Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters











Database
Type of study
Language
Publication year range
1.
iScience ; 27(4): 109354, 2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38500817

ABSTRACT

Glia are the protectors of the nervous system, providing neurons with support and protection from cytotoxic insults. We previously discovered that four astrocyte-like glia can regulate organismal proteostasis and longevity in C. elegans. Expression of the UPRER transcription factor, XBP-1s, in these glia increases stress resistance, and longevity, and activates the UPRER in intestinal cells via neuropeptides. Autophagy, a key regulator of metabolism and aging, has been described as a cell autonomous process. Surprisingly, we find that glial XBP-1s enhances proteostasis and longevity by cell non-autonomously reprogramming organismal lipid metabolism and activating autophagy. Glial XBP-1s regulates the activation of another transcription factor, HLH-30/TFEB, in the intestine. HLH-30 activates intestinal autophagy, increases intestinal lipid catabolism, and upregulates a robust transcriptional program. Our study reveals a novel role for glia in regulating peripheral lipid metabolism, autophagy, and organellar health through peripheral activation of HLH-30 and autophagy.

2.
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
3.
J Neuroimmune Pharmacol ; 16(1): 74-89, 2021 03.
Article in English | MEDLINE | ID: mdl-31209776

ABSTRACT

HIV infection is associated with comorbidities that are likely to be driven not only by HIV itself, but also by the toxicity of long-term use of antiretroviral therapy (ART). Indeed, increasing evidence demonstrates that the antiretroviral drugs used for HIV treatment have toxic effects resulting in various cellular and tissue pathologies. The blood-brain barrier (BBB) is a modulated anatomophysiological interface which separates and controls substance exchange between the blood and the brain parenchyma; therefore, it is particularly exposed to ART-induced toxicity. Balancing the health risks and gains of ART has to be considered in order to maximize the positive effects of therapy. The current review discusses the cerebrovascular toxicity of ART, with the focus on mitochondrial dysfunction. Graphical Abstract Graphical representation of the interactions between HIV, antiretroviral therapy (ART), and the blood-brain barrier (BBB).


Subject(s)
Anti-HIV Agents/pharmacology , Blood-Brain Barrier/drug effects , AIDS Dementia Complex/drug therapy , AIDS Dementia Complex/etiology , ATP Binding Cassette Transporter, Subfamily B/physiology , Aging , Anti-HIV Agents/adverse effects , Anti-HIV Agents/pharmacokinetics , Anti-HIV Agents/therapeutic use , Antiretroviral Therapy, Highly Active , Apoptosis/drug effects , Autophagy/drug effects , Biological Transport , Blood-Brain Barrier/physiology , DNA Polymerase gamma/physiology , DNA, Mitochondrial/metabolism , Drug Resistance , HIV Infections/drug therapy , HIV Infections/physiopathology , HIV-1/pathogenicity , Humans , Inflammation , Mitochondria/pathology , Models, Biological , Nervous System Diseases/chemically induced , Neural Stem Cells/drug effects , Neural Stem Cells/pathology , Organ Specificity
4.
Mol Pharm ; 16(2): 724-736, 2019 02 04.
Article in English | MEDLINE | ID: mdl-30592424

ABSTRACT

HIV infection is associated with symptoms of accelerated or accentuated aging that are likely to be driven not only by HIV itself but also by the toxicity of long-term use of antiretroviral drugs. Therefore, it is crucially important to understand the mechanisms by which antiretroviral drugs may contribute to aging. The aim of this study was to investigate the hypothesis that antiretroviral drugs cause increased reactive oxygen species (ROS) generation that results in mitochondrial dysfunction and culminates in promoting cellular senescence. In addition, we applied targeted nanoparticle (NP)-based delivery to specifically enrich mitochondria with coenzyme Q10 (CoQ10) in order to enhance antioxidant protection. The studies employed neural progenitor cells (NPCs), as differentiation of these cells into mature neurons is affected both during HIV infection and in the aging process. Exposure of cultured NPCs to various combinations of HIV antiretroviral therapy (ART) induced a more than 2-fold increase in mitochondrial ROS generation and mitochondrial membrane potential, a more than 50% decrease in oxygen consumption and ATP levels, a 60% decrease in SIRT3 expression, and a 42% decrease in cell proliferation relative to control levels. These alterations were accompanied by a 37% increase in beta-galactosidase staining and a shortening of the telomere length to more than half of the length of controls as assessed by quantitative telomere-FISH labeling, indicating accelerated NPC senescence in response to ART exposure. Importantly, CoQ10 delivered by targeted nanoparticles effectively attenuated these effects. Overall, these results indicate that ART promotes cellular senescence by causing mitochondrial dysfunction, which can be successfully reversed by supplementation with mitochondria-targeted CoQ10.


