Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Mol Cell Biochem ; 478(3): 581-595, 2023 Mar.
Article in English | MEDLINE | ID: mdl-35976519

ABSTRACT

We evaluated the mechanistic link between circadian rhythms and gut barrier permeability. Mice were subjected to either constant 24-h light (LL) or 12-h light/dark cycles (LD). Mice housed in LL experienced a significant increase in gut barrier permeability that was associated with dysregulated ß-catenin expression and altered expression of tight junction (TJ) proteins. Silencing of ß-catenin resulted in disruption of barrier function in SW480 cells, with ß-catenin appearing to be an upstream regulator of the core circadian components, such as Bmal1, Clock, and Per1/2. In addition, ß-catenin silencing downregulated ZO-1 and occludin TJ proteins with only limited or no changes at their mRNA levels, suggesting post transcriptional regulation. Indeed, silencing of ß-catenin significantly upregulated expression of matrix metallopeptidase (MMP)-2 and MMP-9, and blocking MMP-2/9 activity attenuated epithelial disruption induced by ß-catenin silencing. These results indicate the regulatory role of circadian disruption on gut barrier integrity and the associations between TJ proteins and circadian rhythms, while demonstrating the regulatory role of ß-catenin in this process.


Subject(s)
Catenins , Circadian Rhythm , Animals , Mice , Catenins/genetics , Gene Expression Regulation
2.
Toxicol Appl Pharmacol ; 287(3): 258-66, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26080028

ABSTRACT

Occludin is an essential integral transmembrane protein regulating tight junction (TJ) integrity in brain endothelial cells. Phosphorylation of occludin is associated with its localization to TJ sites and incorporation into intact TJ assembly. The present study is focused on the role of lipid rafts in polychlorinated biphenyl (PCB)-induced disruption of occludin and endothelial barrier function. Exposure of human brain endothelial cells to 2,2',4,4',5,5'-hexachlorobiphenyl (PCB153) induced dephosphorylation of threonine residues of occludin and displacement of occludin from detergent-resistant membrane (DRM)/lipid raft fractions within 1h. Moreover, lipid rafts modulated the reduction of occludin level through activation of matrix metalloproteinase 2 (MMP-2) after 24h PCB153 treatment. Inhibition of protein phosphatase 2A (PP2A) activity by okadaic acid or fostriecin markedly protected against PCB153-induced displacement of occludin and increased permeability of endothelial cells. The implication of lipid rafts and PP2A signaling in these processes was further defined by co-immunoprecipitation of occludin with PP2A and caveolin-1, a marker protein of lipid rafts. Indeed, a significant MMP-2 activity was observed in lipid rafts and was increased by exposure to PCB153. The pretreatment of MMP-2 inhibitors protected against PCB153-induced loss of occludin and disruption of lipid raft structure prevented the increase of endothelial permeability. Overall, these results indicate that lipid raft-associated processes, such as PP2A and MMP-2 activation, participate in PCB153-induced disruption of occludin function in brain endothelial barrier. This study contributes to a better understanding of the mechanisms leading to brain endothelial barrier dysfunction in response to exposure to environmental pollutants, such as ortho-substituted PCBs.


Subject(s)
Blood-Brain Barrier/drug effects , Capillary Permeability/drug effects , Endothelial Cells/drug effects , Environmental Pollutants/toxicity , Matrix Metalloproteinase 2/metabolism , Membrane Microdomains/drug effects , Occludin/metabolism , Polychlorinated Biphenyls/toxicity , Protein Phosphatase 2/metabolism , Animals , Blood-Brain Barrier/enzymology , Blood-Brain Barrier/pathology , Caveolin 1/metabolism , Cell Line , Dose-Response Relationship, Drug , Endothelial Cells/metabolism , Endothelial Cells/pathology , Humans , Male , Membrane Microdomains/enzymology , Membrane Microdomains/pathology , Mice, Inbred C57BL , Signal Transduction/drug effects , Time Factors
3.
J Nutr Biochem ; 26(2): 120-9, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25465156

