Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Mol Neurobiol ; 60(11): 6441-6465, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37458985

ABSTRACT

Brain endothelial extracellular vesicles carrying amyloid beta (EV-Aß) can be transferred to neural progenitor cells (NPCs) leading to NPC dysfunction. However, the events involved in this EV-mediated Aß pathology are unclear. EV-proteomics studies identified Serpine-1 (plasminogen activator inhibitor 1, PAI-1) as a major connecting "hub" on several protein-protein interaction maps. Serpine-1 was described as a key player in Aß pathology and was linked to HIV-1 infection as well. Therefore, the aim of this work was to address the hypothesis that Serpine-1 can be transferred via EVs from brain endothelial cells (HBMEC) to NPCs and contribute to NPC dysfunction. HBMEC concentrated and released Serpine-1 via EVs, the effect that was potentiated by HIV-1 and Aß. EVs loaded with Serpine-1 were readily taken up by NPCs, and HIV-1 enhanced this event. Interestingly, a highly specific Serpine-1 inhibitor PAI039 increased EV-Aß transfer to NPCs in the presence of HIV-1. PAI039 also partially blocked mitochondrial network morphology alterations in the recipient NPCs, which developed mainly after HIV + Aß-EV transfer. PAI039 partly attenuated HIV-EV-mediated decreased synaptic protein levels in NPCs, while increased synaptic protein levels in NPC projections. These findings contribute to a better understanding of the complex mechanisms underlying EV-Serpine-1 related Aß pathology in the context of HIV infection. They are relevant to HIV-1 associated neurocognitive disorders (HAND) in an effort to elucidate the mechanisms of neuropathology in HIV infection.


Subject(s)
Extracellular Vesicles , HIV Infections , HIV-1 , Neural Stem Cells , Humans , Amyloid beta-Peptides/metabolism , HIV Infections/metabolism , HIV-1/metabolism , Endothelial Cells/metabolism , Neural Stem Cells/metabolism , Extracellular Vesicles/metabolism
2.
Res Sq ; 2023 Feb 15.
Article in English | MEDLINE | ID: mdl-36824983

ABSTRACT

Brain endothelial extracellular vesicles carrying amyloid beta (EV-Aß) can be transferred to neural progenitor cells (NPCs) leading to NPC dysfunction. However, the events involved in this EV-mediated Aß pathology are unclear. EV-proteomics studies identified Serpine-1 (plasminogen activator inhibitor 1, PAI-1) as a major connecting "hub" on several protein-protein interaction maps. Serpine-1 was described as a key player in Aß pathology and was linked to HIV-1 infection as well. Therefore, the aim of this work was to address the hypothesis that Serpine-1 can be transferred via EVs from brain endothelial cells to NPCs and contribute to NPC dysfunction. HBMEC concentrated and released Serpine-1 via EVs, the effect that was potentiated by HIV-1 and Aß. EVs loaded with Serpine-1 were readily taken up by NPCs, and HIV-1 enhanced this event. Interestingly, a highly specific Serpine-1 inhibitor PAI039 increased EV-Aß transfer to NPCs in the presence of HIV-1. PAI039 also partially blocked mitochondrial network morphology and mitochondrial function alterations in the recipient NPCs, which developed mainly after HIV + Aß-EV transfer. PAI039 partly attenuated HIV-EV-mediated decreased synaptic protein levels in NPCs, while increased synaptic protein levels in NPC projections. These findings contribute to a better understanding of the complex mechanisms underlying EV-Serpine-1 related Aß pathology in the context of HIV infection. They are relevant to HIV-1 associated neurocognitive disorders (HAND) in an effort to elucidate the mechanisms of neuropathology in HIV infection.

