Previous Article in Journal
Anti-Inflammatory Role of the Klotho Protein and Relevance to Aging
Previous Article in Special Issue
HIV Infection, Antiretroviral Drugs, and the Vascular Endothelium
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Structural and Functional Dysregulation of the Brain Endothelium in HIV Infection and Substance Abuse

by
Narendran Annadurai
and
Georgette D. Kanmogne
*
Department of Anesthesiology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-4455, USA
*
Author to whom correspondence should be addressed.
Cells 2024, 13(17), 1415; https://doi.org/10.3390/cells13171415 (registering DOI)
Submission received: 19 June 2024 / Revised: 12 August 2024 / Accepted: 22 August 2024 / Published: 24 August 2024

Abstract

:
Blood–brain barrier (BBB) injury and dysfunction following infection with the human immunodeficiency virus (HIV) enables viral entry into the brain, infection of resident brain cells, neuronal injury and subsequent neurodegeneration leading to HIV-associated neurocognitive disorders (HAND). Although combination antiretroviral therapy has significantly reduced the incidence and prevalence of acquired immunodeficiency syndrome and increased the life expectancy of people living with HIV, the prevalence of HAND remains high. With aging of people living with HIV associated with increased comorbidities, the prevalence of HIV-related central nervous system (CNS) complications is expected to remain high. Considering the principal role of the brain endothelium in HIV infection of the CNS and HAND, the purpose of this manuscript is to review the current literature on the pathobiology of the brain endothelium structural and functional dysregulation in HIV infection, including in the presence of HIV-1 and viral proteins (gp120, Tat, Nef, and Vpr). We summarize evidence from human and animal studies, in vitro studies, and associated mechanisms. We further summarize evidence of synergy or lack thereof between commonly abused substances (cocaine, methamphetamine, alcohol, tobacco, opioids, and cannabinoids) and HIV- or viral protein-induced BBB injury and dysfunction.

1. Introduction

The blood–brain barrier (BBB) is a semi-permeable membrane that separates circulating blood from brain tissues, regulates the transport of oxygen and essential nutrients into the central nervous system (CNS), protects the brain from harmful substances, and maintains brain homeostasis [1,2,3,4]. It is primarily composed of endothelial cells (ECs) lining the brain blood vessels, a basement membrane that encases the ECs on the abluminal side, pericytes, and astrocyte endfeet [2,3,4]. The molecular components of the BBB responsible for its protective and barrier functions include endothelial tight junction (TJ) proteins (occludin, claudins, and zonula occludens (ZO)), adherent junction (AJ) proteins, and efflux transporters [2,3]. Endothelial injury and subsequent BBB dysfunction are implicated in the pathogenesis of several CNS infections and diseases, including human immunodeficiency virus (HIV) CNS infection and neuroHIV.
In the early stages of HIV infection, endothelial injury following exposure to circulating infected leukocytes, virions, and viral proteins, as well as subsequent BBB dysfunction, enables HIV entry into the brain and infection of resident CNS cells, including productive microglial infection [5,6]. Virions and viral proteins released by infected brain cells are associated with increased inflammation and oxidative stress, which leads to neuronal injury and subsequent neurodegeneration [7,8] (Figure 1). Increased neurodegeneration over time results in motor, cognitive, and behavioral abnormalities termed HIV-associated neurocognitive disorders (HAND) [9,10]. HAND subclassification includes HIV-associated asymptomatic neurocognitive impairment, mild neurocognitive disorder, and HIV-associated dementia [10,11,12,13]. Although the incidence and prevalence of the most severe form of HAND has decreased with the advent of combination antiretroviral therapy (ART), the overall prevalence of HAND and its associated morbidity remains high (up to 40%) [14,15]. With increased life expectancy and aging of people living with HIV (PLWH) [16,17], the prevalence of HAND and HIV-associated CNS comorbidities is expected to remain high. Considering the principal role of the brain endothelium in HIV infection of the CNS and HAND, the purpose of this manuscript is to review the current literature on BBB structure and function in HIV infection and substance abuse. We summarize evidence from human and animal studies, in vitro studies, and associated mechanisms. We also summarize evidence of synergy, or lack thereof, between commonly abused substances (cocaine, methamphetamine, alcohol, tobacco, opioids, and cannabinoids) and HIV- or viral protein-induced BBB injury and dysfunction.

2. HIV Infection, BBB Injury, and Dysfunction

2.1. Evidence from Human Studies

Studies have shown that HIV-infected patients have BBB injury that is associated with increased permeability, infiltration of circulating leukocytes into the CNS, and advanced disease. In these studies, patients with HAND had significantly more damage to the brain endothelium, and increased BBB permeability was observed in 50% of AIDS patients compared to 22% in asymptomatic PLWH [18,19,20]. There is other evidence of increased BBB breakdown in HIV-infected humans. Contrast-enhanced magnetic resonance imaging of PLWH showed altered myoinositol/creatine (mI/Cr) in the basal ganglia associated with high plasma viral load [21]. High cerebrospinal fluid (CSF) albumin and CSF/serum albumin quotient (QAlb) in PLWH correlates with increased neuronal injury (higher CSF neurofilament-L) and impaired brain bioenergetics (lower N-acetylaspartate-to-creatine ratio) in the parietal gray matter [22].
Evidence suggests that ART does not prevent or abrogate HIV-induced BBB injury and can increase BBB permeability. In PLWH who had an abnormal QAlb and N-acetylaspartate-to-creatine ratio, one year of ART did not improve QA1b [22]. Impaired BBB integrity has been reported in both HIV-infected subjects with HAND and neuroasymptomatic PLWH, despite a decreased QA1b post-ART [17]. Although ART suppressed CSF and plasma viral loads, it did not significantly change albumin and neurofilament-L levels [19]. A study of virally suppressed patients with HAND showed BBB disruptions in the basal ganglia and frontal white matter, despite a lack of peripheral inflammation [23]. HIV-infected adults on suppressive ART also showed impaired BBB associated with increased levels of amyloid-β (1–42), and phosphorylated and total tau correlating with CSF albumin and QAlb [24].
Autopsy evidence of brain endothelium injury and impaired BBB in HIV-infected patients has included enlarged microvessels and loss of hippocampal capillaries associated with degeneration of perivascular tissues [25], fragmentation and loss of microvessels TJ proteins, including claudin-5, occludin, ZO-1 and ZO-2 in several brain regions (frontal cortex, basal ganglia, subcortical white matter, and cortical gray matter) [26,27,28]. Brain tissues of HIV-infected humans has also showed significantly more capillary loss and decreased TJs in patients with HAND compared to HIV-infected subjects without HAND and seronegative controls subjects [20,23,26,27,28,29].

2.2. Evidence from Animal Studies

Primate models: Simian immunodeficiency virus (SIV)-infected macaques, including animals with SIV encephalitis, showed endothelial activation and BBB injury, reduced expression of endothelial TJ proteins occludin and ZO-1, and fragmentation of TJs in the basal ganglia microvessels associated with increased pro-inflammatory cytokines and accumulation of perivascular macrophages in the CNS [30,31,32,33]. SIV-infected macaques showed increased expression of vascular cell adhesion molecule (VCAM)-1 that correlated with endothelial activation, BBB injury, and increased production of cytokines such as tumor necrosis factor (TNF)-α and interferon-γ [32,34]. SIV-infected animals also showed increased production of chemokines such as the chemokine ligand-5 (CCL5), macrophage inflammatory protein-1α and -1β, monocyte chemotactic protein-3, and interferon-inducible protein-10 [35]. These chemokines likely contribute to increased diapedesis and recruitment of circulating leukocytes into the brain.
Murine models: Evidence from several HIV/AIDS murine models show that mice and rats infected with HIV-1 [36] or exposed to HIV-1 glycoprotein (gp)120 [37,38,39,40], transactivator of transcription (Tat) [41,42], or negative factor (Nef) [43,44] had reduced expression of cerebral TJ proteins (e.g., occludin, claudin-5, ZO-1 and ZO-2). These animals also had increased BBB disruption and permeability associated with upregulation of endothelial adhesion molecules, matrix metalloproteinases (MMPs), and reactive oxygen species (ROS) [37,38,39,40,41,42,43]. In HIV-infected humanized mice, treatments that prevented or reduced TJ downregulation and BBB dysfunction were also associated with less gliosis, reduced inflammation and leukocyte infiltration into the CNS, and less neuronal injury [36,45]. Induction of Tat expression in transgenic (Tg) mice resulted in BBB breakdown associated with increased leakage of circulating dyes into the brain, activation of perivascular macrophages, and microgliosis [42]. HIV-1 Tg26 mice and gp120 Tg mice showed reduced ZO-1 expression and increased BBB permeability, including enhanced leakage of albumin into the brain associated with increased intercellular adhesion molecule (ICAM)-1, VCAM-1, and substance-P [37,38]. Injection of HIV-1 gp120 proteins to rats induced ROS, caused the upregulation of MMP-2 and MMP-9, and reduced brain claudin-5 and laminin (major component of endothelial basement membrane) [39,40]. Injection of HIV-1 Nef protein into rats also induced BBB disruption, reduced claudin-5, and increased the permeability of the BBB and other vascular tissues. This was associated with increased MMP-9 activity, inflammation, and leukocyte infiltration into tissues [43,44]. MMP-9 inhibitors prevented Nef-mediated BBB injury [43].

2.3. Evidence from In Vitro Studies

Several studies using in vitro BBB models showed that HIV-1 [26,46,47] and viral proteins, including gp120 [39,48,49,50,51], Tat [52,53,54,55,56,57,58], and Nef [59,60], induced endothelial injury and BBB dysfunction; they decreased the expression of endothelial TJs occludin, claudin-5, claudin-1, ZO-1 and ZO-2, decreased transendothelial electrical resistance (TEER), and increased endothelial paracellular permeability, leukocyte adhesion, and transendothelial migration. These HIV- and viral protein-induced endothelial injuries and BBB dysfunction also involved an altered endothelial cytoskeleton, increased cytotoxicity, mitochondrial dysfunction and oxidative stress, and increased expression and secretion of inflammatory cytokines and chemokines, vascular adhesion molecules, MMPs, and other extracellular matrix-degrading enzymes [26,39,46,47,51].

3. Factors and Mechanisms Involved in HIV-Mediated Endothelial Injury and BBB Dysfunction

3.1. HIV and Infected Leukocytes

Several mechanisms have been shown to mediate HIV transport across the BBB and CNS entry, including transport via mannose-6 phosphate receptor-mediated transcytosis [61] and inflammation-mediated diapedesis of infected leukocytes [62,63,64,65,66]. BBB dysfunction and increased CNS infiltration of leukocytes in HIV infection and HAND have been shown to involve dynamin-related protein-1-mediated mitochondrial dysfunction and oxidative injury [67], inflammation, and downregulation of ZO-1, ZO-2, claudin-1, claudin-5, and occludin [46,68,69]. These HIV-induced effects on the BBB were mediated by Rho-associated kinases, nuclear factor NF-κB, and signal transducer and transcription (STAT)-1 [26,46,68] pathways. Moreover, they were associated with increased MMP-2 and MMP-9 and the dysregulation of brain endothelial efflux transporters, voltage-gated ion channels, calcium-binding proteins, and cytoskeletal and regulatory proteins [64,69,70,71]. Patients with HAND or HIV encephalitis had increased CSF levels of oxidized proteins that correlated with decreased mitochondrial activity and cognitive impairment [72].
In HIV/AIDS, cell–cell communications also impact endothelial properties and function. Interactions between primary human brain microvascular endothelial cells (HBMECs) and HIV-infected macrophages have induced endothelial upregulation and activation of pro-inflammatory cytokines and chemokines, including interleukin (IL)-6, TNF-α-induced proteins, interferon-inducible genes, adhesion molecules such as ICAM-1, and transcription factors associated with NF-κB and STAT-1 pathways [46,69]. HIV-induced IL-6 and downregulation of endothelial TJ proteins was associated with increased activation of STAT-1 and STAT-3 and STAT1-mediated induction of interferon-stimulated response element/interferon-γ-activated sequence promoter activity [26,47]. Other studies using HBMECs and hCMEC/D3 cell lines showed that ECs’ exposure to HIV-infected monocytes decreased endothelial TJ and AJ proteins, and increased MMP-9, IL-1β, TNF-α, chemokine ligand-2 (CCL2), and E-selectin. Furthermore, ECs’ interaction with infected monocytes increased monocytes’ transendothelial migration via peroxisome proliferator-activated receptor (PPAR)-α, PPAR-γ, Rho GTPases, and caveolin-1-mediated activation of extracellular signal-regulated kinase-1/2 and protein kinase-B (AKT) [68,73,74,75,76].