ABSTRACT

Selenium-containing compounds and selenized yeast have anticancer properties. In order to address possible mechanisms involved in these effects, selenoglycoproteins (SGPs) were extracted from selenium-enriched yeast at pH 4.0 and 6.5 (the fractions are called SGP40 and SGP65, respectively), followed by evaluation of their impact on the interactions of lung and breast tumor cells with human brain microvascular endothelial cells (HBMECs). Extracted SGPs, especially SGP40, significantly inhibited adhesion of tumor cells to HBMECs and their transendothelial migration. Because the active components of SGPs are unknown, small selenium-containing compounds [leucyl-valyl-selenomethionyl-arginine (LVSe-MR) and methylselenoadenosine (M-Se-A)], which are normally present in selenized yeast, were introduced as additional treatment groups. Treatment of HBMECs with SGP40, LVSe-MR and M-Se-A induced changes in gene signatures, which suggested a central involvement of nuclear factor (NF)-κB-dependent pathway. These observations were confirmed in the subsequent analysis of NF-κB DNA binding activity, quantitative measurements of the expression of selected genes and proteins, and tumor cell adhesion assay with a specific NF-κB inhibitor as the additional treatment factor. These findings indicate that specific organic selenium-containing compounds have the ability to inhibit tumor cell adhesion to brain endothelial cells via down-regulation of NF-κB. SGPs appear to be more effective than small selenium-containing compounds, suggesting the role of not only selenium but also the glycoprotein component in the observed protective impact.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Endothelium, Vascular/drug effects , Glycoproteins/pharmacology , Lung Neoplasms/drug therapy , Saccharomyces cerevisiae Proteins/pharmacology , Selenoproteins/pharmacology , Antineoplastic Agents/isolation & purification , Antineoplastic Agents/metabolism , Brain/blood supply , Brain/cytology , Brain/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Adhesion/drug effects , Cell Line , Cell Line, Tumor , Endothelium, Vascular/cytology , Female , Gene Expression Regulation, Neoplastic/drug effects , Glycoproteins/biosynthesis , Glycoproteins/isolation & purification , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Microvessels/cytology , Microvessels/drug effects , NF-kappa B/agonists , NF-kappa B/genetics , NF-kappa B/metabolism , Neoplasm Proteins/agonists , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Organoselenium Compounds/isolation & purification , Organoselenium Compounds/metabolism , Organoselenium Compounds/pharmacology , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/biosynthesis , Saccharomyces cerevisiae Proteins/isolation & purification , Selenium/metabolism , Selenomethionine/analogs & derivatives , Selenomethionine/isolation & purification , Selenomethionine/metabolism , Selenomethionine/pharmacology , Selenoproteins/biosynthesis , Selenoproteins/isolation & purification , Transendothelial and Transepithelial Migration/drug effects
4.
Arch Med Res ; 45(8): 744-52, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25446617

ABSTRACT

BACKGROUND AND AIMS: Increased amyloid deposition in HIV-infected brains may contribute to the pathogenesis of neurocognitive dysfunction in infected patients. We have previously shown that exposure to HIV results in enhanced amyloid ß (Aß) levels in human brain microvascular endothelial cells, suggesting that brain endothelial cells contribute to accumulation of Aß in HIV-infected brains. Importantly, Aß not only accumulates in the cytoplasm of HIV-exposed cells but also enters the nuclei of brain endothelial cells. METHODS: cDNA microarray analysis was performed in order to examine changes in the transcriptional profile associated with Aß nuclear entry in the presence of HIV-1. RESULTS: Gene network analysis indicated that inhibition of nuclear entry of Aß resulted in enrichment in gene sets involved in apoptosis and survival, endoplasmic reticulum stress response, immune response, cell cycle, DNA damage, oxidative stress, cytoskeleton remodeling and transforming growth factor ß (TGFß) receptor signaling. CONCLUSIONS: The obtained data indicate that HIV-induced Aß nuclear uptake affects several cellular stress-related pathways relevant for HIV-induced Aß pathology.


Subject(s)
Active Transport, Cell Nucleus/genetics , Amyloid beta-Peptides/metabolism , Brain/pathology , HIV Infections/metabolism , HIV-1 , Protein Transport/genetics , Apoptosis/genetics , Brain/metabolism , Cell Line , Cells, Cultured , Dynamins/antagonists & inhibitors , Endoplasmic Reticulum Stress/genetics , Endothelial Cells/metabolism , HEK293 Cells , Humans , Oxidative Stress/genetics , Signal Transduction/genetics , Transforming Growth Factor beta/metabolism
5.
Am J Physiol Gastrointest Liver Physiol ; 306(11): G992-G1001, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24742991

ABSTRACT

The intestinal epithelium forms a selective barrier maintained by tight junctions (TJs) and separating the luminal environment from the submucosal tissues. N-acylhomoserine lactone (AHL) quorum-sensing molecules produced by gram-negative bacteria in the gut can influence homeostasis of the host intestinal epithelium. In the present study, we evaluated the regulatory mechanisms affecting the impact of two representative long- and short-chain AHLs, N-3-(oxododecanoyl)-homoserine lactone (C12-HSL) and N-butyryl homoserine lactone (C4-HSL), on barrier function of human intestinal epithelial Caco-2 cells. Treatment with C12-HSL, but not with C4-HSL, perturbed Caco-2 barrier function; the effect was associated with decreased levels of the TJ proteins occludin and tricellulin and their delocalization from the TJs. C12-HSL also induced matrix metalloprotease (MMP)-2 and MMP-3 activation via lipid raft- and protease-activated receptor (PAR)-dependent signaling. Pretreatment with lipid raft disruptors, PAR antagonists, or MMP inhibitors restored the C12-HSL-induced loss of the TJ proteins and increased permeability of Caco-2 cell monolayers. These results indicate that PAR/lipid raft-dependent MMP-2 and -3 activation followed by degradation of occludin and tricellulin are involved in C12-HSL-induced alterations of epithelial paracellular barrier functions.