3.
Article in English | MEDLINE | ID: mdl-36649440

ABSTRACT

Aim: Elevated brain deposits of amyloid beta (Aß40) contribute to neuropathology and cognitive dysfunction in Alzheimer's disease (AD). However, the role of the blood-brain barrier (BBB) as an interface for the transfer of Aß40 from the periphery into the brain is not well characterized. In addition, a substantial population of neural progenitor cells (NPCs) resides in close proximity to brain capillaries that form the BBB. The aim of this study is to understand the impact of brain endothelium-derived extracellular vesicles (EV) containing Aß40 on metabolic functions and differentiation of NPCs. Methods: Endothelial EVs were derived from an in vitro model of the brain endothelium treated with 100 nM Aß40 or PBS. We then analyzed the impact of these EVs on mitochondrial morphology and bioenergetic disruption of NPCs. In addition, NPCs were differentiated and neurite development upon exposure to EVs was assessed using the IncuCyte Zoom live cell imaging system. Results: We demonstrate that physiological concentrations of Aß40 can be transferred to accumulate in NPCs via endothelial EVs. This transfer results in mitochondrial dysfunction, disrupting crista morphology, metabolic rates, fusion and fission dynamics of NPCs, as well as their neurite development. Conclusion: Intercellular transfer of Aß40 is carried out by brain endothelium-derived EVs, which can affect NPC differentiation and induce mitochondrial dysfunction, leading to aberrant neurogenesis. This has pathological implications because NPCs growing into neurons are incorporated into cerebral structures involved in learning and memory, two common phenotypes affected in AD and related dementias.

4.
Neurobiol Dis ; 155: 105388, 2021 07.
Article in English | MEDLINE | ID: mdl-33962010

ABSTRACT

Human immunodeficiency virus-1 (HIV-1) has been shown to cross the blood-brain barrier and cause HIV-associated neurocognitive disorders (HAND) through a process that may involve direct or indirect interactions with the central nervous system (CNS) cells and alterations of amyloid ß (Aß) homeostasis. The present study focused on the mechanisms of HIV-1 infecting human neural progenitor cells (hNPCs) and affecting NPC intercellular communications with human brain endothelial cells (HBMEC). Despite the lack of the CD4 receptor, hNPCs were effectively infected by HIV-1 via a mechanism involving the chemokine receptors, CXCR4 and CCR5. HIV-1 infection increased expression of connexin-43 (Cx43), phosphorylated Cx43 (pCx43), and pannexin 2 (Panx2) protein levels in hNPCs, suggesting alterations in gap-junction (GJ) and pannexin channel communication. Indeed, a functional GJ assay indicated an increase in communication between HIV-infected hNPCs and non-infected HBMEC. We next analyzed the impact of HBMEC-derived extracellular vesicles (EVs) and EVs carrying Aß (EV-Aß) on the expression of Cx43, pCx43, and Panx2 in HIV-1 infected and non-infected hNPCs. Exposure to EV-Aß resulted in significant reduction of Cx43 and pCx43 protein expression in non-infected hNPCs when compared to EV controls. Interestingly, EV-Aß treatment significantly increased levels of Cx43, pCx43, and Panx2 in HIV-1-infected hNPCs when compared to non-infected controls. These results were confirmed in a GJ functional assay and an ATP release assay, which is an indicator of connexin hemichannel and/or pannexin channel functions. Overall, the current study demonstrates the importance of hNPCs in HIV-1 infection and indicates that intercellular communications between infected hNPCs and HBMEC can be effectively modulated by EVs carrying Aß as their cargo.


Subject(s)
Cell Communication/physiology , Extracellular Vesicles/metabolism , Gap Junctions/metabolism , HIV Infections/metabolism , HIV-1/metabolism , Neural Stem Cells/metabolism , Amyloid beta-Peptides/metabolism , Cell Line , Cells, Cultured , Endothelium, Vascular/metabolism , Endothelium, Vascular/virology , Extracellular Vesicles/virology , Gap Junctions/virology , Humans , Neural Stem Cells/virology
5.
Int J Mol Sci ; 21(8)2020 Apr 15.
Article in English | MEDLINE | ID: mdl-32326569

ABSTRACT

Amyloid beta (Aß) depositions are more abundant in HIV-infected brains. The blood-brain barrier, with its backbone created by endothelial cells, is assumed to be a core player in Aß homeostasis and may contribute to Aß accumulation in the brain. Exposure to HIV increases shedding of extracellular vesicles (EVs) from human brain endothelial cells and alters EV-Aß levels. EVs carrying various cargo molecules, including a complex set of proteins, can profoundly affect the biology of surrounding neurovascular unit cells. In the current study, we sought to examine how exposure to HIV, alone or together with Aß, affects the surface and total proteomic landscape of brain endothelial EVs. By using this unbiased approach, we gained an unprecedented, high-resolution insight into these changes. Our data suggest that HIV and Aß profoundly remodel the proteome of brain endothelial EVs, altering the pathway networks and functional interactions among proteins. These events may contribute to the EV-mediated amyloid pathology in the HIV-infected brain and may be relevant to HIV-1-associated neurocognitive disorders.


