neurofibromin 1/physiology , Smad Proteins/physiology , Sp Transcription Factors/physiology , Cell Proliferation/drug effects , Cells, Cultured , Culture Media, Serum-Free/pharmacology , Down-Regulation/drug effects , Humans , Models, Biological , Multigene Family/physiology , Multiprotein Complexes/metabolism , Multiprotein Complexes/physiology , Neurofibromin 1/metabolism , Promoter Regions, Genetic , Protein Binding/drug effects , Repressor Proteins/metabolism , Repressor Proteins/physiology , Smad Proteins/metabolism , Sp Transcription Factors/metabolism , Transcription, Genetic
10.
Article in English | MEDLINE | ID: mdl-15380819

ABSTRACT

Acyl-CoA thioesterases hydrolyze acyl-CoAs to free fatty acids and CoASH, thereby regulating fatty acid metabolism. This activity is catalyzed by numerous structurally related and unrelated enzymes, of which several acyl-CoA thioesterases have been shown to be regulated via the peroxisome proliferator-activated receptor alpha, strongly linking them to fatty acid metabolism. Two protein families have recently been characterized, the type I acyl-CoA thioesterase gene family and the type II protein family, which are expressed in cytosol, mitochondria and peroxisomes. Still, only little is known about regulation of their expression and precise functions in vivo. In the present study, we have investigated the activity and expression of acyl-CoA thioesterase in the porcine ovary during different phases of the estrus cycle. The activity was low in homogenates obtained during the immature and follicular phases, increasing nearly 4-fold during the luteal phase, with the highest activity being found in the pregnant corpus luteum (about 7-fold higher than in immature follicles). The increase in homogenate activity in corpus luteum from pregnant pigs was due to a moderate increase in the cytosolic activity, and an approximately 20-25-fold increase in the mitochondrial fraction. Western blot analysis showed no detectable expression of the type I acyl-CoA thioesterases (CTE-I and MTE-I) and revealed that the increased activity in cytosol and mitochondria is due to increased expression of the type II acyl-CoA thioesterases (CTE-II and MTE-II). This apparent hormonal regulation of expression of the type II acyl-CoA thioesterase may provide new insights into the functions of these enzymes in the mammalian ovary.


Subject(s)
Corpus Luteum/enzymology , Cytosol/enzymology , Fatty Acid Synthases/genetics , Gene Expression Regulation, Enzymologic , Mitochondria/enzymology , Thiolester Hydrolases/genetics , Adrenal Glands/enzymology , Adrenal Glands/ultrastructure , Animals , Blotting, Western , Corpus Luteum/ultrastructure , Fatty Acid Synthases/metabolism , Female , Ovary/enzymology , Ovary/ultrastructure , Pregnancy , Swine , Thiolester Hydrolases/metabolism
11.
Eur J Biochem ; 271(9): 1781-8, 2004 May.
Article in English | MEDLINE | ID: mdl-15096217

ABSTRACT

The human adenine nucleotide translocase-2 (ANT2) promoter contains a silencer region that confers partial repression on the heterologous herpes simplex virus thymidine kinase (HSVtk) promoter [Barath, P., Albert-Fournier, B., Luciakova, K., Nelson, B.D. (1999) J. Biol. Chem.274, 3378-3384]. Two sequences in the silencer (Site-2 and Site-3) are protected in the DNase I assay in vitro, and one of these is a repeated GTCCTG element previously shown to act as the active repressor element. We have now purified the DNA binding protein, and identified it using MALDI-TOF MS as a 33-kDa member of the nuclear factor 1 (NF1) family of transcription factors. NF1 purified from rat liver and HeLa cell nuclei bind to both silencer Site-2 and Site-3, resulting in a DNase I footprint identical to that obtained with purified recombinant NF1. Furthermore, transient transfection experiments with reporter constructs containing mutated silencer Site-2 and/or Site-3 show that both sites contribute to repression of the HSVtk promoter. Finally, chromatin immunoprecipitation analysis reveals that NF1 is bound to both elements on the endogenous HeLa cell ANT2 promoter. Our data support the belief that NF1 acts as a repressor when bound to silencing Site-2 and Site-3 of the ANT2 gene.