Subject(s)
Epithelial Cells/drug effects , Homoserine/analogs & derivatives , Intestinal Mucosa/physiology , Lactones/pharmacology , Matrix Metalloproteinases/metabolism , Acyl-Butyrolactones/pharmacology , Caco-2 Cells , Enzyme Activation , Gene Expression Regulation , Homoserine/pharmacology , Humans , Intestinal Mucosa/cytology , MARVEL Domain Containing 2 Protein/metabolism , Matrix Metalloproteinases/genetics , Membrane Microdomains/physiology , Occludin/metabolism , Permeability , Tight Junction Proteins/metabolism
6.
Environ Health Perspect ; 121(6): 725-30, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23632211

ABSTRACT

BACKGROUND: The gut microbiome, a dynamic bacterial community that interacts with the host, is integral to human health because it regulates energy metabolism and immune functions. The gut microbiome may also play a role in risks from environmental toxicants. OBJECTIVES: We investigated the effects of polychlorinated biphenyls (PCBs) and exercise on the composition and structure of the gut microbiome in mice. METHODS: After mice exercised voluntarily for 5 weeks, they were treated by oral gavage with a mixture of environmentally relevant PCB congeners (PCB153, PCB138, and PCB180; total PCB dose, 150 µmol/kg) for 2 days. We then assessed the microbiome by determination of 16S rRNA using microarray analysis. RESULTS: Oral exposure to PCBs significantly altered the abundance of the gut microbiome in mice primarily by decreasing the levels of Proteobacteria. The activity level of the mice correlated with a substantial shift in abundance, biodiversity, and composition of the microbiome. Importantly, exercise attenuated PCB-induced changes in the gut microbiome. CONCLUSIONS: Our results show that oral exposure to PCBs can induce substantial changes in the gut microbiome, which may then influence their systemic toxicity. These changes can be attenuated by behavioral factors, such as voluntary exercise.


Subject(s)
Environmental Pollutants/toxicity , Intestines/microbiology , Metagenome/drug effects , Physical Conditioning, Animal , Polychlorinated Biphenyls/toxicity , Animals , Cholic Acid/pharmacology , Fatty Acids, Volatile/pharmacology , Humans , Male , Mice , Mice, Inbred C57BL
7.
PLoS One ; 8(5): e63159, 2013.
Article in English | MEDLINE | ID: mdl-23690990

ABSTRACT

PCBs bind to environmental particles; however, potential toxicity exhibited by such complexes is not well understood. The aim of the present study is to study the hypothesis that assembling onto nanoparticles can influence the PCB153-induced brain endothelial toxicity via interaction with the toll-like receptor 4 (TLR4). To address this hypothesis, TLR4-deficient and wild type control mice (males, 10 week old) were exposed to PCB153 (5 ng/g body weight) bound to chemically inert silica nanoparticles (PCB153-NPs), PCB153 alone, silica nanoparticles (NPs; diameter, 20 nm), or vehicle. Selected animals were also subjected to 40 min ischemia, followed by a 24 h reperfusion. As compared to exposure to PCB153 alone, treatment with PCB153-NP potentiated the brain infarct volume in control mice. Importantly, this effect was attenuated in TLR4-deficient mice. Similarly, PCB153-NP-induced proinflammatory responses and disruption of tight junction integrity were less pronounced in TLR4-deficient mice as compared to control animals. Additional in vitro experiments revealed that TLR4 mediates toxicity of PCB153-NP via recruitment of tumor necrosis factor-associated factor 6 (TRAF6). The results of current study indicate that binding to seemingly inert nanoparticles increase cerebrovascular toxicity of PCBs and suggest that targeting the TLR4/TRAF6 signaling may protect against these effects.


Subject(s)
Blood Vessels/drug effects , Brain/blood supply , Nanoparticles/chemistry , Polychlorinated Biphenyls/chemistry , Polychlorinated Biphenyls/toxicity , Signal Transduction/drug effects , Toll-Like Receptor 4/metabolism , Animals , Blood Vessels/metabolism , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Brain/drug effects , Brain/metabolism , Cerebral Infarction/pathology , Environmental Pollutants/chemistry , Environmental Pollutants/toxicity , Gene Expression Regulation/drug effects , Male , Mice , TNF Receptor-Associated Factor 6/metabolism , Tight Junctions/drug effects , Tight Junctions/metabolism , Toll-Like Receptor 4/deficiency
8.
Toxicology ; 302(2-3): 212-20, 2012 Dec 16.
Article in English | MEDLINE | ID: mdl-22906770