Subject(s)
Amyloid beta-Peptides/metabolism , Brain/metabolism , Endothelial Cells/metabolism , Extracellular Vesicles/metabolism , HIV Infections/metabolism , HIV-1/metabolism , Proteome/metabolism , Amyloid beta-Peptides/pharmacology , Blood-Brain Barrier/metabolism , Brain/virology , Cells, Cultured , Chromatography, Liquid , Extracellular Vesicles/virology , Gene Ontology , HEK293 Cells , Humans , Protein Interaction Maps , Proteomics , Software , Tandem Mass Spectrometry
6.
Mol Brain ; 13(1): 21, 2020 02 17.
Article in English | MEDLINE | ID: mdl-32066471

ABSTRACT

Amyloid beta (Aß) deposition was demonstrated to be elevated in the brains of HIV-infected patients and associated with neurocognitive decline; however, the mechanisms of these processes are poorly understood. The goal of the current study was to address the hypothesis that Aß can be transferred via extracellular vesicles (ECVs) from brain endothelial cells to neural progenitor cells (NPCs) and that this process can contribute to abnormal NPC differentiation. Mechanistically, we focused on the role of the receptor for advanced glycation end products (RAGE) and activation of the inflammasome in these events. ECVs loaded with Aß (Aß-ECVs) were readily taken up by NPCs and Aß partly colocalized with the inflammasome markers ASC and NLRP3 in the nuclei of the recipient NPCs. This colocalization was affected by HIV and RAGE inhibition by a high-affinity specific inhibitor FPS-ZM1. Blocking RAGE resulted also in an increase in ECV number produced by brain endothelial cells, decreased Aß content in ECVs, and diminished Aß-ECVs transfer to NPC nuclei. Interestingly, both Aß-ECVs and RAGE inhibition altered NPC differentiation. Overall, these data indicate that RAGE inhibition affects brain endothelial ECV release and Aß-ECVs transfer to NPCs. These events may modulate ECV-mediated amyloid pathology in the HIV-infected brain and contribute to the development of HIV-associated neurocognitive disorders.


Subject(s)
Amyloid beta-Peptides/metabolism , Extracellular Vesicles/metabolism , HIV Infections/metabolism , Neural Stem Cells/metabolism , Receptor for Advanced Glycation End Products/metabolism , Brain/pathology , Cell Differentiation , Chemokines/metabolism , Humans , Inflammasomes/metabolism , Inflammation Mediators/metabolism , Models, Biological , Neurons/metabolism , Solubility
7.
Mol Cell Neurosci ; 79: 12-22, 2017 03.
Article in English | MEDLINE | ID: mdl-28040512

ABSTRACT

HIV-infected brains are characterized by increased amyloid beta (Aß) deposition. It is believed that the blood-brain barrier (BBB) is critical for Aß homeostasis and contributes to Aß accumulation in the brain. Extracellular vesicles (ECV), like exosomes, recently gained a lot of attention as potentially playing a significant role in Aß pathology. In addition, HIV-1 hijacks the exosomal pathway for budding and release. Therefore, we investigated the involvement of BBB-derived ECV in the HIV-1-induced Aß pathology in the brain. Our results indicate that HIV-1 increases ECV release from brain endothelial cells as well as elevates their Aß cargo when compared to controls. Interestingly, brain endothelial cell-derived ECV transferred Aß to astrocytes and pericytes. Infusion of brain endothelial ECV carrying fluorescent Aß into the internal carotid artery of mice resulted in Aß fluorescence associated with brain microvessels and in the brain parenchyma. These results suggest that ECV carrying Aß can be successfully transferred across the BBB into the brain. Based on these observations, we conclude that HIV-1 facilitates the shedding of brain endothelial ECV carrying Aß; a process that may increase Aß exposure of cells of neurovascular unit, and contribute to amyloid deposition in HIV-infected brain.


Subject(s)
AIDS Dementia Complex/metabolism , Amyloid beta-Peptides/metabolism , Blood-Brain Barrier/metabolism , Extracellular Vesicles/metabolism , Amyloid beta-Peptides/toxicity , Animals , Blood-Brain Barrier/virology , Cell Line , Cells, Cultured , Endothelium, Vascular/metabolism , Endothelium, Vascular/virology , HEK293 Cells , HIV-1/pathogenicity , Humans , Male , Mice , Mice, Inbred C57BL
8.
Tissue Barriers ; 4(1): e1131804, 2016.
Article in English | MEDLINE | ID: mdl-27141419

ABSTRACT

Extracellular vesicles (ECV), like exosomes, gained recently a lot of attention as potentially playing a significant role in neurodegenerative diseases, particularly in Aß pathology. While there are a lot of reports on ECV/exosomes derived from a variety of cell types, there is limited information on ECV/exosomes originated from brain microvascular endothelial cells forming the blood-brain barrier (BBB). In this review, we summarize the literature data on brain endothelial ECV/exosomes and present our own data on BBB-derived ECV and their possible involvement in the brain's Aß pathology. We propose that ECV/exosome release from brain endothelial cells associated with Aß affects different cells of the neurovascular unit and may be an important contributor to the Aß deposition in the central nervous system.