Subject(s)
Adenine Nucleotide Translocator 2/genetics , CCAAT-Enhancer-Binding Proteins/physiology , Repressor Proteins/physiology , Transcription Factors/physiology , Amino Acid Sequence , Base Sequence , DNA/metabolism , Gene Silencing , Humans , Molecular Sequence Data , NFI Transcription Factors , Promoter Regions, Genetic
12.
Chem Biol Interact ; 147(1): 23-34, 2004 Jan 15.
Article in English | MEDLINE | ID: mdl-14726150

ABSTRACT

A detailed subfractionation of the non-pregnant porcine corpus luteum (CL) was performed employing differential centrifugation. Marker enzyme assays (i.e., lactate dehydrogenase for the cytosol, NADPH-cytochrome P450 reductase for the endoplasmatic reticulum, catalase (CAT) for peroxisomes, glutamate dehydrogenase for the mitochondrial matrix and acid phosphatase for lysosomes) in all subfractions obtained exhibited a pattern of distribution similar to that observed with rat liver. These subfractions should be useful in connection with many types of future studies. In disagreement with previous biochemical and morphological studies, peroxisomes (identified on the basis of catalase activity and by Western blotting of catalase and of the major peroxisomal membrane protein (PMP-70)) sedimented together with mitochondria (i.e., at 5000 x g(av) for 10 min) and not in the post-mitochondrial fraction prepared at 30,000 x g(av) for 20 min by Peterson and Stevensson. No other classical peroxisomal enzymes were detectable in the porcine ovary, raising questions concerning the function of peroxisomes in this organ. Furthermore, UDP-glucuronosyltransferase (UGT), generally considered to be an integral membrane protein anchored in the endoplasmatic reticulum, was recovered in both the cytosolic (i.e., the supernatant after centrifugation at 50,000 x g(av) for 1h) and the microsomal fraction of the porcine corpus luteum, even upon further centrifugation of the former. In contrast, UGT sediments exclusively in the microsomal fraction upon subfractionation of the liver and ovary from rat.


Subject(s)
Cell Fractionation/methods , Corpus Luteum/enzymology , Glucuronosyltransferase/metabolism , Peroxisomes/enzymology , Animals , Catalase/metabolism , Centrifugation, Density Gradient , Corpus Luteum/metabolism , Cytosol/enzymology , Endoplasmic Reticulum/metabolism , Female , Membrane Proteins/metabolism , Mitochondria/metabolism , Subcellular Fractions/enzymology , Subcellular Fractions/metabolism , Swine
13.
Biochem Pharmacol ; 66(5): 749-56, 2003 Sep 01.
Article in English | MEDLINE | ID: mdl-12948855

ABSTRACT

We have previously demonstrated that severe adipose tissue atrophy occurs upon dietary treatment of mice with potent peroxisome proliferators (PPs). This atrophy occurs subsequent to peroxisome proliferation in the liver and may represent a novel addition to the pleiotropic effects exerted by PPs. In the present study we have characterized the recovery of mice from such atrophy following cessation of exposure. Following termination of treatment with perfluorooctanoic acid (PFOA) for 7 days, the adipose tissue atrophy was rapidly reversed, beginning on 2-5 days of recovery and being complete within 10 days. In contrast, hepatic peroxisome proliferation recovered much more slowly, indicating that these processes are not strictly coordinated. Analysis of lipoprotein lipase and hormone-sensitive lipase activities in adipose tissue revealed that the decrease and increase in these activities, respectively, caused by PFOA were both reversed within 10 days of recovery. Overall, these data provide further support for our previous conclusion that the adipose tissue atrophy induced by PFOA is caused, at least in part, by changes in the activities of lipoprotein lipase and hormone-sensitive lipase. The serum level of cholesterol, which increased after termination of PFOA treatment, returned to normal with a time-course similar to the recovery of adipose tissue weight, although hepatic peroxisome proliferation was still present. The possible relationship between the reduction in serum cholesterol and/or in its availability to peripheral tissues and the associated atrophy of adipose tissues caused by PPs is discussed.