ABSTRACT

Exposure to polychlorinated biphenyls (PCBs) is associated with numerous adverse health effects. Although the main route of exposure to PCBs is through the gastrointestinal tract, little is known about the contribution of the gut to the health effects of PCBs. We hypothesize that PCBs can disrupt intestinal integrity, causing lipopolysaccharide (LPS) translocation into the bloodstream and potentiation of the systemic toxicity of PCBs. C57BL/6 mice were exposed to individual PCB congeners by oral gavage, followed by the assessment of small intestine morphology and plasma levels of proinflammatory mediators. In addition, mice and human brain endothelial cells were exposed to PCB118 in the presence or absence of LPS to evaluate the contribution of LPS to PCB-induced toxicity at the blood-brain barrier (BBB) level. Oral administration of PCB153, PCB118, or PCB126 disrupted intestinal morphology and increased plasma levels of LPS and proinflammatory cytokines. Direct injection of LPS and PCB118 into the cerebral microvasculature resulted in synergistic disruption of BBB integrity and decreased expression of tight junction proteins in brain microvessels. In vitro experiments confirmed these effects and indicated that stimulation of the toll-like receptor 4 (TLR4) pathway can be responsible for these effects via activation of interferon regulatory factor-3 (IRF-3). These results indicate that LPS may be a contributing factor in PCB-induced dysfunction of the brain endothelium via stimulation of the TLR4/IRF-3 pathway.


Subject(s)
Interferon Regulatory Factor-3/metabolism , Lipopolysaccharides/adverse effects , Polychlorinated Biphenyls/toxicity , Signal Transduction , Toll-Like Receptor 4/metabolism , Administration, Oral , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Cell Line , Cell Movement , Cytokines/blood , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Gene Silencing , Humans , Interferon Regulatory Factor-3/genetics , Lipopolysaccharides/blood , Male , Mice , Mice, Inbred C57BL , Polychlorinated Biphenyls/administration & dosage , Toll-Like Receptor 4/genetics
9.
Biochem Biophys Res Commun ; 421(2): 177-83, 2012 May 04.
Article in English | MEDLINE | ID: mdl-22490665

ABSTRACT

Amyloid beta (Aß) levels are increased in HIV-1 infected brains due to not yet fully understood mechanisms. In the present study, we investigate the role of lipid rafts, functional caveolae, and caveolae-associated signaling in HIV-1-induced Aß accumulation in HBMEC. Both silencing of caveolin-1 (cav-1) and disruption of lipid rafts by pretreatment with beta-methyl-cyclodextrin (MCD) protected against Aß accumulation in HBMEC. Exposure to HIV-1 and Aß activated caveolae-associated Ras and p38. While inhibition of Ras by farnesylthiosalicylic acid (FTS) effectively protected against HIV-1-induced accumulation of Aß, blocking of p38 did not have such an effect. We also evaluated the role of caveolae in HIV-1-induced upregulation of the receptor for advanced glycation end products (RAGE), which regulates Aß transfer from the blood stream into the central nervous system. HIV-1-induced RAGE expression was prevented by infecting HBMEC with cav-1 specific shRNA lentiviral particles or by pretreatment of cells with FTS. Overall, the present results indicate that Aß accumulation in HBMEC is lipid raft and caveolae dependent and involves the caveolae-associated Ras signaling.


Subject(s)
Amyloid beta-Peptides/metabolism , Brain/metabolism , Endothelial Cells/metabolism , HIV Infections/metabolism , HIV-1 , Membrane Microdomains/metabolism , Brain/virology , Caveolae/metabolism , Cells, Cultured , Endothelial Cells/virology , Enzyme Inhibitors/pharmacology , Farnesol/analogs & derivatives , Farnesol/pharmacology , Humans , Receptor for Advanced Glycation End Products , Receptors, Immunologic/metabolism , Salicylates/pharmacology , ras Proteins/antagonists & inhibitors , ras Proteins/metabolism
10.
J Neurosci ; 32(1): 143-50, 2012 Jan 04.
Article in English | MEDLINE | ID: mdl-22219277

ABSTRACT

The human immunodeficiency virus (HIV)-specific protein trans-activator of transcription (Tat) can contribute to the dysfunction of brain endothelial cells and HIV trafficking into the brain by disrupting tight junction (TJ) integrity at the blood-brain barrier (BBB) level. Specific TJ proteins, such as zonula occludens (ZO) proteins, localize not only at the cell-cell borders but are also present in the nuclei. The objective of the present study was to evaluate the mechanisms and significance of Tat-induced nuclear localization of ZO-1. Treatment of a brain endothelial cell line (hCMEC/D3 cells) with Tat resulted in a decrease in total levels of ZO-1 but significantly upregulated ZO-1 protein expression in the nuclei. In addition, exposure to Tat stimulated Rho signaling and induced phosphorylation and activity of transcription factor cAMP response element-binding protein (CREB), binding sites that have been identified in the proximal region of the ZO-1 promoter. Interestingly, inhibition of the Rho cascade protected against Tat-induced upregulation of ZO-1 in the nuclei and activation of CREB. Depletion of CREB by infection of cells with specific shRNA lentiviral particles attenuated both Tat-induced Rho signaling and nuclear targeting of ZO-1. A decrease in CREB levels also attenuated Tat-induced endothelial and BBB hyperpermeability as well as transendothelial migration of monocytic cells. The role of CREB in Tat-mediated alterations of ZO-1 was confirmed in brain microvessels in mice with CREB shRNA lentiviral particles injected into the cerebral circulation. The present results indicate the crucial role of Rho signaling and CREB in modulation of nuclear localization of ZO-1 and maintaining the integrity of endothelial monolayers.