Subject(s)
Blood-Brain Barrier/metabolism , Extracellular Vesicles/metabolism , Amyloid beta-Peptides/metabolism , Animals , Blood-Brain Barrier/cytology , Blood-Brain Barrier/drug effects , Drug Carriers/pharmacokinetics , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Humans
9.
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
10.
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
11.
PLoS One ; 9(5): e97033, 2014.
Article in English | MEDLINE | ID: mdl-24804765

ABSTRACT

Tumor cell extravasation into the brain requires passage through the blood-brain barrier (BBB). There is evidence that exercise can alter the oxidation status of the brain microvasculature and protect against tumor cell invasion into the brain, although the mechanisms are not well understood. In the current study, we focused on the role of microenvironment generated by exercise and metastasizing tumor cells at the levels of brain microvessels, influencing oxidative stress-mediated responses and activation of redox-sensitive small GTPases. Mature male mice were exercised for four weeks using a running wheel with the average voluntary running distance 9.0 ± 0.3 km/day. Mice were then infused with 1.0 × 10(6) D122 (murine Lewis lung carcinoma) cells into the brain microvasculature, and euthanized either 48 hours (in short-term studies) or 2-3 weeks (in long-term studies) post tumor cell administration. A significant increase in the level of reactive oxygen species was observed following 48 hours or 3 weeks of tumor cells growth, which was accompanied by a reduction in MnSOD expression in the exercised mice. Activation of the small GTPase Rho was negatively correlated with running distance in the tumor cell infused mice. Together, these data suggest that exercise may play a significant role during aggressive metastatic invasion, especially at higher intensities in pre-trained individuals.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/therapy , Carcinoma, Lewis Lung/therapy , Monomeric GTP-Binding Proteins/biosynthesis , Physical Conditioning, Animal , Animals , Blood-Brain Barrier/metabolism , Brain Neoplasms/pathology , Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/pathology , Disease Models, Animal , Exercise Therapy , Humans , Mice , Monomeric GTP-Binding Proteins/genetics , Oxidation-Reduction , Oxidative Stress/genetics , Reactive Oxygen Species/metabolism
12.
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
13.
Exp Cell Res ; 323(1): 66-76, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24491918

ABSTRACT

Clinical evidence indicates increased amyloid deposition in HIV-1-infected brains, which contributes to neurocognitive dysfunction in infected patients. Here we show that HIV-1 exposure stimulates amyloid beta (Aß) nuclear entry in human brain endothelial cells (HBMEC), the main component of the blood-brain barrier (BBB). Treatment with HIV-1 and/or Aß resulted in concurrent increase in early endosomal antigen-1 (EEA1), Smad, and phosphorylated Smad (pSmad) in nuclear fraction of HBMEC. A series of inhibition and silencing studies indicated that Smad and EEA1 closely interact by influencing their own nuclear entry; the effect that was attenuated by dynasore, a blocker of GTP-ase activity of dynamin. Importantly, inhibition of dynamin, EEA1, or TGF-ß/Smad effectively attenuated HIV-1-induced Aß accumulation in the nuclei of HBMEC. The present study indicates that nuclear uptake of Aß involves the dynamin-dependent EEA1 and TGF-ß/Smad signaling pathways. These results identify potential novel targets to protect against HIV-1-associated dysregulation of amyloid processes at the BBB level.