3.2. HIV Proteins

3.2.1. HIV gp120

Several in vitro and in vivo studies showed that HIV-1 gp120 induced brain endothelial injury, increased BBB permeability and leukocytes’ transendothelial migration. These gp120-induced BBB dysfunctions were associated with calcium dysregulation, were mediated by the C-C chemokine receptor-5, and involved signaling via myosin light-chain kinase, protein kinase-C, and p38 mitogen-activated protein kinase (MAPK) pathways [48,49,50,77,78]. Exposure of HBMECs to gp120 decreased TJ proteins ZO-1, ZO-2, and occludin, decreased AJ proteins such as junctional adhesion molecule (JAM)-1, increased the transcription, expression, and secretion of pro-inflammatory cytokines and chemokines such as IL-6 and IL-8, increased leukocyte adhesion and transendothelial migration via STAT-1 pathways and crosstalk with phosphatidyl inositol-3 kinase (PI3K) and MAP2K pathways [49,50,51], and may also involve endocannabinoids [79]. Exposure of HBMECs to gp120, Tat, or inflammatory mediators (TNF-α and lipopolysaccharides) enhanced the release of occludin-containing extracellular vesicles via small GTPase ADP-ribosylation factor-6 [80]. Endothelial injury and impaired BBB integrity following exposure of brain ECs and mice to gp120 and Tat involved increased ROS, intracellular accumulation of lipid peroxidation products such as malondialdehyde, and decreased activity of glutathione, glutathione peroxidase, and glutathione reductase. The thiol antioxidant N-acetylcysteine amide abrogated these gp120- and Tat-induced effects [81,82,83].

3.2.2. HIV Tat

Exposure of brain ECs to HIV-1 Tat induced cytoskeletal changes, dysregulated efflux transporters such as P-glycoprotein and multidrug resistance protein-1, and decreased TEER and the TJ proteins occludin, claudin-1, claudin-5, and ZO-2 [52,53,54,55,56,57,58]. Tat induced apoptosis in ECs and this was associated with mitochondrial dysfunction, increased ROS, and endoplasmic reticulum stress mediated by phosphorylation of eukaryotic translation initiation factor-2α and activating transcription factor-4 [84]. ECs injury following Tat exposure involved increased activator protein-1 (AP-1), ICAM-1, and E-selectin, and the secretion of plasminogen activator inhibitor-1, cytokines and chemokines such as CCL2 and IL-6. These Tat-induced effects involved signaling via AKT, protein kinase-C and -A pathways [85,86,87,88]. Tat-induced brain endothelial injury and BBB dysfunction was also associated with activation/phosphorylation of VE-cadherin [89,90], Rho/Rho-kinase, NF-κB, AP-1, and vascular endothelial growth factor receptor-2 signaling pathways [55,58,91,92,93], oxidative stress and MMP9-mediated cleavage of occludin, resulting in increased BBB permeability [54,55,56], increased ROS and nitric oxide, and decreased intracellular glutathione via NF-κB, AP-1, and Ras pathways [94,95].

3.2.3. HIV Nef, Viral Protein R (Vpr), and p17

Intracellular and extracellular Nef induced mitochondrial dysfunction, caspase activation, and apoptosis in HBMECs [59]. Nef also decreased ZO-1, reduced TEER, increased BBB permeability, and increased chemokines and cytokines such as IL-12, IL-8, IL-6, CCL5, and IL-17A [60]. HIV-1 Vpr induced apoptosis in HBMECs [96], and the HIV-1 matrix protein p17 cross the BBB in vitro and in mice via transcytosis mediated by the CXC motif chemokine receptor-2 [97].

4. Endothelial Injury and BBB Dysfunction in HIV Infection and Substance Abuse

Substance abuse is associated with increased risk of HIV transmission and infection, and PLWH with substance use disorders are more likely to be diagnosed late, have poor adherence to ART, and experience accelerated disease progression, increased morbidity, neuropathological complications, and cognitive impairment [98,99]. Substances of abuse that have been investigated for effects on the brain endothelium in HIV/AIDS include cocaine, methamphetamine (meth), alcohol, nicotine/tobacco, opioids, and cannabinoids.

4.1. Cocaine

Cocaine alters the brain endothelium properties and function, and HIV and viral proteins further exacerbate these effects. Exposure of HBMECs to cocaine induced the breakdown of ZO-1 and JAM-2, decreased TEER, altered the endothelial cytoskeleton, upregulated the adhesion molecules ICAM-1, VCAM-1, platelet/endothelial cell adhesion molecule-1, and endothelial leukocyte adhesion molecule-1, and activated leukocyte cell adhesion molecule (ALCAM). Cocaine also upregulated inflammatory cytokines in ECs and increased endothelial paracellular permeability, leukocytes adhesion, and transendothelial migration [100,101,102,103,104,105]. Administration of cocaine to mice increased ALCAM, monocyte adhesion to brain microvessels, and transmigration into brain tissues [101]. HIV and viral proteins further potentiated cocaine-mediated endothelial injury and dysfunction. Cocaine and Tat synergistically decreased HBMECs TEER, ZO-1, and JAM-2, increased endothelial paracellular permeability, and increased monocytes transendothelial migration [102]. Cocaine increased cytokines and neuroinflammation in the CNS of HIV-1 Tg rats [106]. Compared to HIV+ subjects not using cocaine and seronegative controls, brain tissues of HIV+ subjects who abused cocaine showed significant ALCAM upregulation associated with macrophage accumulation around brain microvessels [101]. These cocaine-mediated BBB injuries and dysfunctions have been shown to involve the CCL2/C-C chemokine receptor-2 axis, sigma receptor and platelet-derived growth factor-β activation, NF-κB, MAPK, and PI3K/AKT pathways [100,101].

4.2. Meth

Meth, HIV-1, and viral proteins can individually and synergistically impair brain cells’ structure, properties, and functions, including cells of the BBB and neurovascular unit [107,108,109]. In vitro studies using hCMEC/D3 cells [110,111,112] and HBMECs [113] showed that Meth and Tat [110,111] or gp120 [113] decreased TEER, decreased the expression of glucose transporter-1 and -3, decreased the expression of TJ proteins ZO-1, occludin, claudin-3, and claudin-5, decreased the expression of AJ proteins JAM-A and JAM-2, and increased endothelial paracellular permeability [110,111,112,113]. Administration of Meth to HIV Tg rats did not alter the HIV-induced decrease in hippocampus TJs and did not increase BBB permeability or gliosis [114]. However, other in vivo studies showed that Meth and Tat or gp120 synergistically decreased TJs and increased BBB permeability in mice, rats, and treeshrews [81,111,115]. Common mechanisms associated with Meth and Tat- or gp120- induced endothelial injury and dysfunction included increased oxidative stress and lipid peroxidation, with increased ROS and malondialdehyde and decreased intracellular glutathione, glutathione peroxidase, and superoxide dismutase activity. Antioxidants such as N-acetylcysteine amide reduced or abrogated Meth and Tat- or gp120-induced BBB impairment [81,111,112,113,115].

4.3. Alcohol

Alcohol abuse negatively impacts brain function and can exacerbate HIV-associated CNS injury. Binge exposure of HIV Tg rats to ethanol, as well as in vitro exposure of rats and human brain ECs to ethanol and gp120, altered endothelial barrier properties (increased BBB permeability and decreased claudin-3, occludin, and JAM-2) via mechanisms involving the transient receptor potential melastatin-7 ion channel [116]. In vitro studies of HBMECs exposed to ethanol and HIV-1 Nef, Vpr, Tat, or gp120 proteins showed that ethanol potentiated viral protein-induced ECs apoptosis and TNF-α production [96]. Tat and alcohol individually and synergistically altered the BBB, increased macrophage transendothelial migration, and increased neurotoxicity [117]. However, another study showed that alcohol and gp120 individually increased HBMEC permeability, ROS production, and cytoskeletal remodeling, but found no synergistic effect [118].

4.4. Tobacco

In vivo and in vitro studies showed that nicotine, cotinine, and tobacco smoke extracts have limited effect on the BBB but their combination with protease inhibitors (saquinavir [SQV] and ritonavir [RTV/r]) or gp120 proteins significantly increased BBB injury and dysfunction. Exposure of rats’ brain ECs to nicotine, SQV, or RTV increased ROS, and combination of nicotine and SQV or SQV/r synergistically increased ROS and decreased ZO-1 [119]. In rats treated with nicotine, cotinine, SQV or SQV/r, individual treatment had no major effect on ZO-1 or BBB permeability, but a combination of nicotine or tobacco smoke extracts with SQV or SQV/r significantly decreased ZO-1 and increased BBB permeability (by 2 to 4-fold). These effects were associated with Notch-4 downregulation [119]. Nicotine, cotinine, and their combination target efflux transporters and dose-dependently increased BBB permeability to 14C-sucrose and SQV, and increased SQV accumulation in several brain regions (frontal, parietal, and occipital cortex, hippocampus, thalamus, hypo-thalamus, and caudate/putamen) [119,120]. HBMECs exposed to tobacco smoke extracts or gp120 showed decreased viability, mitochondrial membrane potential, ZO-1, occludin, and TEER, and increased BBB permeability. Combined treatment of HBMECs with tobacco smoke extracts and gp120 resulted in a synergistic decrease in TEER and an increase in paracellular permeability, associated with a 2- to 4-fold decrease in endothelial oxygen consumption, mitochondrial membrane potential, ZO-1, and occludin [121]. These data suggest that tobacco use in HIV infection may be associated with increased risk of BBB oxidative injury and dysfunction.

4.5. Opioids

Opioids abuse and overdoses have dramatically increased in the USA and worldwide, and there is evidence that opioids impact HIV immuno- and neuro-pathogenesis [122,123]. In vitro studies showed that chronic low doses of morphine (MOR) had no major effect on HBMECs [124]. However, evidence from other in vivo and in vitro studies suggests that in the presence of HIV proteins, opioids alter the BBB properties and function. Administration of MOR and Tat to mice increased leukocytes trafficking into the CNS and the combination of Tat and MOR potentiated CCL5 upregulation in the CNS [125]. Tat Tg mice treated with fentanyl [126] or MOR [127,128] showed increased BBB disruption and brain inflammatory markers [126,127,128] and increased BBB leakage and recruitment of circulating macrophages into the striatum, hippocampus parenchyma, and perivascular space [127,128]. However, buprenorphine, a partial opioid agonist, decreased CCL2-mediated adhesion of monocytes to HBMECs and transendothelial migration [129]. These results led to suggestions that buprenorphine can reduce HIV-mediated neuroinflammation in addition to its use for the treatment of opioid addiction [129]. Exposure of brain microvascular ECs to MOR and/or Tat altered the expression of ZO-1, occludin and JAM-2, and decreased TEER, which was associated with intracellular calcium release, activation of myosin light-chain kinase, and increased production of pro-inflammatory cytokines [130]. MOR also increased the accumulation of tenofovir, emtricitabine, and dolutegravir in HBMECs and hCMEC/D3 cells [131]. However, these in vitro findings may not necessarily translate to increased CNS entry of these antiretrovirals in vivo. Although MOR increased BBB leakage in Tat Tg mice, it significantly reduced CNS entry of abacavir and dolutegravir, and increased brain P-glycoprotein levels [127], which suggests that MOR may block CNS entry of antiretrovirals by upregulating BBB efflux transporters.