Subject(s)
Blood-Brain Barrier/metabolism , Cell Nucleus/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Endothelial Cells/metabolism , Membrane Proteins/metabolism , Phosphoproteins/metabolism , Signal Transduction/physiology , rho GTP-Binding Proteins/physiology , tat Gene Products, Human Immunodeficiency Virus/physiology , Animals , Blood-Brain Barrier/virology , Cell Nucleus/virology , Endothelial Cells/cytology , Endothelial Cells/virology , HEK293 Cells , HIV-1/physiology , Humans , Male , Mice , Mice, Inbred C57BL , Primary Cell Culture , Zonula Occludens-1 Protein
11.
Toxicol Sci ; 126(2): 362-71, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22240979

ABSTRACT

Polychlorinated biphenyls (PCBs) are environmental toxicants that cause vascular inflammation and facilitate the development of brain metastases. The crucial event in metastasis formation is adhesion of blood-borne tumor cells to the vascular endothelium, followed by their transcapillary migration. The aim of the present study was to examine the mechanisms of PCB118-induced brain metastasis formation at the blood-brain barrier level with the focus on tumor cell adhesion to the brain endothelium. PCB118 was administered orally to wild-type or intercellular cell adhesion molecule-1 (ICAM-1)-deficient mice, followed by an injection of Lewis lung carcinoma cells into the carotid artery. Treatment with PCB118 resulted in enhanced development of brain metastases. Injection of tumor cells induced overexpression of ICAM-1 and vascular endothelial cell adhesion molecule-1 (VCAM-1) in brain endothelium that was further potentiated in mice exposed to PCB118. PCB118 did not affect the number of adhered and extravasated tumor cells in ICAM-1-deficient mice. Additional in vitro studies indicated that VCAM-1-neutralizing antibody protected against PCB118-induced adhesion of tumor cells to cultured brain endothelial cells. These results indicate that exposure to selected PCB congeners, such as PCB118, induces adhesion and transcapillary migration of tumor cells. This process is facilitated by proinflammatory adhesion molecules and results in potentiation of brain metastasis formation.


Subject(s)
Brain Neoplasms/secondary , Cell Adhesion Molecules/physiology , Inflammation Mediators/physiology , Polychlorinated Biphenyls/toxicity , Animals , Brain Neoplasms/chemically induced , Brain Neoplasms/pathology , Cell Adhesion , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
12.
J Neurosci Res ; 88(13): 2859-68, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20564349

ABSTRACT

Epidemiology and genetic studies indicate that patients with telomere length shorter than average are at higher risk of dying from heart disease or stroke. Telomeres are located at the ends of eukaryotic chromosomes, which demonstrate progressive length reduction in most somatic cells during aging. The enzyme telomerase can compensate for telomere loss during cell replication. The present study sought to investigate the contribution of telomerase to stroke and blood-brain barrier (BBB) dysfunction. Telomerase reverse transcriptase knockout (TERT(-/-)) mice and littermate controls with normal TERT expression were subjected to a 24-hr permanent middle cerebral artery occlusion (pMCAO). The stroke outcomes were assessed in terms of neurological scores and infarct volumes. In addition, we evaluated oxidative stress, permeability across the BBB, and integrity of tight junctions in brain microvessels. Neurological testing revealed that TERT(-/-) mice showed enhanced deficits compared with controls. These changes were associated with a greater infarct volume. The expression of tight junction protein ZO-1 decreased markedly in ischemic hemispheres of TERT(-/-) mice. The brain microvessels of TERT(-/-) mice also were more susceptible to oxidative stress, revealing higher superoxide and lower glutathione levels compared with mice with normal TERT expression. Importantly, TERT deficiency potentiated the production of inflammatory mediators, such as tumor necrosis factor-alpha, interleukin-1 beta, and intercellular adhesion molecule-1, in the ischemic hemispheres of mice with pMCAO. Our study suggests that TERT deficiency can predispose to the development of stroke in an experimental model of this disease.


Subject(s)
Blood-Brain Barrier/physiopathology , Encephalitis/etiology , Encephalitis/genetics , Infarction, Middle Cerebral Artery/complications , Telomerase/deficiency , Animals , Brain/pathology , Brain Infarction/etiology , Brain Infarction/pathology , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Gene Expression Regulation/genetics , Glutathione/metabolism , Mice , Mice, Knockout , Microvessels/physiopathology , Nervous System Diseases/etiology , Nervous System Diseases/genetics , Oxidative Stress/genetics , Oxidative Stress/physiology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA, Messenger/metabolism , Statistics, Nonparametric , Superoxides/metabolism
13.
Toxicol Appl Pharmacol ; 246(1-2): 74-82, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20406653