Subject(s)
Amyloid beta-Peptides/metabolism , Dynamins/metabolism , HIV Infections/metabolism , HIV-1 , Smad2 Protein/metabolism , Transforming Growth Factor beta/metabolism , Vesicular Transport Proteins/metabolism , Active Transport, Cell Nucleus/physiology , Benzamides/pharmacology , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/virology , Brain/metabolism , Brain/virology , Cell Line , Cognitive Dysfunction/virology , Dioxoles/pharmacology , Dynamins/antagonists & inhibitors , Endothelial Cells/metabolism , Humans , Hydrazones/pharmacology , Phosphorylation , RNA Interference , RNA, Small Interfering , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Signal Transduction/drug effects , Smad2 Protein/antagonists & inhibitors , Transforming Growth Factor beta/antagonists & inhibitors , Vesicular Transport Proteins/antagonists & inhibitors , Vesicular Transport Proteins/genetics
14.
Mol Neurodegener ; 8: 22, 2013 Jun 24.
Article in English | MEDLINE | ID: mdl-23799892

ABSTRACT

BACKGROUND: There is no effective therapeutic intervention developed targeting cerebrovascular toxicity of drugs of abuse, including methamphetamine (METH). We hypothesize that exercise protects against METH-induced disruption of the blood-brain barrier (BBB) by enhancing the antioxidant capacity of cerebral microvessels and modulating caveolae-associated signaling. Mice were subjected to voluntary wheel running for 5 weeks resembling the voluntary pattern of human exercise, followed by injection with METH (10 mg/kg). The frequency, duration, and intensity of each running session were monitored for each mouse via a direct data link to a computer and the running data are analyzed by Clock lab™ Analysis software. Controls included mice sedentary that did not have access to running wheels and/or injections with saline. RESULTS: METH induced oxidative stress in brain microvessels, resulting in up regulation of caveolae-associated NAD(P)H oxidase subunits, and phosphorylation of mitochondrial protein 66Shc. Treatment with METH disrupted also the expression and colocalization of tight junction proteins. Importantly, exercise markedly attenuated these effects and protected against METH-induced disruption of the BBB integrity. CONCLUSIONS: The obtained results indicate that exercise is an important modifiable behavioral factor that can protect against METH-induced cerebrovascular toxicity. These findings may provide new strategies in preventing the toxicity of drug of abuse.


Subject(s)
Blood-Brain Barrier/drug effects , Brain/drug effects , Central Nervous System Stimulants/toxicity , Methamphetamine/toxicity , Oxidative Stress/drug effects , Physical Conditioning, Animal/physiology , Animals , Blood-Brain Barrier/physiology , Brain/blood supply , Brain/physiology , Mice , Microvessels/drug effects , Oxidative Stress/physiology
15.
IUBMB Life ; 65(1): 43-9, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23225609

ABSTRACT

In recent years, we face an increase in the aging of the HIV-1-infected population, which is not only due to effective antiretroviral therapy but also to new infections among older people. Even with the use of the antiretroviral therapy, HIV-associated neurocognitive disorders represent an increasing problem as the HIV-1-infected population ages. Increased amyloid beta (Aß) deposition is characteristic of HIV-1-infected brains, and it has been hypothesized that brain vascular dysfunction contributes to this phenomenon, with a critical role suggested for the blood-brain barrier in brain Aß homeostasis. This review will describe the mechanisms by which the blood-brain barrier may contribute to brain Aß accumulation, and our findings in the context of HIV-1 infection will be discussed.


Subject(s)
Amyloid beta-Peptides/metabolism , Blood-Brain Barrier , Brain/metabolism , HIV Infections/metabolism , HIV-1 , Humans , Membrane Microdomains/metabolism , Proteoglycans/metabolism
16.
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
17.
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
18.
FASEB J ; 25(11): 3979-88, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21840940

ABSTRACT

Activation of matrix metalloproteinase-9 (MMP-9) is involved in HIV-1-induced disruption of the blood-brain barrier (BBB). In the present study, we hypothesize that peroxisome proliferator-activated receptor (PPAR)-α or PPARγ can protect against HIV-1-induced MMP-9 overexpression in brain endothelial cells (hCMEC cell line) by attenuating cellular oxidative stress and down-regulation of caveolae-associated redox signaling. Exposure to HIV-1-infected monocytes induced phosphorylation of ERK1/2 and Akt in hCMEC by 2.5- and 3.6-fold, respectively; however, these effects were attenuated by overexpression of PPARα or PPARγ and by silencing of caveolin-1 (cav-1). Coculture of hCMEC with HIV-1-infected monocytes significantly induced MMP-9 promoter and enzyme activity by 3- to 3.5-fold. Promoter mutation studies indicated that SP-1 (g1940t_g1941t) is an essential transcription factor involved in induction of MMP-9 promoter by HIV-1. In addition, HIV-1-stimulated activity of MMP-9 promoter was inhibited by mutation of AP-1 site 2 (c1918t_a1919g) and both (but not individual) NF-κB binding sites (g1389c and g1664c). PPAR overexpression, ERK1/2 or Akt inhibition, and silencing of cav-1 all effectively protected against HIV-1-induced MMP-9 promoter activity, indicating a close relationship among HIV-1-induced cerebrovascular toxicity, redox-regulated mechanisms, and functional caveolae. Such a link was further confirmed in MMP-9-deficient mice exposed to PPARα or PPARγ agonist and injected with the HIV-1-specific protein Tat into cerebral vasculature. Overall, our results indicate that ERK1/2, Akt, and cav-1 are involved in the regulatory mechanisms of PPAR-mediated protection against HIV-1-induced MMP-9 expression in brain endothelial cells.