4.6. Cannabinoids

Cannabinoids have been shown to have anti-inflammatory and analgesic properties, and to have a protective effect on the BBB following brain injury. Cannabinoids increased TEER, decreased endothelial VCAM-1, reduced BBB permeability and ischemia-induced BBB damage [132,133], reduced ischemia-induced IL-6 and lactate dehydrogenase in astrocytes [133], increased endogenous antioxidant via NF-κB pathways, reduced cytokines (including TNF-α, IL-1β, and IL-6) expression, and decreased BBB permeability in mice following traumatic brain injury [134]. Cannabinoid-type 2 receptor (CB2R) is expressed in ECs, glial cells, neurons, and brain macrophages [135], and has been shown to mediate cannabinoid effects in vitro and in vivo. Activation of CB2R decreased inflammatory responses in human macrophages and HBMECs, preserved endothelial AJ and TJ proteins, and decreased BBB permeability [136]. CB2R activation decreased inflammation-induced ICAM-1 and VCAM-1, decreased monocytes’ adhesion to the brain endothelium, and decreased BBB permeability in vivo [137,138]. CB2R agonists increased endothelial AJs and TJs occludin, ZO-1, and claudin-5, restored BBB properties, reduced neuroinflammation and neurodegeneration, and attenuated brain injury in murine models of brain hemorrhage [139,140] and traumatic brain injury [141,142].
Cannabis is widely used among PLWH. Considering the evidence above, that cannabinoids attenuated inflammation and BBB damage following brain injury, it has been suggested that cannabinoids’ anti-inflammatory properties could counter HIV-induced neuroinflammation, BBB damage, and CNS injury [135,143]. CB1R and CB2R agonists decreased gp120-induced synapse loss and neuronal death in vitro [144], decreased gp120-induced downregulation of ZO-1, claudin-5, and JAM-1 in HBMECs, and decreased gp120-induced calcium influx, BBB permeability, and monocyte transmigration in vitro and in vivo [79]. In cross-sectional studies of an observational cohort, analyses of the blood and CSF of HIV-infected adults and seronegative controls, cannabis users (moderate and daily users), and non-users, showed that recent use of cannabis was associated with reduced inflammatory markers in both the CSF and blood [143]. Compared to HIV+ non-cannabis users, frequent and daily cannabis use was associated with lower BBB permeability (lower CSF/serum albumin ratio and CSF levels of soluble urokinase plasminogen activator receptor), lower CSF neurofilament-L [145], and lower CSF CCL2 and interferon gamma-induced protein-10 levels associated with better neurocognitive (learning) performance [146]. Longitudinal studies of older HIV-infected adults who were frequent cannabis users, occasional users, and non-users, showed better global cognitive performance among occasional users compared to HIV+ non-cannabis users. However, recent cannabis use was linked to worse cognition, especially regarding memory function [147]. In autopsy studies of brain tissues from HIV-infected humans on long-term ART, subjects with HAND [148], HIV encephalitis, and CNS comorbidities [149] had increased CB1R and CB2R in several brain regions, in neurons, glial cells, and meningeal and perivascular macrophages [148,149]. Increased CB1R was also associated with worse cognition, including poorer memory function and speed of information processing [148]. Furthermore, although CB2R agonists decreased gp120-induced neuronal injury, they had no effect on Tat-induced synapse loss or neuronal death [144]. Thus, it is not yet clear whether cannabinoids abrogate or exacerbate HIV-induced CNS injury and HAND, or the potential effect of the dosage, potency (e.g., delta-9- tetrahydrocannabinol content), and frequency of cannabis use, and polysubstance use.

5. Conclusions

The BBB is central to HAND neuropathogenesis. BBB dysfunction during the early stages of HIV infection enables viral entry into the brain, infection of brain cells, and subsequent inflammation, oxidative injury of CNS cells, neurodegeneration, and HAND. This comprehensive review of current evidence from studies in humans, human autopsy brain samples, animals, and in vitro BBB models shows that there is increased injury of the cerebral endothelium, BBB breakdown, and permeability in both symptomatic and asymptomatic HIV infections. HIV-1 proteins such as gp120, Tat, and Nef also induce BBB injury and dysfunction in vivo (Table 1) and in vitro (Table 2), and commonly abused substances such as cocaine, meth, opioids, and tobacco potentiate HIV- and viral protein-induced BBB injury and dysfunction (Table 1, Table 2 and Table 3).
This review also discussed the molecular mechanisms involved in BBB injury and dysfunction induced by HIV, viral proteins, and substances of abuse (overall findings are summarized in Table 1, Table 2 and Table 3). Synergistic increases in BBB damage with substance abuse and HIV or viral proteins would result in increased risk of CNS infection and HAND in HIV-infected substance abusers. As current evidence suggests that ART does not prevent or abrogate HIV-induced BBB injury, it would be important to determine whether ART alters HIV-induced neuropathology, and if any such CNS ART effects are influenced by antiretrovirals drug classes and CNS penetration.
Current evidence suggests that cannabinoids reduce inflammation and HIV-induced BBB injury. However, human studies show conflicting findings, including recent and frequent/daily cannabis use being associated with improved BBB function and better learning in cross-sectional studies and associated with worse cognition/poor memory function in longitudinal studies. Furthermore, human autopsy studies showed that HAND is associated with increased CB1R and CB2R, and demonstrate that increased CB1R is associated with worse memory function and speed of information processing. Thus, whether cannabinoids are neuroprotective or harmful in HIV CNS pathologies and HAND remains to be established.

Author Contributions

Conceptualization, investigation, supervision, data curation, formal analysis, writing—original draft preparation, review, and editing, G.D.K. Writing—original draft preparation, review, and editing, N.A. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by NIH grant R01 MH132517-01A1.

Acknowledgments

We thank Wendy Claire Kanmogne for the critical reading and editing of our manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

AIDSAcquired immunodeficiency syndrome
ALCAMActivated leukocyte cell adhesion molecule
AJsAdherent junctions
AKTProtein kinase B
AP-1Activator protein 1
ARTAntiretroviral therapy
BBBBlood–brain barrier
CB1RCannabinoid-type 1 receptor
CB2RCannabinoid-type 2 receptor
CCL2:Chemokine ligand 2
CCL5Chemokine ligand 5
CNSCentral nervous system
CSFCerebrospinal fluid
ECsEndothelial cells
Gp120Glycoprotein 120
HANDHIV-associated neurocognitive disorder
HBMECsHuman brain microvascular endothelial cells
HIVHuman immunodeficiency virus
ICAM-1Intercellular adhesion molecule 1
ILInterleukin
JAMJunctional adhesion molecule
MAPKMitogen-activated protein kinase
MMPsMatrix metalloproteinases
MORMorphine
NefNegative factor
NF-κBNuclear factor kappa B
QAIbCSF/serum albumin quotient
PLWHPeople living with HIV
PI3KPhosphatidyl inositol 3 kinase
PPARα/γPeroxisome proliferator-activated receptor alpha/gamma
ROSReactive oxygen species
RTV/rRitonavir
SQVSaquinavir
STATSignal transducer and activator of transcription
TatTrans-activator of transcription
TgTransgenic
TEERTransendothelial electrical resistance
TJsTight junctions
TNF-αTumor necrosis factor-alpha
SIVSimian immunodeficiency virus
VCAM-1Vascular cell adhesion molecule 1
VprViral protein R
ZOZonula occludens