ABSTRACT

Exposure to environmental contaminants, such as polychlorinated biphenyls (PCBs), is a risk factor for the development of cardiovascular diseases such as atherosclerosis. Vascular cell adhesion molecule-1 (VCAM-1) is a critical mediator for adhesion and uptake of monocytes across the endothelium in the early stages of atherosclerosis development. The upregulation of VCAM-1 by PCBs may be dependent on functional membrane domains called caveolae. Caveolae are particularly abundant in endothelial cell membranes and involved in trafficking and signal transduction. The objective of this study was to investigate the role of caveolae in PCB-induced endothelial cell dysfunction. Primary mouse aortic endothelial cells (MAECs) isolated from caveolin-1-deficient mice and background C57BL/6 mice were treated with coplanar PCBs, such as PCB77 and PCB126. In addition, siRNA gene silencing technique was used to knockdown caveolin-1 in porcine vascular endothelial cells. In MAECs with functional caveolae, VCAM-1 protein levels were increased after exposure to both coplanar PCBs, whereas expression levels of VCAM-1 were not significantly altered in cells deficient of caveolin-1. Furthermore, PCB-induced monocyte adhesion was attenuated in caveolin-1-deficient MAECs. Similarly, siRNA silencing of caveolin-1 in porcine endothelial cells confirmed the caveolin-1-dependent VCAM-1 expression. Treatment of cells with PCB77 and PCB126 resulted in phosphorylation of extracellular signal-regulated kinase-1/2 (ERK1/2), and pharmacological inhibition of ERK1/2 diminished the observed PCB-induced increase in monocyte adhesion. These findings suggest that coplanar PCBs induce adhesion molecule expression, such as VCAM-1, in endothelial cells, and that this response is regulated by caveolin-1 and functional caveolae. Our data demonstrate a critical role of functional caveolae in the activation and dysfunction of endothelial cells by coplanar PCBs.


Subject(s)
Caveolin 1/deficiency , Endothelial Cells/drug effects , Polychlorinated Biphenyls/pharmacology , Vascular Cell Adhesion Molecule-1/biosynthesis , Animals , Aorta , Blotting, Western , Caveolin 1/genetics , Cell Adhesion/drug effects , Endothelial Cells/chemistry , Endothelial Cells/metabolism , Fluorescent Antibody Technique , Gene Expression/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Monocytes/drug effects , RNA Interference/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Swine , Vascular Cell Adhesion Molecule-1/analysis
14.
Environ Health Perspect ; 118(7): 976-81, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20299304

ABSTRACT

BACKGROUND: Polychlorinated biphenyls (PCBs) are widely distributed environmental toxicants that contribute to numerous disease states. The main route of exposure to PCBs is through the gastrointestinal tract; however, little is known about the effects of PCBs on intestinal epithelial barrier functions. OBJECTIVE: The aim of the present study was to address the hypothesis that highly chlorinated PCBs can disrupt gut integrity at the level of tight junction (TJ) proteins. METHODS: Caco-2 human colon adenocarcinoma cells were exposed to one of the following PCB congeners: PCB153, PCB118, PCB104, and PCB126. We then assessed NAD(P)H oxidase (NOX) activity and expression and the barrier function of Caco-2 cells. In addition, the integrity of intestinal barrier function and expression of TJ proteins were evaluated in C57BL/6 mice exposed to individual PCBs by oral gavage. RESULTS: Exposure of Caco-2 cells to individual PCB congeners resulted in activation of NOX and increased permeability of fluorescein isothiocyanate (FITC)-labeled dextran (4 kDa). Treatment with PCB congeners also disrupted expression of TJ proteins zonula occludens-1 (ZO-1) and occludin in Caco-2 cells. Importantly, inhibition of NOX by apocynin significantly protected against PCB-mediated increase in epithelial permeability and alterations of ZO-1 protein expression. Exposure to PCBs also resulted in alterations of gut permeability via decreased expression of TJ proteins in an intact physiological animal model. CONCLUSIONS: These results suggest that oral exposure to highly chlorinated PCBs disrupts intestinal epithelial integrity and may directly contribute to the systemic effects of these toxicants.


Subject(s)
Environmental Pollutants/toxicity , Intestinal Mucosa/drug effects , NADPH Oxidases/metabolism , Polychlorinated Biphenyls/toxicity , Tight Junctions/drug effects , Analysis of Variance , Animals , Blotting, Western , Caco-2 Cells , Electrophoresis, Polyacrylamide Gel , Enzyme Activation/drug effects , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Immunoprecipitation , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Occludin , Phosphoproteins/metabolism , Zonula Occludens-1 Protein
15.
Mol Cell Neurosci ; 43(2): 232-43, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19944163

ABSTRACT

HIV-1-infected brains are characterized by increased amyloid deposition. To study the influence of HIV-1 on amyloid beta (Abeta) homeostasis at the blood-brain barrier (BBB) level, we employed a model of brain microvascular endothelial cells exposed to HIV-1 in the presence or absence of Abeta. HIV-1 markedly increased endogenous Abeta levels and elevated accumulation of exogenous Abeta. Simvastatin, the HMG-CoA reductase inhibitor, blocked these effects. We next evaluated the effects of HIV-1 and/or simvastatin on expression of the receptor for lipoprotein related protein (LRP1) and the receptor for advanced glycation end products (RAGE), known to regulate Abeta transport across the BBB. LRP1 expression was not affected by HIV-1; however, it was increased by simvastatin. Importantly, simvastatin attenuated HIV-1-induced RAGE expression. These results suggest that HIV-1 may directly contribute to Abeta accumulation at the BBB level. In addition, statins may protect against increased Abeta levels associated with HIV-1 infection in the brain.