Subject(s)
Adipose Tissue/drug effects , Caprylates/pharmacology , Fluorocarbons/pharmacology , Peroxisome Proliferators/pharmacology , Peroxisomes/drug effects , Substance Withdrawal Syndrome/metabolism , Adipose Tissue/pathology , Animals , Apolipoproteins/blood , Cholesterol/blood , Epidermis/drug effects , Epidermis/enzymology , Fatty Acids/metabolism , Lipids/blood , Lipoprotein Lipase/metabolism , Liver/drug effects , Liver/physiology , Male , Mice , Mice, Inbred C57BL , Mitochondria, Liver/drug effects , Mitochondria, Liver/metabolism , Organ Size/drug effects , Oxidation-Reduction , Peroxisomes/physiology , Substance Withdrawal Syndrome/blood , Time Factors , Triglycerides/blood
14.
J Biol Chem ; 278(33): 30624-33, 2003 Aug 15.
Article in English | MEDLINE | ID: mdl-12777383

ABSTRACT

Adenine nucleotide translocase-2 (ANT2) catalyzes the exchange of ATP for ADP across the mitochondrial membrane, thus playing an important role in maintaining the cytosolic phosphorylation potential required for cell growth. Expression of ANT2 is activated by growth stimulation of quiescent cells and is down-regulated when cells become growth-arrested. In this study, we address the mechanism of growth arrest repression. Using a combination of transfection, in vivo dimethyl sulfate mapping, and in vitro DNase I mapping experiments, we identified two protein-binding elements (Go-1 and Go-2) that are responsible for growth arrest of ANT2 expression in human diploid fibroblasts. Proteins that bound the Go elements were purified and identified by matrix-assisted laser desorption ionization time-of-flight mass spectrometry as members of the NF1 family of transcription factors. Chromatin immunoprecipitation analysis showed that NF1 was bound to both Go-1 and Go-2 in quiescent human diploid cells in vivo, but not in the same cells stimulated to growth by serum. NF1 binding correlated with the disappearance of ANT2 transcripts in quiescent cells. Furthermore, overexpression of NF1-A, -C, and -X in NIH3T3 cells repressed expression of an ANT2-driven reporter gene construct. Two additional putative repressor elements in the ANT2 promoter, an Sp1 element juxtaposed to the transcription start site and a silencer centered at nucleotide -332, did not appear to contribute to growth arrest repression. Thus, enhanced binding of NF1 is a key step in the growth arrest repression of ANT2 transcription. To our knowledge, this is the first report showing a role for NF1 in growth arrest.


Subject(s)
Adenine Nucleotide Translocator 2/genetics , DNA-Binding Proteins , Resting Phase, Cell Cycle/physiology , Transcription Factors , 3T3 Cells , Animals , Base Sequence , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Division/physiology , Diploidy , Down-Regulation/physiology , Humans , Mice , Molecular Sequence Data , Mutagenesis/physiology , NFI Transcription Factors , Nuclear Proteins , Promoter Regions, Genetic/physiology , Skin/cytology , Suppression, Genetic , Transcription, Genetic/physiology , Y-Box-Binding Protein 1
15.
Biochem Pharmacol ; 63(10): 1893-900, 2002 May 15.
Article in English | MEDLINE | ID: mdl-12034374

ABSTRACT

Peroxisome proliferators (PPs) are a large class of structurally diverse chemicals, which includes drugs designed to improve the metabolic abnormalities linking hypertriglyceridemia to diabetes, hyperglycemia, insulin-resistance and atherosclerosis. We have recently demonstrated that exposure of rodents to potent PPs indirectly causes a number of immunomodulating effects, resulting in severe adaptive immunosuppression. Since the peroxisome proliferator-activated receptor alpha (PPARalpha) plays a central role in mediating the pleiotropic responses exerted by PPs, we have compared here the immunomodulating effects of the PPs perfluorooctanoic acid (PFOA) and Wy-14,643 in wild-type and PPARalpha-null mice. The reductions in spleen weight and in the number of splenocytes caused by PP treatment in wild-type mice was not observed in PPARalpha-null mice. Furthermore, the reductions in thymus weight and in the number of thymocytes were potently attenuated in the latter animals. Similarly, the dramatic decreases in the size of the CD4(+)CD8(+) population of cells in the thymus and in the number of thymocytes in the S and G2/M phases of the cell cycle observed in wild-type mice administered PPs were much less extensive in PPARalpha-null mice. Finally, in contrast to the case of wild-type animals, the response of splenocytes isolated from the spleen of PP-treated PPARalpha-null mice to appropriate T- or B-cell activators in vitro was not reduced. Altogether, these data indicate that PPARalpha plays a major role in the immunomodulation caused by PPs. The possible relevance of these changes to the alterations in plasma lipids also caused by PPs is discussed.