References

  1. Alahmari, A. Blood-Brain Barrier Overview: Structural and Functional Correlation. Neural Plast. 2021, 2021, 6564585. [Google Scholar] [CrossRef] [PubMed]
  2. Kadry, H.; Noorani, B.; Cucullo, L. A blood–brain barrier overview on structure, function, impairment, and biomarkers of integrity. Fluids Barriers CNS 2020, 17, 69. [Google Scholar] [CrossRef] [PubMed]
  3. Daneman, R.; Prat, A. The Blood–Brain Barrier. Cold Spring Harb. Perspect. Biol. 2015, 7, a020412. [Google Scholar] [CrossRef]
  4. Segarra, M.; Aburto, M.R.; Acker-Palmer, A. Blood–Brain Barrier Dynamics to Maintain Brain Homeostasis. Trends Neurosci. 2021, 44, 393–405. [Google Scholar] [CrossRef]
  5. Borrajo, A.; Spuch, C.; Penedo, M.A.; Olivares, J.M.; Agís-Balboa, R.C. Important role of microglia in HIV-1 associated neurocognitive disorders and the molecular pathways implicated in its pathogenesis. Ann. Med. 2021, 53, 43–69. [Google Scholar] [CrossRef] [PubMed]
  6. Ginsberg, S.D.; Alldred, M.J.; Gunnam, S.M.; Schiroli, C.; Lee, S.H.; Morgello, S.; Fischer, T. Expression profiling suggests microglial impairment in human immunodeficiency virus neuropathogenesis. Ann. Neurol. 2018, 83, 406–417. [Google Scholar] [CrossRef]
  7. Mattson, M.P.; Haughey, N.J.; Nath, A. Cell death in HIV dementia. Cell Death Differ. 2005, 12, 893–904. [Google Scholar] [CrossRef]
  8. Kaul, M.; Zheng, J.; Okamoto, S.; Gendelman, H.E.; Lipton, S.A. HIV-1 infection and AIDS: Consequences for the central nervous system. Cell Death Differ. 2005, 12, 878–892. [Google Scholar] [CrossRef] [PubMed]
  9. Saylor, D.; Dickens, A.M.; Sacktor, N.; Haughey, N.; Slusher, B.; Pletnikov, M.; Mankowski, J.L.; Brown, A.; Volsky, D.J.; McArthur, J.C. HIV-associated neurocognitive disorder—Pathogenesis and prospects for treatment. Nat. Rev. Neurol. 2016, 12, 234–248. [Google Scholar] [CrossRef]
  10. Antinori, A.; Arendt, G.; Becker, J.T.; Brew, B.J.; Byrd, D.A.; Cherner, M.; Clifford, D.B.; Cinque, P.; Epstein, L.G.; Goodkin, K.; et al. Updated research nosology for HIV-associated neurocognitive disorders. Neurology 2007, 69, 1789–1799. [Google Scholar] [CrossRef]
  11. Bandera, A.; Taramasso, L.; Bozzi, G.; Muscatello, A.; Robinson, J.A.; Burdo, T.H.; Gori, A. HIV-Associated Neurocognitive Impairment in the Modern ART Era: Are We Close to Discovering Reliable Biomarkers in the Setting of Virological Suppression? Front. Aging Neurosci. 2019, 11, 187. [Google Scholar] [CrossRef] [PubMed]
  12. Clifford, D.B.; Ances, B.M. HIV-associated neurocognitive disorder. Lancet Infect. Dis. 2013, 13, 976–986. [Google Scholar] [CrossRef] [PubMed]
  13. Nightingale, S.; Ances, B.; Cinque, P.; Dravid, A.; Dreyer, A.J.; Gisslén, M.; Joska, J.A.; Kwasa, J.; Meyer, A.-C.; Mpongo, N.; et al. Cognitive impairment in people living with HIV: Consensus recommendations for a new approach. Nat. Rev. Neurol. 2023, 19, 424–433. [Google Scholar] [CrossRef] [PubMed]
  14. Heaton, R.K.; Clifford, D.B.; Franklin, D.R.; Woods, S.P.; Ake, C.; Vaida, F.; Ellis, R.J.; Letendre, S.L.; Marcotte, T.D.; Atkinson, J.H.; et al. HIV-associated neurocognitive disorders persist in the era of potent antiretroviral therapy. Neurology 2010, 75, 2087–2096. [Google Scholar] [CrossRef] [PubMed]
  15. Sacktor, N.; McDermott, M.P.; Marder, K.; Schifitto, G.; Selnes, O.A.; McArthur, J.C.; Stern, Y.; Albert, S.; Palumbo, D.; Kieburtz, K.; et al. HIV-associated cognitive impairment before and after the advent of combination therapy. J. NeuroVirol. 2002, 8, 136–142. [Google Scholar] [CrossRef]
  16. Marcus, J.L.; Leyden, W.A.; Alexeeff, S.E.; Anderson, A.N.; Hechter, R.C.; Hu, H.; Lam, J.O.; Towner, W.J.; Yuan, Q.; Horberg, M.A.; et al. Comparison of Overall and Comorbidity-Free Life Expectancy Between Insured Adults With and Without HIV Infection, 2000-2016. JAMA Netw. Open 2020, 3, e207954. [Google Scholar] [CrossRef]
  17. Trickey, A.; Sabin, C.A.; Burkholder, G.; Crane, H.; d‘Arminio Monforte, A.; Egger, M.; Gill, M.J.; Grabar, S.; Guest, J.L.; Jarrin, I.; et al. Life expectancy after 2015 of adults with HIV on long-term antiretroviral therapy in Europe and North America: A collaborative analysis of cohort studies. Lancet HIV 2023, 10, e295–e307. [Google Scholar] [CrossRef]
  18. Anesten, B.; Zetterberg, H.; Nilsson, S.; Brew, B.J.; Fuchs, D.; Price, R.W.; Gisslén, M.; Yilmaz, A. Effect of antiretroviral treatment on blood-brain barrier integrity in HIV-1 infection. BMC Neurol. 2021, 21, 494. [Google Scholar] [CrossRef]
  19. Calcagno, A.; Alberione, M.C.; Romito, A.; Imperiale, D.; Ghisetti, V.; Audagnotto, S.; Lipani, F.; Raviolo, S.; Di Perri, G.; Bonora, S. Prevalence and predictors of blood-brain barrier damage in the HAART era. J. NeuroVirol. 2014, 20, 521–525. [Google Scholar] [CrossRef]
  20. Caligaris, G.; Trunfio, M.; Ghisetti, V.; Cusato, J.; Nigra, M.; Atzori, C.; Imperiale, D.; Bonora, S.; Di Perri, G.; Calcagno, A. Blood–Brain Barrier Impairment in Patients Living with HIV: Predictors and Associated Biomarkers. Diagnostics 2021, 11, 867. [Google Scholar] [CrossRef]
  21. Avison, M.J.; Nath, A.; Greene-Avison, R.; Schmitt, F.A.; Greenberg, R.N.; Berger, J.R. Neuroimaging correlates of HIV-associated BBB compromise. J. Neuroimmunol. 2004, 157, 140–146. [Google Scholar] [CrossRef] [PubMed]
  22. Rahimy, E.; Li, F.-Y.; Hagberg, L.; Fuchs, D.; Robertson, K.; Meyerhoff, D.J.; Zetterberg, H.; Price, R.W.; Gisslén, M.; Spudich, S. Blood-Brain Barrier Disruption Is Initiated During Primary HIV Infection and Not Rapidly Altered by Antiretroviral Therapy. J. Infect. Dis. 2017, 215, 1132–1140. [Google Scholar] [CrossRef] [PubMed]
  23. Chaganti, J.; Marripudi, K.; Staub, L.P.; Rae, C.D.; Gates, T.M.; Moffat, K.J.; Brew, B.J. Imaging correlates of the blood–brain barrier disruption in HIV-associated neurocognitive disorder and therapeutic implications. AIDS 2019, 33, 1843–1852. [Google Scholar] [CrossRef] [PubMed]
  24. Calcagno, A.; Atzori, C.; Romito, A.; Vai, D.; Audagnotto, S.; Stella, M.L.; Montrucchio, C.; Imperiale, D.; Di Perri, G.; Bonora, S. Blood brain barrier impairment is associated with cerebrospinal fluid markers of neuronal damage in HIV-positive patients. J. NeuroVirol. 2016, 22, 88–92. [Google Scholar] [CrossRef] [PubMed]
  25. Sharma, A.L.; Wang, H.; Zhang, Z.; Millien, G.; Tyagi, M.; Hongpaisan, J. HIV Promotes Neurocognitive Impairment by Damaging the Hippocampal Microvessels. Mol. Neurobiol. 2022, 59, 4966–4986. [Google Scholar] [CrossRef]
  26. Chaudhuri, A.; Yang, B.; Gendelman, H.E.; Persidsky, Y.; Kanmogne, G.D. STAT1 signaling modulates HIV-1–induced inflammatory responses and leukocyte transmigration across the blood-brain barrier. Blood 2008, 111, 2062–2072. [Google Scholar] [CrossRef]
  27. Dallasta, L.M.; Pisarov, L.A.; Esplen, J.E.; Werley, J.V.; Moses, A.V.; Nelson, J.A.; Achim, C.L. Blood-Brain Barrier Tight Junction Disruption in Human Immunodeficiency Virus-1 Encephalitis. Am. J. Pathol. 1999, 155, 1915–1927. [Google Scholar] [CrossRef]
  28. Bai, F.; Bono, V.; Borghi, L.; Bonazza, F.; Falcinella, C.; Vitaletti, V.; Miraglia, F.; Trunfio, M.; Calcagno, A.; Cusato, J.; et al. Association between tight junction proteins and cognitive performance in untreated Persons living with HIV (PLWH). AIDS 2024, 38, 1292–1303. [Google Scholar] [CrossRef]
  29. Petito, C.K.; Cash, K.S. Blood-brain barrier abnormalities in acquired immunodeficiency syndrome: Immunohistochemical localization of serum proteins in postmortem brain. Ann. Neurol. 1992, 32, 658–666. [Google Scholar] [CrossRef] [PubMed]
  30. MacLean, A.G.; Belenchia, G.E.; Bieniemy, D.N.; Moroney-Rasmussen, T.A.; Lackner, A.A. Simian immunodeficiency virus disrupts extended lengths of the blood–brain barrier. J. Med. Primatol. 2005, 34, 237–242. [Google Scholar] [CrossRef]
  31. Luabeya, M.K.; Dallasta, L.M.; Achim, C.L.; Pauza, C.D.; Hamilton, R.L. Blood–brain barrier disruption in simian immunodeficiency virus encephalitis. Neuropathol. Appl. Neurobiol. 2000, 26, 454–462. [Google Scholar] [CrossRef] [PubMed]
  32. Orandle Marlene, S.; MacLean Andrew, G.; Sasseville Vito, G.; Alvarez, X.; Lackner Andrew, A. Enhanced Expression of Proinflammatory Cytokines in the Central Nervous System Is Associated with Neuroinvasion by Simian Immunodeficiency Virus and the Development of Encephalitis. J. Virol. 2002, 76, 5797–5802. [Google Scholar] [CrossRef] [PubMed]
  33. Clay Candice, C.; Rodrigues Denise, S.; Ho Yan, S.; Fallert Beth, A.; Janatpour, K.; Reinhart Todd, A.; Esser, U. Neuroinvasion of Fluorescein-Positive Monocytes in Acute Simian Immunodeficiency Virus Infection. J. Virol. 2007, 81, 12040–12048. [Google Scholar] [CrossRef] [PubMed]
  34. Sasseville, V.G.; Lane, J.H.; Walsh, D.; Ringler, D.J.; Lackner, A.A. VCAM-1 expression and leukocyte trafficking to the CNS occur early in infection with pathogenic isolates of SIV. J. Med. Primatol. 1995, 24, 123–131. [Google Scholar] [CrossRef] [PubMed]
  35. Sasseville, V.G.; Smith, M.M.; Mackay, C.R.; Pauley, D.R.; Mansfield, K.G.; Ringler, D.J.; Lackner, A.A. Chemokine expression in simian immunodeficiency virus-induced AIDS encephalitis. Am. J. Pathol. 1996, 149, 1459. [Google Scholar]
  36. Bhargavan, B.; Woollard, S.M.; McMillan, J.E.; Kanmogne, G.D. CCR5 antagonist reduces HIV-induced amyloidogenesis, tau pathology, neurodegeneration, and blood-brain barrier alterations in HIV-infected hu-PBL-NSG mice. Mol. Neurodegener. 2021, 16, 78. [Google Scholar] [CrossRef]
  37. Cioni, C.; Annunziata, P. Circulating gp120 alters the blood–brain barrier permeability in HIV-1 gp120 transgenic mice. Neurosci. Lett. 2002, 330, 299–301. [Google Scholar] [CrossRef]
  38. Toneatto, S.; Finco, O.; van der Putten, H.; Abrignani, S.; Annunziata, P. Evidence of blood-brain barrier alteration and activation in HIV-1 gp120 transgenic mice. AIDS 1999, 13, 2343–2348. [Google Scholar] [CrossRef]
  39. Louboutin, J.-P.; Agrawal, L.; Reyes, B.A.S.; Van Bockstaele, E.J.; Strayer, D.S. HIV-1 gp120-Induced Injury to the Blood-Brain Barrier: Role of Metalloproteinases 2 and 9 and Relationship to Oxidative Stress. J. Neuropathol. Exp. Neurol. 2010, 69, 801–816. [Google Scholar] [CrossRef]