Subject(s)
Amyloid beta-Peptides/metabolism , Endothelial Cells , HIV-1/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Simvastatin/pharmacology , Amyloid beta-Peptides/pharmacology , Brain/cytology , Cell Line, Transformed , Coculture Techniques/methods , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/virology , Gene Expression Regulation/drug effects , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Peptide Fragments/pharmacology , Receptor for Advanced Glycation End Products , Receptors, Immunologic/metabolism , Time Factors
16.
Toxicol Appl Pharmacol ; 240(2): 299-305, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19632255

ABSTRACT

Exposure to persistent organic pollutants, such as polychlorinated biphenyls (PCBs), can lead to chronic inflammation and the development of vascular diseases. Because cell adhesion molecules (CAMs) of the cerebrovascular endothelium regulate infiltration of inflammatory cells into the brain, we have explored the molecular mechanisms by which ortho-substituted polychlorinated biphenyls (PCBs), such as PCB153, can upregulate CAMs in brain endothelial cells. Exposure to PCB153 increased expression of intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1), as well as elevated adhesion of leukocytes to brain endothelial cells. These effects were impeded by inhibitors of EGFR, JAKs, or Src activity. In addition, pharmacological inhibition of NADPH oxidase or disruption of lipid rafts by cholesterol depleting agents blocked PCB153-induced phosphorylation of JAK and Src kinases and upregulation of CAMs. In contrast, silencing of caveolin-1 by siRNA interference did not affect upregulation of ICAM-1 and VCAM-1 in brain endothelial cells stimulated by PCB153. Results of the present study indicate that lipid raft-dependent NADPH oxidase/JAK/EGFR signaling mechanisms regulate the expression of CAMs in brain endothelial cells and adhesion of leukocytes to endothelial monolayers. Due to its role in leukocyte infiltration, induction of CAMs may contribute to PCB-induced cerebrovascular disorders and neurotoxic effects in the CNS.


Subject(s)
Brain/blood supply , Cell Adhesion Molecules/metabolism , Endothelial Cells/drug effects , Environmental Pollutants/toxicity , Membrane Microdomains/drug effects , NADPH Oxidases/metabolism , Polychlorinated Biphenyls/toxicity , Signal Transduction/drug effects , Caveolin 1/genetics , Caveolin 1/metabolism , Cell Adhesion/drug effects , Cell Line, Transformed , Cholesterol/metabolism , Coculture Techniques , Dose-Response Relationship, Drug , Endothelial Cells/enzymology , ErbB Receptors/drug effects , ErbB Receptors/metabolism , Humans , Intercellular Adhesion Molecule-1/metabolism , Janus Kinases/metabolism , Leukocytes/drug effects , Leukocytes/metabolism , Membrane Microdomains/enzymology , Oxidation-Reduction , Phosphorylation , Protein Kinase Inhibitors/pharmacology , RNA Interference , Time Factors , U937 Cells , Up-Regulation , Vascular Cell Adhesion Molecule-1/metabolism , src-Family Kinases/metabolism
17.
FASEB J ; 23(5): 1596-606, 2009 May.
Article in English | MEDLINE | ID: mdl-19141539

ABSTRACT

The blood-brain barrier (BBB) plays an important role in HIV trafficking into the brain and the development of the central nervous system complications in HIV infection. Tight junctions are the main structural and functional elements that regulate the BBB integrity. Exposure of human brain microvascular endothelial cells (hCMEC/D3 cell line) to HIV-infected monocytes resulted in decreased expression of tight junction proteins, such as junctional adhesion molecule-A (JAM)-A, occludin, and zonula occludens (ZO)-1. Control experiments involved exposure to uninfected monocytes. Alterations of tight junction protein expression were associated with increased endothelial permeability and elevated transendothelial migration of HIV-infected monocytes across an in vitro model of the BBB. Notably, overexpression of the peroxisome proliferator-activated receptor (PPAR)alpha or PPARgamma attenuated HIV-mediated dysregulation of tight junction proteins. With the use of exogenous PPARgamma agonists and silencing of PPARalpha or PPARgamma, these protective effects were connected to down-regulation of matrix metalloproteinase (MMP) and proteasome activities. Indeed, the HIV-induced decrease in the expression of JAM-A and occludin was restored by inhibition of MMP activity. Moreover, both MMP and proteasome inhibitors attenuated HIV-mediated altered expression of ZO-1. The present data indicate that down-regulation of MMP and proteasome activities constitutes a novel mechanism of PPAR-induced protections against HIV-induced disruption of brain endothelial cells.