Subject(s)
Caveolin 1/physiology , Extracellular Signal-Regulated MAP Kinases/physiology , HIV-1/metabolism , Matrix Metalloproteinase 9/biosynthesis , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinase 3/physiology , PPAR alpha/physiology , PPAR gamma/physiology , Proto-Oncogene Proteins c-akt/physiology , Animals , Blood-Brain Barrier/metabolism , Caveolae/metabolism , HEK293 Cells , Humans , Mice , Phosphorylation/drug effects , Signal Transduction/drug effects , U937 Cells
19.
Methods Mol Biol ; 762: 355-70, 2011.
Article in English | MEDLINE | ID: mdl-21717370

ABSTRACT

HIV-1 crosses the blood-brain barrier (BBB) early in the course of systemic infection and resides in brain macrophages and microglia. The integrity of the brain endothelium is regulated by intercellular tight junctions, which also play a critical role in HIV-1-entry into the brain. Disruption of tight junctions, including changes in claudin-5 expression, is common in HIV-1-infected patients. Recent evidence indicates that both exposure to HIV-1 and HIV-1 specific proteins, such as Tat protein, can contribute to alterations of expression and distribution of claudin-5 in brain endothelial cells and brain microvessels.


Subject(s)
Blood-Brain Barrier/metabolism , Gene Expression Regulation/physiology , HIV-1/physiology , Membrane Proteins/metabolism , Tight Junctions/metabolism , Blood-Brain Barrier/virology , Blotting, Western/methods , Cell Line, Tumor , Claudin-5 , Electrophoresis, Polyacrylamide Gel/methods , Humans , Microscopy, Fluorescence/methods , Tight Junctions/virology
20.
Am J Physiol Heart Circ Physiol ; 298(4): H1136-45, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20139322

ABSTRACT

Telomerase, via its catalytic component telomerase reverse transcriptase (TERT), extends telomeres of eukaryotic chromosomes. The importance of this reaction is related to the fact that telomere shortening is a rate-limiting mechanism for human life span that induces cell senescence and contributes to the development of age-related pathologies. The aim of the present study was to evaluate whether the modulation of telomerase activity can influence human immunodeficiency virus type 1 (HIV-1)-mediated dysfunction of human brain endothelial cells (hCMEC/D3 cells) and transendothelial migration of HIV-1-infected cells. Telomerase activity was modulated in hCMEC/D3 cells via small interfering RNA-targeting human TERT (hTERT) or by using a specific pharmacological inhibitor of telomerase, TAG-6. The inhibition of hTERT resulted in the upregulation of HIV-1-induced overexpression of intercellular adhesion molecule-1 via the nuclear factor-kappaB-regulated mechanism and induced the transendothelial migration of HIV-1-infected monocytic U937 cells. In addition, the blocking of hTERT activity potentiated a HIV-induced downregulation of the expression of tight junction proteins. These results were confirmed in TERT-deficient mice injected with HIV-1-specific protein Tat into the cerebral vasculature. Further studies revealed that the upregulation of matrix metalloproteinase-9 is the underlying mechanisms of disruption of tight junction proteins in hCMEC/D3 cells with inhibited TERT and exposed to HIV-1. These results indicate that the senescence of brain endothelial cells may predispose to the HIV-induced upregulation of inflammatory mediators and the disruption of the barrier function at the level of the brain endothelium.


Subject(s)
Cell Movement/physiology , Endothelium, Vascular/virology , HIV-1/isolation & purification , Membrane Proteins/metabolism , Telomerase/antagonists & inhibitors , Tight Junctions/metabolism , Animals , Cell Line , Cells, Cultured , Claudin-5 , Disease Models, Animal , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Female , Gene Silencing , HIV Infections/metabolism , HIV Infections/pathology , Humans , Intercellular Adhesion Molecule-1/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Telomerase/genetics , Telomerase/metabolism , U937 Cells , tat Gene Products, Human Immunodeficiency Virus/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...