  40. Louboutin, J.-P.; Reyes, B.A.S.; Agrawal, L.; Van Bockstaele, E.J.; Strayer, D.S. HIV-1 gp120 upregulates matrix metalloproteinases and their inhibitors in a rat model of HIV encephalopathy. Eur. J. Neurosci. 2011, 34, 2015–2023. [Google Scholar] [CrossRef] [PubMed]
  41. Liao, K.; Niu, F.; Hu, G.; Guo, M.-L.; Sil, S.; Buch, S. HIV Tat-mediated induction of autophagy regulates the disruption of ZO-1 in brain endothelial cells. Tissue Barriers 2020, 8, 1748983. [Google Scholar] [CrossRef]
  42. Leibrand, C.R.; Paris, J.J.; Ghandour, M.S.; Knapp, P.E.; Kim, W.-K.; Hauser, K.F.; McRae, M. HIV-1 Tat disrupts blood-brain barrier integrity and increases phagocytic perivascular macrophages and microglia in the dorsal striatum of transgenic mice. Neurosci. Lett. 2017, 640, 136–143. [Google Scholar] [CrossRef]
  43. Sporer, B.; Koedel, U.; Paul, R.; Kohleisen, B.; Erfle, V.; Fontana, A.; Pfister, H.W. Human immunodeficiency virus type-1 Nef protein induces blood-brain barrier disruption in the rat: Role of matrix metalloproteinase-9. J. Neuroimmunol. 2000, 102, 125–130. [Google Scholar] [CrossRef]
  44. Rivera, J.; Isidro, R.A.; Loucil-Alicea, R.Y.; Cruz, M.L.; Appleyard, C.B.; Isidro, A.A.; Chompre, G.; Colon-Rivera, K.; Noel, R.J., Jr. Infusion of HIV-1 Nef-expressing astrocytes into the rat hippocampus induces enteropathy and interstitial pneumonitis and increases blood–brain-barrier permeability. PLoS ONE 2019, 14, e0225760. [Google Scholar] [CrossRef] [PubMed]
  45. Singh, V.B.; Singh, M.V.; Gorantla, S.; Poluektova, L.Y.; Maggirwar, S.B. Smoothened Agonist Reduces Human Immunodeficiency Virus Type-1-Induced Blood-Brain Barrier Breakdown in Humanized Mice. Sci. Rep. 2016, 6, 26876. [Google Scholar] [CrossRef] [PubMed]
  46. Chaudhuri, A.; Duan, F.; Morsey, B.; Persidsky, Y.; Kanmogne, G.D. HIV-1 Activates Proinflammatory and Interferon-Inducible Genes in Human Brain Microvascular Endothelial Cells: Putative Mechanisms of Blood—Brain Barrier Dysfunction. J. Cereb. Blood Flow Metab. 2008, 28, 697–711. [Google Scholar] [CrossRef] [PubMed]
  47. Yang, B.; Singh, S.; Bressani, R.; Kanmogne, G.D. Cross-talk between STAT1 and PI3K/AKT signaling in HIV-1-induced blood–brain barrier dysfunction: Role of CCR5 and implications for viral neuropathogenesis. J. Neurosci. Res. 2010, 88, 3090–3101. [Google Scholar] [CrossRef] [PubMed]
  48. Kanmogne, G.D.; Kennedy, R.C.; Grammas, P. HIV-1 gp120 Proteins and gp160 Peptides Are Toxic to Brain Endothelial Cells and Neurons: Possible Pathway for HIV Entry into the Brain and HIV-Associated Dementia. J. Neuropathol. Exp. Neurol. 2002, 61, 992–1000. [Google Scholar] [CrossRef]
  49. Kanmogne, G.D.; Primeaux, C.; Grammas, P. HIV-1 gp120 Proteins Alter Tight Junction Protein Expression and Brain Endothelial Cell Permeability: Implications for the Pathogenesis of HIV-Associated Dementia. J. Neuropathol. Exp. Neurol. 2005, 64, 498–505. [Google Scholar] [CrossRef]
  50. Kanmogne, G.D.; Schall, K.; Leibhart, J.; Knipe, B.; Gendelman, H.E.; Persidsky, Y. HIV-1 gp120 Compromises Blood–Brain Barrier Integrity and Enhance Monocyte Migration across Blood–Brain Barrier: Implication for Viral Neuropathogenesis. J. Cereb. Blood Flow Metab. 2007, 27, 123–134. [Google Scholar] [CrossRef]
  51. Yang, B.; Akhter, S.; Chaudhuri, A.; Kanmogne, G.D. HIV-1 gp120 induces cytokine expression, leukocyte adhesion, and transmigration across the blood–brain barrier: Modulatory effects of STAT1 signaling. Microvasc. Res. 2009, 77, 212–219. [Google Scholar] [CrossRef] [PubMed]
  52. Avraham, H.K.; Jiang, S.; Lee, T.-H.; Prakash, O.; Avraham, S. HIV-1 Tat-Mediated Effects on Focal Adhesion Assembly and Permeability in Brain Microvascular Endothelial Cells1. J. Immunol. 2004, 173, 6228–6233. [Google Scholar] [CrossRef]
  53. Bhargavan, B.; Kanmogne, G.D. Differential Mechanisms of Inflammation and Endothelial Dysfunction by HIV-1 Subtype-B and Recombinant CRF02_AG Tat Proteins on Human Brain Microvascular Endothelial Cells: Implications for Viral Neuropathogenesis. Mol. Neurobiol. 2018, 55, 1352–1363. [Google Scholar] [CrossRef] [PubMed]
  54. András, I.E.; Pu, H.; Deli, M.A.; Nath, A.; Hennig, B.; Toborek, M. HIV-1 Tat protein alters tight junction protein expression and distribution in cultured brain endothelial cells. J. Neurosci. Res. 2003, 74, 255–265. [Google Scholar] [CrossRef] [PubMed]
  55. Chen, Y.; Huang, W.; Jiang, W.; Wu, X.; Ye, B.; Zhou, X. HIV-1 Tat Regulates Occludin and Aβ Transfer Receptor Expression in Brain Endothelial Cells via Rho/ROCK Signaling Pathway. Oxidative Med. Cell. Longev. 2016, 2016, 4196572. [Google Scholar] [CrossRef]
  56. Xu, R.; Feng, X.; Xie, X.; Zhang, J.; Wu, D.; Xu, L. HIV-1 Tat protein increases the permeability of brain endothelial cells by both inhibiting occludin expression and cleaving occludin via matrix metalloproteinase-9. Brain Res. 2012, 1436, 13–19. [Google Scholar] [CrossRef] [PubMed]
  57. Hayashi, K.; Pu, H.; Tian, J.; Andras, I.E.; Lee, Y.W.; Hennig, B.; Toborek, M. HIV-Tat protein induces P-glycoprotein expression in brain microvascular endothelial cells. J. Neurochem. 2005, 93, 1231–1241. [Google Scholar] [CrossRef] [PubMed]
  58. Zhong, Y.; Hennig, B.; Toborek, M. Intact Lipid Rafts Regulate HIV-1 Tat Protein-Induced Activation of the Rho Signaling and Upregulation of P-Glycoprotein in Brain Endothelial Cells. J. Cereb. Blood Flow Metab. 2010, 30, 522–533. [Google Scholar] [CrossRef]
  59. Acheampong Edward, A.; Parveen, Z.; Muthoga Lois, W.; Kalayeh, M.; Mukhtar, M.; Pomerantz Roger, J. Human Immunodeficiency Virus Type 1 Nef Potently Induces Apoptosis in Primary Human Brain Microvascular Endothelial Cells via the Activation of Caspases. J. Virol. 2005, 79, 4257–4269. [Google Scholar] [CrossRef] [PubMed]
  60. Raymond, A.D.; Diaz, P.; Chevelon, S.; Agudelo, M.; Yndart-Arias, A.; Ding, H.; Kaushik, A.; Jayant, R.D.; Nikkhah-Moshaie, R.; Roy, U.; et al. Microglia-derived HIV Nef+ exosome impairment of the blood–brain barrier is treatable by nanomedicine-based delivery of Nef peptides. J. NeuroVirol. 2016, 22, 129–139. [Google Scholar] [CrossRef]
  61. Dohgu, S.; Ryerse, J.S.; Robinson, S.M.; Banks, W.A. Human Immunodeficiency Virus-1 Uses the Mannose-6-Phosphate Receptor to Cross the Blood-Brain Barrier. PLoS ONE 2012, 7, e39565. [Google Scholar] [CrossRef] [PubMed]
  62. Wang, H.; Sun, J.; Goldstein, H. Human Immunodeficiency Virus Type 1 Infection Increases the In Vivo Capacity of Peripheral Monocytes To Cross the Blood-Brain Barrier into the Brain and the In Vivo Sensitivity of the Blood-Brain Barrier to Disruption by Lipopolysaccharide. J. Virol. 2008, 82, 7591–7600. [Google Scholar] [CrossRef] [PubMed]
  63. Williams, D.W.; Eugenin, E.A.; Calderon, T.M.; Berman, J.W. Monocyte maturation, HIV susceptibility, and transmigration across the blood brain barrier are critical in HIV neuropathogenesis. J. Leukoc. Biol. 2012, 91, 401–415. [Google Scholar] [CrossRef] [PubMed]
  64. Eugenin, E.A.; Osiecki, K.; Lopez, L.; Goldstein, H.; Calderon, T.M.; Berman, J.W. CCL2/Monocyte Chemoattractant Protein-1 Mediates Enhanced Transmigration of Human Immunodeficiency Virus (HIV)-Infected Leukocytes across the Blood–Brain Barrier: A Potential Mechanism of HIV–CNS Invasion and NeuroAIDS. J. Neurosci. 2006, 26, 1098–1106. [Google Scholar] [CrossRef] [PubMed]
  65. Nottet, H.S.L.M. Interactions between macrophages and brain microvascular endothelial cells: Role in pathogenesis of HIV-1 infection and blood-brain barrier function. J. Neurovirol. 1999, 5, 659–669. [Google Scholar] [CrossRef]
  66. Persidsky, Y.; Zheng, J.; Miller, D.; Gendelman, H.E. Mononuclear phagocytes mediate blood-brain barrier compromise and neuronal injury during HIV-1-associated dementia. J. Leukoc. Biol. 2000, 68, 413–422. [Google Scholar] [CrossRef]
  67. Chandra, P.K.; Rutkai, I.; Kim, H.; Braun, S.E.; Abdel-Mageed, A.B.; Mondal, D.; Busija, D.W. Latent HIV-Exosomes Induce Mitochondrial Hyperfusion Due to Loss of Phosphorylated Dynamin-Related Protein 1 in Brain Endothelium. Mol. Neurobiol. 2021, 58, 2974–2989. [Google Scholar] [CrossRef]
  68. Persidsky, Y.; Heilman, D.; Haorah, J.; Zelivyanskaya, M.; Persidsky, R.; Weber, G.A.; Shimokawa, H.; Kaibuchi, K.; Ikezu, T. Rho-mediated regulation of tight junctions during monocyte migration across the blood-brain barrier in HIV-1 encephalitis (HIVE). Blood 2006, 107, 4770–4780. [Google Scholar] [CrossRef] [PubMed]
  69. Ricardo-Dukelow, M.; Kadiu, I.; Rozek, W.; Schlautman, J.; Persidsky, Y.; Ciborowski, P.; Kanmogne, G.D.; Gendelman, H.E. HIV-1 infected monocyte-derived macrophages affect the human brain microvascular endothelial cell proteome: New insights into blood-brain barrier dysfunction for HIV-1-associated dementia. J. Neuroimmunol. 2007, 185, 37–46. [Google Scholar] [CrossRef]
  70. McRae, M. HIV and viral protein effects on the blood brain barrier. Tissue Barriers 2016, 4, e1143543. [Google Scholar] [CrossRef]
  71. Langford, D.; Grigorian, A.; Hurford, R.; Adame, A.; Ellis, R.J.; Hansen, L.; Masliah, E. Altered P-Glycoprotein Expression in AIDS Patients with HIV Encephalitis. J. Neuropathol. Exp. Neurol. 2004, 63, 1038–1047. [Google Scholar] [CrossRef] [PubMed]
  72. Turchan, J.; Pocernich, C.B.; Gairola, C.; Chauhan, A.; Schifitto, G.; Butterfield, D.A.; Buch, S.; Narayan, O.; Sinai, A.; Geiger, J.; et al. Oxidative stress in HIV demented patients and protection ex vivo with novel antioxidants. Neurology 2003, 60, 307–314. [Google Scholar] [CrossRef]
  73. Huang, W.; András, I.E.; Rha, G.B.; Hennig, B.; Toborek, M. PPARα and PPARγ protect against HIV-1-induced MMP-9 overexpression via caveolae-associated ERK and Akt signaling. FASEB J. 2011, 25, 3979–3988. [Google Scholar] [CrossRef] [PubMed]
  74. Huang, W.; Eum, S.Y.; András, I.E.; Hennig, B.; Toborek, M. PPARα and PPARγ attenuate HIV-induced dysrégulation of tight junction proteins by modulations of matrix metalloproteinase and proteasome activities. FASEB J. 2009, 23, 1596–1606. [Google Scholar] [CrossRef] [PubMed]
  75. Huang, W.; Rha, G.B.; Han, M.-J.; Eum, S.Y.; András, I.E.; Zhong, Y.; Hennig, B.; Toborek, M. PPARα and PPARγ effectively protect against HIV-induced inflammatory responses in brain endothelial cells. J. Neurochem. 2008, 107, 497–509. [Google Scholar] [CrossRef]