Subject(s)
Blood-Brain Barrier/physiopathology , HIV-1/physiology , PPAR alpha/physiology , PPAR gamma/physiology , Proteasome Endopeptidase Complex/metabolism , Tight Junctions/metabolism , Brain/metabolism , Cell Adhesion Molecules/biosynthesis , Cell Line , Coculture Techniques , Endopeptidases/metabolism , Endothelial Cells/metabolism , Gene Silencing , Humans , Immunoglobulins/biosynthesis , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Membrane Proteins/biosynthesis , Occludin , Phosphoproteins/biosynthesis , Receptors, Cell Surface , Zonula Occludens-1 Protein
18.
J Neurochem ; 107(2): 497-509, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18710415

ABSTRACT

Peroxisome proliferator-activated receptors (PPARs) are nuclear receptors which down-regulate inflammatory signaling pathways. Therefore, we hypothesized that alterations of PPAR functions can contribute to human immunodeficiency virus-1 (HIV-1)-induced dysfunction of brain endothelial cells. Indeed, treatment with HIV-1 transactivator of transcription (Tat) protein decreased PPAR transactivation in brain endothelial cells. We next stably over-expressed PPARalpha and PPARgamma in a newly developed cell line of human brain endothelial cells (hCMEC/D3 cells). Tat-induced up-regulation of inflammatory mediators, such as interleukin (IL)-1beta, tumor necrosis factor-alpha, CCL2, and E-selectin were markedly attenuated in hCMEC/D3 over-expressing PPARalpha or PPARgamma. These results were confirmed in CCL2 and E-selectin promoter activity studies. Similar protective effects were observed in hCMEC/D3 after activation of PPARgamma by exogenous PPAR agonists (dPGJ(2) and rosiglitazone). PPAR over-expression also prevented Tat-induced binding activity and transactivation of nuclear factor-kappaB. Importantly, increased PPAR activity attenuated induction of IL-1beta, tumor necrosis factor-alpha, CCL2, and E-selectin in hCMEC/D3 cells co-cultured with HIV-1-infected Jurkat cells. The protective effects of PPAR over-expression were reversed by the antagonists of PPARalpha (MK886) or PPARgamma (GW9662). The present data suggest that targeting PPAR signaling may provide a novel therapeutic approach to attenuate HIV-1-induced local inflammatory responses in brain endothelial cells.


Subject(s)
Endothelial Cells/metabolism , Endothelial Cells/virology , HIV/physiology , Microvessels/cytology , PPAR alpha/metabolism , PPAR gamma/metabolism , Antineoplastic Agents/pharmacology , Brain/anatomy & histology , Cell Line, Transformed , Cytokines/metabolism , Dose-Response Relationship, Drug , E-Selectin/metabolism , Endothelial Cells/drug effects , Gene Products, tat/pharmacology , Humans , Prostaglandin D2/analogs & derivatives , Prostaglandin D2/pharmacology , Transcriptional Activation/drug effects , Transfection/methods , Up-Regulation/drug effects
19.
Environ Toxicol Pharmacol ; 25(2): 251-9, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18438459

ABSTRACT

Exposure to specific congeners of polychlorinated biphenyls (PCBs) can induce proinflammatory alterations, which may contribute to the formation of blood-borne tumor metastasis. The main aim of the present study was to establish an experimental model of PCB exposure in which PCBs are administered by oral gavage, which resembles the human exposure through the food chain. To determine structure-function relationship, we studied induction of inflammatory responses in the livers, lungs and brains of mice treated with PCB77 (a major coplanar PCB), PCB104 (a non-coplanar PCB with multiple ortho-chlorine substituents), and PCB153 (a major non-coplanar PCB) after a single gavage dose (150 µmol/kg body weight). The strongest expression of proinflammatory proteins occurred 24 h following the PCB administration independent of the class of PCB congeners. These data indicate that food-chain exposure to PCBs can induce proinflammatory mediators in organs that are potential targets for PCB-induced toxicity.

20.
Environ Toxicol Pharmacol ; 25(2): 234-40, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18438464

ABSTRACT

Polychlorinated biphenyl (PCB) congeners exhibit a broad range of adverse biological effects including neurotoxicity. The mechanisms by which PCBs cause neurotoxic effects are still not completely understood. The blood-brain barrier (BBB) is a physical and metabolic barrier separating brain microenvironment from the peripheral circulation and is mainly composed of endothelial cells connected by tight junctions. We examined the effects of several highly-chlorinated PCB congeners on expression of tight junction proteins in human brain endothelial cells. Treatment for 24 h with selective PCB congeners disrupted expression of the cytosolic scaffold proteins of tight junctions, such as zonula occludens (ZO)-1, ZO-2, and AF6. In contrast, PCB exposure did not alter expression of integral membrane proteins, junctional adhesion molecule-A (JAM-A), and claudin-1. Based on these data, we suggest that PCB-mediated selective alterations of tight junction protein expression may contribute to their neurotoxic effects in the central nervous system.

SELECTION OF CITATIONS
SEARCH DETAIL
...