Subject(s)
Adjuvants, Immunologic/pharmacology , Caprylates/pharmacology , Fluorocarbons/pharmacology , Peroxisome Proliferators/pharmacology , Pyrimidines/pharmacology , Receptors, Cytoplasmic and Nuclear/immunology , Transcription Factors/immunology , Animals , Cell Cycle/drug effects , Cell Division/drug effects , Cells, Cultured , Liver/drug effects , Liver/physiology , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Models, Animal , Organ Size/drug effects , Peroxisomes/drug effects , Spleen/drug effects , Spleen/physiology , Thymus Gland/drug effects , Thymus Gland/physiology
16.
Lipids ; 37(2): 139-46, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11908906

ABSTRACT

In the present study, we characterized the effects of peroxisome proliferators (PP) on adipose tissue in mice. Treatment with potent PP, such as perfluorooctanoic acid (PFOA), 2-methyl-2-(p(1,2,3,4-tetrahydroxy-naphthyl)-phenoxy)propionic acid, (4-chloro-6-(2,3-xylidino)2-pyrimidinylthio) acetic acid, and di(2-ethylhexyl)phthalate, caused dramatic decreases in adipose tissue weight, whereas the moderately potent PP, acetylsalicylic acid, had a relatively weak effect. This decrease in weight reflects a loss of fat from adipocytes rather than a loss of cells, as demonstrated by constant DNA content. The dose-dependency and time-course experiments indicate that peroxisome proliferation occurs simultaneously with or prior to adipose tissue atrophy. Thus, hepatic peroxisome proliferation might result in the increased mobilization of lipids and lipid utilization in liver. The enhanced adipose tissue hormone-sensitive lipase (HSL) activity and down-regulated lipoprotein lipase (LPL) activity observed upon PP treatment might, at least in part, explain the loss of fat via increased FA release from adipocytes and/or decreased FA uptake from the circulation, respectively. In addition, the possible involvement of the increased tumor necrosis factor alpha expression found upon PFOA treatment in reducing the insulin sensitivity of adipose tissue and thereby altering LPL and HSL activities is discussed.


Subject(s)
Adipose Tissue/drug effects , Peroxisome Proliferators/pharmacology , Adipose Tissue/enzymology , Adipose Tissue/pathology , Animals , Blotting, Western , Electrophoresis, Polyacrylamide Gel , Lipolysis , Lipoprotein Lipase/metabolism , Liver/drug effects , Male , Mice , Mice, Inbred C57BL , Organ Size/drug effects , Sterol Esterase/metabolism , Tumor Necrosis Factor-alpha/metabolism
17.
Int Immunopharmacol ; 2(2-3): 389-97, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11811941

ABSTRACT

In a previous investigation, we demonstrated that severe thymus and spleen atrophy occurs in mice upon dietary exposure to several potent peroxisome proliferators (PPs). In the present investigation, the effects of the potent PP perfluorooctanoic acid (PFOA) on the adaptive immunity of mice was evaluated both in vivo and ex vivo. The in vivo immune response examined involved immunization of mice with horse red blood cells (HRBCs), displaying T-cell-dependent antigens after pre-treatment with a PFOA-containing diet for 10 days. Subsequent quantitation of the primary humoral response was performed employing both the plaque-forming cell (PFC) assay and determination of the antibody titer by ELISA. The results clearly demonstrate that oral administration of PFOA prevents both the increases in plaque formations by anti-IgM and -IgG and in serum levels of IgM and IgG normally evoked by such immunization. Ex vivo spleen cells proliferation (assayed as incorporation of 3H-thymidine) in response to both T- and B-cell activators was attenuated by dietary treatment with PFOA, although the analogous in vitro treatment of mouse spleen cells with this same compound had no such effects. Thus, the relatively metabolically inert PP PFOA may exert adaptive immunosuppression in mice by an indirect mechanism. The possible relevance of this immunosuppression to the alterations in plasma lipids caused by PPs is discussed.


Subject(s)
Caprylates/administration & dosage , Caprylates/pharmacology , Diet/adverse effects , Fluorocarbons/administration & dosage , Fluorocarbons/pharmacology , Immunosuppressive Agents/pharmacology , Peroxisome Proliferators/administration & dosage , Peroxisome Proliferators/pharmacology , Administration, Oral , Animals , Antibody Formation/drug effects , Cells, Cultured/drug effects , Cells, Cultured/immunology , Erythrocytes/immunology , Horses , Immunization , Lymphocyte Activation/drug effects , Male , Mice , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL
...