  76. Ramirez, S.H.; Heilman, D.; Morsey, B.; Potula, R.; Haorah, J.; Persidsky, Y. Activation of Peroxisome Proliferator-Activated Receptor γ (PPARγ) Suppresses Rho GTPases in Human Brain Microvascular Endothelial Cells and Inhibits Adhesion and Transendothelial Migration of HIV-1 Infected Monocytes1. J. Immunol. 2008, 180, 1854–1865. [Google Scholar] [CrossRef]
  77. Khan, N.A.; Di Cello, F.; Stins, M.; Kim, K.S. Gp120-mediated cytotoxicity of human brain microvascular endothelial cells is dependent on p38 mitogen-activated protein kinase activation. J. NeuroVirol. 2007, 13, 242–251. [Google Scholar] [CrossRef]
  78. Banks, W.A.; Kastin, A.J.; Brennan, J.M.; Vallance, K.L. Adsorptive Endocytosis of HIV-1gp120 by Blood–Brain Barrier Is Enhanced by Lipopolysaccharide. Exp. Neurol. 1999, 156, 165–171. [Google Scholar] [CrossRef]
  79. Lu, T.-S.; Avraham, H.K.; Seng, S.; Tachado, S.D.; Koziel, H.; Makriyannis, A.; Avraham, S. Cannabinoids Inhibit HIV-1 Gp120-Mediated Insults in Brain Microvascular Endothelial Cells1. J. Immunol. 2008, 181, 6406–6416. [Google Scholar] [CrossRef]
  80. Ramirez, S.H.; Buzhdygan, T.P.; Hale, J.F.; Cheng, L.; Li, G.; Hoover-Hankerson, B.; Razmpour, R.; Sriram, U.; Su, L.; Potula, R.; et al. Extracellular Microvesicles Released From Brain Endothelial Cells are Detected in Animal Models Of HIV-1 Signifying Unresolved Inflammation. J. Neuroimmune Pharmacol. 2021, 16, 785–795. [Google Scholar] [CrossRef]
  81. Banerjee, A.; Zhang, X.; Manda, K.R.; Banks, W.A.; Ercal, N. HIV proteins (gp120 and Tat) and methamphetamine in oxidative stress-induced damage in the brain: Potential role of the thiol antioxidant N-acetylcysteine amide. Free Radic. Biol. Med. 2010, 48, 1388–1398. [Google Scholar] [CrossRef] [PubMed]
  82. Price, T.O.; Ercal, N.; Nakaoke, R.; Banks, W.A. HIV-1 viral proteins gp120 and Tat induce oxidative stress in brain endothelial cells. Brain Res. 2005, 1045, 57–63. [Google Scholar] [CrossRef]
  83. Price, T.O.; Uras, F.; Banks, W.A.; Ercal, N. A novel antioxidant N-acetylcysteine amide prevents gp120- and Tat-induced oxidative stress in brain endothelial cells. Exp. Neurol. 2006, 201, 193–202. [Google Scholar] [CrossRef]
  84. Ma, R.; Yang, L.; Niu, F.; Buch, S. HIV Tat-Mediated Induction of Human Brain Microvascular Endothelial Cell Apoptosis Involves Endoplasmic Reticulum Stress and Mitochondrial Dysfunction. Mol. Neurobiol. 2016, 53, 132–142. [Google Scholar] [CrossRef] [PubMed]
  85. ZIDOVETZKI, R.; WANG, J.-L.; CHEN, P.; JEYASEELAN, R.; HOFMAN, F. Human Immunodeficiency Virus Tat Protein Induces Interleukin 6 mRNA Expression in Human Brain Endothelial Cells via Protein Kinase C- and cAMP-Dependent Protein Kinase Pathways. AIDS Res. Hum. Retroviruses 1998, 14, 825–833. [Google Scholar] [CrossRef] [PubMed]
  86. Weiss, J.M.; Nath, A.; Major, E.O.; Berman, J.W. HIV-1 Tat Induces Monocyte Chemoattractant Protein-1-Mediated Monocyte Transmigration Across a Model of the Human Blood-Brain Barrier and Up-Regulates CCR5 Expression on Human Monocytes. J. Immunol. 1999, 163, 2953–2959. [Google Scholar] [CrossRef] [PubMed]
  87. Hofman, F.M.; Dohadwala, M.M.; Wright, A.D.; Hinton, D.R.; Walker, S.M. Exogenous tat protein activates central nervous system-derived endothelial cells. J. Neuroimmunol. 1994, 54, 19–28. [Google Scholar] [CrossRef]
  88. Huang, W.; Mo, X.; Wu, X.; Luo, W.; Chen, Y. Rosiglitazone suppresses HIV-1 Tat-induced vascular inflammation via Akt signaling. Mol. Cell. Biochem. 2015, 407, 173–179. [Google Scholar] [CrossRef]
  89. Mishra, R.; Singh, S.K. HIV-1 Tat C phosphorylates VE-cadherin complex and increases human brain microvascular endothelial cell permeability. BMC Neurosci. 2014, 15, 80. [Google Scholar] [CrossRef]
  90. Mishra, R.; Singh, S.K. HIV-1 Tat C Modulates Expression of miRNA-101 to Suppress VE-Cadherin in Human Brain Microvascular Endothelial Cells. J. Neurosci. 2013, 33, 5992–6000. [Google Scholar] [CrossRef]
  91. Toborek, M.; Lee, Y.W.; Pu, H.; Malecki, A.; Flora, G.; Garrido, R.; Hennig, B.; Bauer, H.-C.; Nath, A. HIV-Tat protein induces oxidative and inflammatory pathways in brain endothelium. J. Neurochem. 2003, 84, 169–179. [Google Scholar] [CrossRef] [PubMed]
  92. András, I.E.; Pu, H.; Tian, J.; Deli, M.A.; Nath, A.; Hennig, B.; Toborek, M. Signaling Mechanisms of HIV-1 Tat-Induced Alterations of Claudin-5 Expression in Brain Endothelial Cells. J. Cereb. Blood Flow Metab. 2005, 25, 1159–1170. [Google Scholar] [CrossRef] [PubMed]
  93. Khan, N.A.; Di Cello, F.; Nath, A.; Kim, K.S. Human immunodeficiency virus type 1 Tat-mediated cytotoxicity of human brain microvascular endothelial cells. J. NeuroVirol. 2003, 9, 584–593. [Google Scholar] [CrossRef]
  94. Kim, T.-A.; Avraham, H.K.; Koh, Y.-H.; Jiang, S.; Park, I.-W.; Avraham, S. HIV-1 Tat-Mediated Apoptosis in Human Brain Microvascular Endothelial Cells1. J. Immunol. 2003, 170, 2629–2637. [Google Scholar] [CrossRef]
  95. Jiang, W.; Huang, W.; Chen, Y.; Zou, M.; Peng, D.; Chen, D. HIV-1 Transactivator Protein Induces ZO-1 and Neprilysin Dysfunction in Brain Endothelial Cells via the Ras Signaling Pathway. Oxidative Med. Cell. Longev. 2017, 2017, 3160360. [Google Scholar] [CrossRef] [PubMed]
  96. Acheampong, E.; Mukhtar, M.; Parveen, Z.; Ngoubilly, N.; Ahmad, N.; Patel, C.; Pomerantz, R.J. Ethanol Strongly Potentiates Apoptosis Induced by HIV-1 Proteins in Primary Human Brain Microvascular Endothelial Cells. Virology 2002, 304, 222–234. [Google Scholar] [CrossRef] [PubMed]
  97. Caccuri, F.; Neves, V.; Gano, L.; Correia, J.D.G.; Oliveira, M.C.; Mazzuca, P.; Caruso, A.; Castanho, M. The HIV-1 Matrix Protein p17 Does Cross the Blood-Brain Barrier. J. Virol. 2022, 96, e01200-21. [Google Scholar] [CrossRef]
  98. Chibanda, D.; Benjamin, L.; Weiss, H.A.; Abas, M. Mental, neurological, and substance use disorders in people living with HIV/AIDS in low- and middle-income countries. J. Acquir. Immune Defic. Syndr. 2014, 67 (Suppl. 1), S54–S67. [Google Scholar] [CrossRef]
  99. Chilunda, V.; Calderon, T.M.; Martinez-Aguado, P.; Berman, J.W. The impact of substance abuse on HIV-mediated neuropathogenesis in the current ART era. Brain Res. 2019, 1724, 146426. [Google Scholar] [CrossRef] [PubMed]
  100. Dhillon, N.K.; Peng, F.; Bokhari, S.; Callen, S.; Shin, S.-H.; Zhu, X.; Kim, K.-J.; Buch, S.J. Cocaine-mediated Alteration in Tight Junction Protein Expression and Modulation of CCL2/CCR2 Axis Across the Blood-Brain Barrier: Implications for HIV-Dementia. J. Neuroimmune Pharmacol. 2008, 3, 52–56. [Google Scholar] [CrossRef]
  101. Yao, H.; Kim, K.; Duan, M.; Hayashi, T.; Guo, M.; Morgello, S.; Prat, A.; Wang, J.; Su, T.-P.; Buch, S. Cocaine Hijacks σ1 Receptor to Initiate Induction of Activated Leukocyte Cell Adhesion Molecule: Implication for Increased Monocyte Adhesion and Migration in the CNS. J. Neurosci. 2011, 31, 5942–5955. [Google Scholar] [CrossRef] [PubMed]
  102. Gandhi, N.; Saiyed, Z.M.; Napuri, J.; Samikkannu, T.; Reddy, P.V.B.; Agudelo, M.; Khatavkar, P.; Saxena, S.K.; Nair, M.P.N. Interactive role of human immunodeficiency virus type 1 (HIV-1) clade-specific Tat protein and cocaine in blood-brain barrier dysfunction: Implications for HIV-1-associated neurocognitive disorder. J. NeuroVirol. 2010, 16, 294–305. [Google Scholar] [CrossRef] [PubMed]
  103. Fiala, M.; Eshleman, A.J.; Cashman, J.; Lin, J.; Lossinsky, A.S.; Suarez, V.; Yang, W.; Zhang, J.; Popik, W.; Singer, E.; et al. Cocaine increases human immunodeficiency virus type 1 neuroinvasion through remodeling brain microvascular endothelial cells. J. NeuroVirol. 2005, 11, 281–291. [Google Scholar] [CrossRef]
  104. Fiala, M.; Gan, X.H.; Zhang, L.; House, S.D.; Newton, T.; Graves, M.C.; Shapshak, P.; Stins, M.; Kim, K.S.; Witte, M.; et al. Cocaine Enhances Monocyte Migration Across the Blood-Brain Barrier. In Drugs of Abuse, Immunomodulation, and Aids; Friedman, H., Madden, J.J., Klein, T.W., Eds.; Springer: Boston, MA, USA, 1998; pp. 199–205. [Google Scholar]
  105. Gan, X.; Zhang, L.; Berger, O.; Stins, M.F.; Way, D.; Taub, D.D.; Chang, S.L.; Kim, K.S.; House, S.D.; Weinand, M.; et al. Cocaine Enhances Brain Endothelial Adhesion Molecules and Leukocyte Migration. Clin. Immunol. 1999, 91, 68–76. [Google Scholar] [CrossRef]
  106. Ezeomah, C.; Fongsaran, C.; Persons, A.L.; Napier, T.C.; Cisneros, I.E. Cocaine Self-Administration Influences Central Nervous System Immune Responses in Male HIV-1 Transgenic Rats. Cells 2022, 11, 2405. [Google Scholar] [CrossRef]
  107. Fattakhov, N.; Torices, S.; Stangis, M.; Park, M.; Toborek, M. Synergistic Impairment of the Neurovascular Unit by HIV-1 Infection and Methamphetamine Use: Implications for HIV-1-Associated Neurocognitive Disorders. Viruses 2021, 13, 1883. [Google Scholar] [CrossRef]
  108. Mediouni, S.; Marcondes, M.C.; Miller, C.; McLaughlin, J.P.; Valente, S.T. The cross-talk of HIV-1 Tat and methamphetamine in HIV-associated neurocognitive disorders. Front. Microbiol. 2015, 6, 1164. [Google Scholar] [CrossRef] [PubMed]
  109. Northrop, N.A.; Yamamoto, B.K. Methamphetamine effects on blood-brain barrier structure and function. Front. Neurosci. 2015, 9, 69. [Google Scholar] [CrossRef]
  110. Patel, S.; Leibrand, C.R.; Palasuberniam, P.; Couraud, P.-O.; Weksler, B.; Jahr, F.M.; McClay, J.L.; Hauser, K.F.; McRae, M. Effects of HIV-1 Tat and Methamphetamine on Blood-Brain Barrier Integrity and Function In Vitro. Antimicrob. Agents Chemother. 2017, 61, e01307. [Google Scholar] [CrossRef]
  111. Huang, J.; Zhang, R.; Wang, S.; Zhang, D.; Leung, C.-K.; Yang, G.; Li, Y.; Liu, L.; Xu, Y.; Lin, S.; et al. Methamphetamine and HIV-Tat Protein Synergistically Induce Oxidative Stress and Blood-Brain Barrier Damage via Transient Receptor Potential Melastatin 2 Channel. Front. Pharmacol. 2021, 12, 619436. [Google Scholar] [CrossRef]
  112. Zhang, X.; Banerjee, A.; Banks, W.A.; Ercal, N. N-Acetylcysteine amide protects against methamphetamine-induced oxidative stress and neurotoxicity in immortalized human brain endothelial cells. Brain Res. 2009, 1275, 87–95. [Google Scholar] [CrossRef]