Subject(s)
Anti-Retroviral Agents/pharmacology , Cellular Senescence/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Ubiquinone/analogs & derivatives , Animals , Antioxidants/metabolism , Cell Line , HIV Infections/metabolism , Membrane Potential, Mitochondrial/drug effects , Mice , Nanoparticles/chemistry , Neural Stem Cells/metabolism , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Sirtuin 3/metabolism , Ubiquinone/metabolism
5.
J Biol Chem ; 293(1): 296-311, 2018 01 05.
Article in English | MEDLINE | ID: mdl-29158267

ABSTRACT

HIV-1 infection and methamphetamine (METH) abuse frequently occur simultaneously and may have synergistic pathological effects. Although HIV-positive/active METH users have been shown to have higher HIV viral loads and experience more severe neurological complications than non-users, the direct impact of METH on HIV infection and its link to the development of neurocognitive alternations are still poorly understood. In the present study, we hypothesized that METH impacts HIV infection of neural progenitor cells (NPCs) by a mechanism encompassing NFκB/SP1-mediated HIV LTR activation. Mouse and human NPCs were infected with EcoHIV (modified HIV virus infectious to mice) and HIV, respectively, in the presence or absence of METH (50 or 100 µm). Pretreatment with METH, but not simultaneous exposure, significantly increased HIV production in both mouse and human NPCs. To determine the mechanisms underlying these effects, cells were transfected with different variants of HIV LTR promoters and then exposed to METH. METH treatment induced transcriptional activity of the HIV LTR promotor, an effect that required both NFκB and SP1 signaling. Pretreatment with METH also decreased neuronal differentiation of HIV-infected NPCs in both in vitro and in vivo settings. Importantly, NPC-derived daughter cells appeared to be latently infected with HIV. This study indicates that METH increases HIV infectivity of NPCs, through the NFκB/SP1-dependent activation of the HIV LTR and with the subsequent alterations of NPC neurogenesis. Such events may underlie METH- exacerbated neurocognitive dysfunction in HIV-infected patients.


Subject(s)
HIV Infections/virology , HIV-1/drug effects , Methamphetamine/pharmacology , Animals , Cell Line , HIV Long Terminal Repeat/drug effects , Humans , Male , Methamphetamine/adverse effects , Mice , Mice, Inbred C57BL , NF-kappa B/drug effects , NF-kappa B/metabolism , Neural Stem Cells/drug effects , Neurons/drug effects , Signal Transduction/drug effects , Sp1 Transcription Factor/drug effects , Stem Cells/drug effects , Viral Load
6.
J Cereb Blood Flow Metab ; 38(2): 317-332, 2018 02.
Article in English | MEDLINE | ID: mdl-28718701

ABSTRACT

Energetic regulation at the blood-brain barrier is critical for maintaining its integrity, transport capabilities, and brain demands for glucose. However, the underlying mechanisms that regulate these processes are still poorly explored. We recently characterized the protein occludin as a NADH oxidase and demonstrated its influence on the expression and activation of the histone deacetylase SIRT-1. Because SIRT-1 works in concert with AMP-activated protein kinase (AMPK) (AMPK), we investigated the impact of occludin on this metabolic switch. Here we show that in blood-brain barrier pericytes, occludin promotes AMPK expression and activation, influencing the expression of glucose transporters GLUT-1 and GLUT-4, glucose uptake, and ATP content. Furthermore, occludin expression, AMP-dependent protein kinase activity, and glucose uptake were altered under inflammatory (TNFα) and infectious (HIV) conditions. We also show that pericytes share glucose and mitochondria with astrocytes, and that occludin levels modify the ability of pericytes to share those energetic resources. In addition, we demonstrate that murine mitochondria can be transferred from live brain microvessels to energetically impaired human astrocytes, promoting their survival. Our findings demonstrate that occludin plays an important role in blood-brain barrier pericyte metabolism by influencing AMPK protein kinase activity, glucose uptake, ATP production, and by regulating the ability of pericytes to interact metabolically with astrocytes.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Adenosine Triphosphate/biosynthesis , Glucose/metabolism , Occludin/physiology , Pericytes/metabolism , Astrocytes/metabolism , Astrocytes/ultrastructure , Blood-Brain Barrier/cytology , Blood-Brain Barrier/metabolism , Enzyme Activation , Glucose Transporter Type 1/biosynthesis , Glucose Transporter Type 1/genetics , Glucose Transporter Type 4/biosynthesis , Glucose Transporter Type 4/genetics , HIV Infections/metabolism , Humans , Metabolic Networks and Pathways , Mitochondria/metabolism , Occludin/genetics , Pericytes/ultrastructure , Primary Cell Culture , Tumor Necrosis Factor-alpha
SELECTION OF CITATIONS
SEARCH DETAIL