  113. Mahajan, S.D.; Aalinkeel, R.; Sykes, D.E.; Reynolds, J.L.; Bindukumar, B.; Adal, A.; Qi, M.; Toh, J.; Xu, G.; Prasad, P.N.; et al. Methamphetamine alters blood brain barrier permeability via the modulation of tight junction expression: Implication for HIV-1 neuropathogenesis in the context of drug abuse. Brain Res. 2008, 1203, 133–148. [Google Scholar] [CrossRef] [PubMed]
  114. Ohene-Nyako, M.; Persons, A.L.; Napier, T.C. Hippocampal blood–brain barrier of methamphetamine self-administering HIV-1 transgenic rats. Eur. J. Neurosci. 2021, 53, 416–429. [Google Scholar] [CrossRef]
  115. Li, J.; Huang, J.; He, Y.; Wang, W.; Leung, C.-K.; Zhang, D.; Zhang, R.; Wang, S.; Li, Y.; Liu, L.; et al. The protective effect of gastrodin against the synergistic effect of HIV-Tat protein and METH on the blood–brain barrier via glucose transporter 1 and glucose transporter 3. Toxicol. Res. 2021, 10, 91–101. [Google Scholar] [CrossRef]
  116. Mack, M.L.; Huang, W.; Chang, S.L. Involvement of TRPM7 in Alcohol-Induced Damage of the Blood–Brain Barrier in the Presence of HIV Viral Proteins. Int. J. Mol. Sci. 2023, 24, 1910. [Google Scholar] [CrossRef] [PubMed]
  117. Agas, A.; Garcia, R.; Kalluru, J.; Leiser, B.; Haorah, J. Synergistic effects of alcohol and HIV TAT protein on macrophage migration and neurotoxicity. J. Neuroimmunol. 2022, 368, 577869. [Google Scholar] [CrossRef]
  118. Shiu, C.; Barbier, E.; Cello, F.D.; Choi, H.J.; Stins, M. HIV-1 gp120 as Well as Alcohol Affect Blood–Brain Barrier Permeability and Stress Fiber Formation: Involvement of Reactive Oxygen Species. Alcohol. Clin. Exp. Res. 2007, 31, 130–137. [Google Scholar] [CrossRef] [PubMed]
  119. Manda, V.K.; Mittapalli, R.K.; Geldenhuys, W.J.; Lockman, P.R. Chronic exposure to nicotine and saquinavir decreases endothelial Notch-4 expression and disrupts blood-brain barrier integrity. J. Neurochem. 2010, 115, 515–525. [Google Scholar] [CrossRef]
  120. Manda, V.K.; Mittapalli, R.K.; Bohn, K.A.; Adkins, C.E.; Lockman, P.R. Nicotine and cotinine increases the brain penetration of saquinavir in rat. J. Neurochem. 2010, 115, 1495–1507. [Google Scholar] [CrossRef]
  121. Bhalerao, A.; Cucullo, L. HIV-1 gp120 and tobacco smoke synergistically disrupt the integrity of the blood-brain barrier. Eur. J. Cell Biol. 2022, 101, 151271. [Google Scholar] [CrossRef]
  122. Murphy, A.; Barbaro, J.; Martinez-Aguado, P.; Chilunda, V.; Jaureguiberry-Bravo, M.; Berman, J.W. The Effects of Opioids on HIV Neuropathogenesis. Front. Immunol. 2019, 10, 2445. [Google Scholar] [CrossRef] [PubMed]
  123. Buch, S.; Periyasamy, P.; Thangaraj, A.; Sil, S.; Chivero, E.T.; Tripathi, A. Opioid-Mediated HIV-1 Immunopathogenesis. J. Neuroimmune Pharmacol. 2020, 15, 628–642. [Google Scholar] [CrossRef] [PubMed]
  124. Marino, J.; Maubert, M.E.; Lawrence, J.M.; Wigdahl, B.; Nonnemacher, M.R. Chronic Low Dose Morphine Does Not Alter Two In Vitro BBB Models. Brain Sci. 2022, 12, 888. [Google Scholar] [CrossRef]
  125. Dutta, R.; Roy, S. Chronic morphine and HIV-1 Tat promote differential central nervous system trafficking of CD3+ and Ly6C+ immune cells in a murine Streptococcus pneumoniae infection model. J. Neuroinflamm. 2015, 12, 120. [Google Scholar] [CrossRef] [PubMed]
  126. Rademeyer, K.M.; Nass, S.R.; Jones, A.M.; Ohene-Nyako, M.; Hauser, K.F.; McRae, M. Fentanyl dysregulates neuroinflammation and disrupts blood-brain barrier integrity in HIV-1 Tat transgenic mice. J. NeuroVirol. 2024, 30, 1–21. [Google Scholar] [CrossRef]
  127. Leibrand, C.R.; Paris, J.J.; Jones, A.M.; Masuda, Q.N.; Halquist, M.S.; Kim, W.-K.; Knapp, P.E.; Kashuba, A.D.M.; Hauser, K.F.; McRae, M. HIV-1 Tat and opioids act independently to limit antiretroviral brain concentrations and reduce blood–brain barrier integrity. J. NeuroVirol. 2019, 25, 560–577. [Google Scholar] [CrossRef]
  128. Leibrand, C.R.; Paris, J.J.; Jones, A.M.; Ohene-Nyako, M.; Rademeyer, K.M.; Nass, S.R.; Kim, W.-K.; Knapp, P.E.; Hauser, K.F.; McRae, M. Independent actions by HIV-1 Tat and morphine to increase recruitment of monocyte-derived macrophages into the brain in a region-specific manner. Neurosci. Lett. 2022, 788, 136852. [Google Scholar] [CrossRef]
  129. Jaureguiberry-Bravo, M.; Lopez, L.; Berman, J.W. Frontline Science: Buprenorphine decreases CCL2-mediated migration of CD14+CD16+ monocytes. J. Leukoc. Biol. 2018, 104, 1049–1059. [Google Scholar] [CrossRef]
  130. Mahajan, S.D.; Aalinkeel, R.; Sykes, D.E.; Reynolds, J.L.; Bindukumar, B.; Fernandez, S.F.; Chawda, R.; Shanahan, T.C.; Schwartz, S.A. Tight Junction Regulation by Morphine and HIV-1 Tat Modulates Blood–Brain Barrier Permeability. J. Clin. Immunol. 2008, 28, 528–541. [Google Scholar] [CrossRef] [PubMed]
  131. Patel, S.H.; Ismaiel, O.A.; Mylott, W.R., Jr.; Yuan, M.; McClay, J.L.; Paris, J.J.; Hauser, K.F.; McRae, M. Cell-type specific differences in antiretroviral penetration and the effects of HIV-1 Tat and morphine among primary human brain endothelial cells, astrocytes, pericytes, and microglia. Neurosci. Lett. 2019, 712, 134475. [Google Scholar] [CrossRef] [PubMed]
  132. Hind, W.H.; England, T.J.; O’Sullivan, S.E. Cannabidiol protects an in vitro model of the blood-brain barrier from oxygen-glucose deprivation via PPARgamma and 5-HT1A receptors. Br. J. Pharmacol. 2016, 173, 815–825. [Google Scholar] [CrossRef] [PubMed]
  133. Stone, N.L.; England, T.J.; O’Sullivan, S.E. Protective Effects of Cannabidivarin and Cannabigerol on Cells of the Blood-Brain Barrier Under Ischemic Conditions. Cannabis Cannabinoid Res. 2021, 6, 315–326. [Google Scholar] [CrossRef] [PubMed]
  134. Panikashvili, D.; Shein, N.a.A.; Mechoulam, R.; Trembovler, V.; Kohen, R.; Alexandrovich, A.; Shohami, E. The endocannabinoid 2-AG protects the blood–brain barrier after closed head injury and inhibits mRNA expression of proinflammatory cytokines. Neurobiol. Dis. 2006, 22, 257–264. [Google Scholar] [CrossRef] [PubMed]
  135. Starr, A.; Jordan-Sciutto, K.L.; Mironets, E. Confound, Cause, or Cure: The Effect of Cannabinoids on HIV-Associated Neurological Sequelae. Viruses 2021, 13, 1242. [Google Scholar] [CrossRef] [PubMed]
  136. Persidsky, Y.; Fan, S.; Dykstra, H.; Reichenbach, N.L.; Rom, S.; Ramirez, S.H. Activation of Cannabinoid Type Two Receptors (CB2) Diminish Inflammatory Responses in Macrophages and Brain Endothelium. J. Neuroimmune Pharmacol. 2015, 10, 302–308. [Google Scholar] [CrossRef]
  137. Rom, S.; Zuluaga-Ramirez, V.; Dykstra, H.; Reichenbach, N.L.; Pacher, P.; Persidsky, Y. Selective activation of cannabinoid receptor 2 in leukocytes suppresses their engagement of the brain endothelium and protects the blood-brain barrier. Am. J. Pathol. 2013, 183, 1548–1558. [Google Scholar] [CrossRef]
  138. Ramirez, S.H.; Hasko, J.; Skuba, A.; Fan, S.; Dykstra, H.; McCormick, R.; Reichenbach, N.; Krizbai, I.; Mahadevan, A.; Zhang, M.; et al. Activation of cannabinoid receptor 2 attenuates leukocyte-endothelial cell interactions and blood-brain barrier dysfunction under inflammatory conditions. J. Neurosci. 2012, 32, 4004–4016. [Google Scholar] [CrossRef]
  139. Wang, Z.; Li, Y.; Cai, S.; Li, R.; Cao, G. Cannabinoid receptor 2 agonist attenuates blood-brain barrier damage in a rat model of intracerebral hemorrhage by activating the Rac1 pathway. Int. J. Mol. Med. 2018, 42, 2914–2922. [Google Scholar] [CrossRef]
  140. Fujii, M.; Sherchan, P.; Krafft, P.R.; Rolland, W.B.; Soejima, Y.; Zhang, J.H. Cannabinoid type 2 receptor stimulation attenuates brain edema by reducing cerebral leukocyte infiltration following subarachnoid hemorrhage in rats. J. Neurol. Sci. 2014, 342, 101–106. [Google Scholar] [CrossRef]
  141. Bullock, T.A.; Galpayage Dona, K.N.U.; Hale, J.F.; Morales, P.; Jagerovic, N.; Andrews, A.M.; Ramirez, S.H. Activation of CB2R by synthetic CB2R agonist, PM289, improves brain endothelial barrier properties, decreases inflammatory response and enhances endothelial repair. NeuroImmune Pharm. Ther. 2023, 2, 387–400. [Google Scholar] [CrossRef]
  142. Amenta, P.S.; Jallo, J.I.; Tuma, R.F.; Elliott, M.B. A cannabinoid type 2 receptor agonist attenuates blood-brain barrier damage and neurodegeneration in a murine model of traumatic brain injury. J. Neurosci. Res. 2012, 90, 2293–2305. [Google Scholar] [CrossRef] [PubMed]
  143. Ellis, R.J.; Peterson, S.N.; Li, Y.; Schrier, R.; Iudicello, J.; Letendre, S.; Morgan, E.; Tang, B.; Grant, I.; Cherner, M. Recent cannabis use in HIV is associated with reduced inflammatory markers in CSF and blood. Neurol. Neuroimmunol. Neuroinflamm. 2020, 7, e809. [Google Scholar] [CrossRef]
  144. Kim, H.J.; Shin, A.H.; Thayer, S.A. Activation of cannabinoid type 2 receptors inhibits HIV-1 envelope glycoprotein gp120-induced synapse loss. Mol. Pharmacol. 2011, 80, 357–366. [Google Scholar] [CrossRef]
  145. Ellis, R.J.; Peterson, S.; Cherner, M.; Morgan, E.; Schrier, R.; Tang, B.; Hoenigl, M.; Letendre, S.; Iudicello, J. Beneficial Effects of Cannabis on Blood–Brain Barrier Function in Human Immunodeficiency Virus. Clin. Infect. Dis. 2020, 73, 124–129. [Google Scholar] [CrossRef] [PubMed]
  146. Watson, C.W.-M.; Campbell, L.M.; Sun-Suslow, N.; Hong, S.; Umlauf, A.; Ellis, R.J.; Iudicello, J.E.; Letendre, S.; Marcotte, T.D.; Heaton, R.K.; et al. Daily Cannabis Use is Associated With Lower CNS Inflammation in People With HIV. J. Int. Neuropsychol. Soc. 2021, 27, 661–672. [Google Scholar] [CrossRef] [PubMed]
  147. Watson, C.W.; Sundermann, E.; Helm, J.; Paolillo, E.W.; Hong, S.; Ellis, R.J.; Letendre, S.; Marcotte, T.D.; Heaton, R.K.; Morgan, E.E.; et al. A longitudinal study of cannabis use and risk for cognitive and functional decline among older adults with HIV. AIDS Behav. 2023, 27, 3401–3413. [Google Scholar] [CrossRef]
  148. Swinton, M.K.; Sundermann, E.E.; Pedersen, L.; Nguyen, J.D.; Grelotti, D.J.; Taffe, M.A.; Iudicello, J.E.; Fields, J.A. Alterations in Brain Cannabinoid Receptor Levels Are Associated with HIV-Associated Neurocognitive Disorders in the ART Era: Implications for Therapeutic Strategies Targeting the Endocannabinoid System. Viruses 2021, 13, 1742. [Google Scholar] [CrossRef]
  149. Cosenza-Nashat, M.A.; Bauman, A.; Zhao, M.L.; Morgello, S.; Suh, H.S.; Lee, S.C. Cannabinoid receptor expression in HIV encephalitis and HIV-associated neuropathologic comorbidities. Neuropathol. Appl. Neurobiol. 2011, 37, 464–483. [Google Scholar] [CrossRef]
  150. Jones, L.D.; Jackson, J.W.; Maggirwar, S.B. Modeling HIV-1 Induced Neuroinflammation in Mice: Role of Platelets in Mediating Blood-Brain Barrier Dysfunction. PLoS ONE 2016, 11, e0151702. [Google Scholar] [CrossRef]
  151. Haorah, J.; Knipe, B.; Gorantla, S.; Zheng, J.; Persidsky, Y. Alcohol-induced blood–brain barrier dysfunction is mediated via inositol 1,4,5-triphosphate receptor (IP3R)-gated intracellular calcium release. J. Neurochem. 2007, 100, 324–336. [Google Scholar] [CrossRef]
  152. Muneer, P.M.A.; Alikunju, S.; Szlachetka, A.M.; Haorah, J. The Mechanisms of Cerebral Vascular Dysfunction and Neuroinflammation by MMP-Mediated Degradation of VEGFR-2 in Alcohol Ingestion. Arterioscler. Thromb. Vasc. Biol. 2012, 32, 1167–1177. [Google Scholar] [CrossRef] [PubMed]
  153. Flora, G.; Pu, H.; Lee, Y.W.; Ravikumar, R.; Nath, A.; Hennig, B.; Toborek, M. Proinflammatory synergism of ethanol and HIV-1 Tat protein in brain tissue. Exp. Neurol. 2005, 191, 2–12. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic illustration of HAND neuropathogenesis. Following contact with the brain endothelium, circulating HIV-infected leukocytes, virions, and viral proteins (e.g., gp120, Tat, Nef) induce blood-brain barrier (BBB) injury, associated with increased inflammation and production of reactive oxygen species (ROS), damage to endothelial tight junction (TJ) proteins (occludin, claudins, zonula occludens), and damage to adherent junctions (junctional adhesion molecules (JAMs)) proteins. This results in BBB dysfunction, including increased permeability and infiltration of HIV and infected cells into the central nervous system (CNS). Once in the brain, HIV infects resident brain cells, including microglia (productive infection) and astrocytes. Productively infected brain cells further release HIV and viral proteins, inflammatory cytokines, chemokines, and ROS. These factors further increase BBB injury and dysfunction, CNS infection, and induce neuronal injury. Repeated and prolonged neuronal injury causes neurodegeneration, which can result in HIV-associated neurocognitive disorders (HAND). Created with BioRender.com (accessed on 14 June 2024).
Figure 1. Schematic illustration of HAND neuropathogenesis. Following contact with the brain endothelium, circulating HIV-infected leukocytes, virions, and viral proteins (e.g., gp120, Tat, Nef) induce blood-brain barrier (BBB) injury, associated with increased inflammation and production of reactive oxygen species (ROS), damage to endothelial tight junction (TJ) proteins (occludin, claudins, zonula occludens), and damage to adherent junctions (junctional adhesion molecules (JAMs)) proteins. This results in BBB dysfunction, including increased permeability and infiltration of HIV and infected cells into the central nervous system (CNS). Once in the brain, HIV infects resident brain cells, including microglia (productive infection) and astrocytes. Productively infected brain cells further release HIV and viral proteins, inflammatory cytokines, chemokines, and ROS. These factors further increase BBB injury and dysfunction, CNS infection, and induce neuronal injury. Repeated and prolonged neuronal injury causes neurodegeneration, which can result in HIV-associated neurocognitive disorders (HAND). Created with BioRender.com (accessed on 14 June 2024).
Cells 13 01415 g001
Table 1. In vivo effects of HIV-1, viral proteins, and substances of abuse on the BBB integrity and function, and associated mechanisms.
Table 1. In vivo effects of HIV-1, viral proteins, and substances of abuse on the BBB integrity and function, and associated mechanisms.
Effects on the BBB Structure and FunctionMechanisms
HIVDownregulation of claudin-5, occludin, ZO-1, and ZO-2, [30,36,45,62,150].
Increased BBB permeability, and infiltration of leukocytes into the brain [36,45,62,150].
Altered expression of ICAM-1 and VCAM-1 [34,150].
Increased levels of C-C chemokines, IFN-γ, CCL5, MIP-1α, MIP-1β, MCP-3, and IP-10 [35].
HIV gp120Decreased expression of occludin and ZO-1 [81].
Increased BBB permeability [38,81].
Increased ICAM-1, VCAM-1, substance-P, TIMP-1, TIMP-2, MMP-2, and MMP-9 [38,40].
Increased ROS, MDA and protein carbonyl; decreased GSH and GPx activity [40,81].
HIV TatDecreased expression of claudin-5, and ZO-1 [41,54].
Disrupted BBB integrity; increased recruitment of activated, phagocytic, and perivascular macrophages into the CNS [42].
Upregulation of ICAM-1, VCAM-1, and AKT activation [88]; autophagy [41].
HIV NefDownregulation of claudin-5 [44].
Induced BBB disruption and increased BBB permeability [43,44].
Upregulated MMP-9 [43] and IL-1β [44].
Cocaine and HIV/viral proteinsCocaine downregulated ZO-1 [101].
Cocaine and HIV-1 increased monocyte adhesion and transmigration [101].
Cocaine induced PDGF-β [101].
Cocaine and HIV-1 increased ALCAM [101] and cytokines [106].
Meth and HIV/viral proteinsMeth and HIV or Tat induced downregulation of ZO-1, occludin, claudin-5, and JAM-A [111,114,115].
Meth and HIV or Tat increased BBB permeability [111,114].
Meth and HIV increased MMP-9 [114].
Meth and Tat downregulated GLUT-1 and GLUT-3 [115]; increased NF-κB [114]; induced ROS, reduced CAT, GPx, and SOD activity; and increased MDA [111].
Alcohol and HIV/viral proteinsAlcohol altered occludin, claudin-5, and ZO-1, and phosphorylated ZO-1 [151,152].
Alcohol increased BBB permeability and transmigration of immune cells into the brain [151,152].
Alcohol activated MMP-3 and MMP-9 [152].
Alcohol and Tat downregulated TRPM7 [116]; increased ICAM-1, IL-1β, MCP-1, CREB, and NF-κB DNA-binding activity; activated ERK1/2 [153].
Tobacco and gp120/antiretroviralsNicotine and SQV or SQV/r decreased ZO-1 and increased BBB permeability [119].Nicotine and SQV or SQV/r decreased Notch-4 and increased ROS [119].
MOR and TatMOR or fentanyl and Tat decreased ZO-1, claudin-5, and increased P-gp [126,128].
MOR or fentanyl and Tat increased BBB disruption and permeability [126,128].
Fentanyl and Tat dysregulated inflammatory cytokines [126].
Table 2. In vitro effects of HIV-1, viral proteins, and substances of abuse on the BBB integrity and function, and associated mechanisms.
Table 2. In vitro effects of HIV-1, viral proteins, and substances of abuse on the BBB integrity and function, and associated mechanisms.
Effects on the BBB Structure and FunctionMechanisms
HIVDownregulation of ZO-1, ZO-2, claudin-1, claudin-5, occludin, and JAM-A [46,64,74]; phosphorylation of occludin and claudin-5 [68]; reduced TEER [68].
Increased BBB permeability, leukocyte adhesion and transmigration [26,46,47,64,68,74].
Increased MMP-2 and MMP-9 [64,73,74]; altered ICAM-1, and E-selectin [26,75]; increased IL-6, IL-8, IL-1β, TNF-α, MCP-1, and IFN-inducible genes; activation of SP-1, AP-1, NF-κB, STAT-1, and STAT-3 [26,46,47,73,75].
Increased ROS and redox proteins (peroxiredoxin, SOD); reduced p-eNOS, p-DRP1, and mitochondrial membrane potential [67,69,73].
Activation of Rac1, RhoA GTPases, Rho-kinase, PI3K, PDK1, ERK1/2, and AKT [47,68,73,76].
HIV gp120Decreased expression of ZO-1, ZO-2, and occludin [49].
Increased BBB permeability, monocyte adhesion, and transendothelial migration [37,49,51].
Increased IL-6, IL-8, ROS, and MDA; decreased activity of GSH, GPx, GR, and CAT [51,82,83].
Activation of STAT-1, p38 MAPK, MEK, and PI3K pathways [51,77]; increased caspase-3 and cytotoxicity [77].
HIV TatDecreased expression of claudin-1, claudin-5, ZO-1, ZO-2, occludin, and TEER; increased P-gp, disruption and phosphorylation of VE-cadherin and β-catenin [41,53,54,55,56,57,58,89,90,92,95].
Increased BBB permeability, leukocyte adhesion and transendothelial migration [52,56,86,87,89,90].
Increased E-selectin, ICAM-1 and VCAM-1 [87,88]; MMP-9 [56]; IL-6, PAI-1, MCP-1, and AP-1 [53,85,86,87,91].
Decreased GSH and increased ROS, ER stress, mitochondrial dysfunction, activated UPR, upregulated NOX-2 and NOX-4 [45,89,91,95].
Induced apoptosis, autophagy, and cytotoxicity [41,45,93,95]. Upregulated VEGFR-2 and activated redox-regulated pathways [92,93].
Increased NF-κB binding activity and activation of NF-κB, ERK1/2, IRAK-1/4, MKK JNK, AP-1, PI3K, AKT, FAK, RhoA, ROCK, Ras, PKA, PKC, and PYK2 pathways [52,53,55,56,58,85,87,89,92,93,95].
HIV NefDecreased ZO-1, TEER, and altered BBB permeability [60].Increased IL-12, IL-8, IL-6, CCL5, and IL-17A [60].
Induced PARP cleavage, upregulated Fas/FasL, activation of caspases and mitochondrial apoptotic pathways [59].
Cocaine and HIV/viral proteinsCocaine, or cocaine and HIV-1 or Tat, decreased ZO-1, JAM-2, and TEER, and increased stress fiber formation [100,102].
Cocaine or cocaine and HIV-1 or Tat increased BBB permeability, monocyte adhesion and transendothelial migration [101,102,104,105].
Cocaine induced activation of ERK1/2, p38 MAPK, JNK, and PI3K/AKT pathways [101].
Cocaine or cocaine and HIV-1 upregulated ICAM-1, VCAM-1, PECAM-1, ELAM-1, and ALCAM [101,104,105].
Cocaine or cocaine and Tat increased inflammatory cytokines, MCP-1 and its receptor in monocytes; increased secretion of TNF-α and IL-6, and activation of NF-κB [100,101,105].
Meth and HIV/viral proteinsMeth, or Meth and gp120 or Tat, decreased ZO-1, JAM-A, occludin, claudin-5, claudin-3, JAM-2, and TEER; and impaired P-gp function [110,111,112,113,115].
Meth or Meth and gp120 or Tat induced BBB disruption, increased BBB permeability and leukocyte transmigration [111,113].
Meth and Tat reduced GLUT-1 and GLUT-3 [115].
Meth or Meth and Tat
increased ROS and MDA, reduced CAT, GPx, and SOD activity [111,112]; decreased viability and induced apoptosis in ECs [111,112,115].
Meth and gp120 activate Rho-A GTPase [113].
Alcohol and HIV/viral proteinsAlcohol, alcohol plus gp120 or Tat, decreased claudin-3, claudin-5, occludin, JAM-1, ZO-1, and induced stress fiber formation [116,117,118].
Alcohol and gp120 or Tat increased BBB permeability and macrophage transmigration [117,118].
Alcohol and gp120 or Tat downregulated TRPM7 [116], increased ECs ROS and NO [117,118].
Ethanol and Nef, Tat, gp120, or Vpr increased ECs LDH, TNF-α, and apoptosis [96].
Tobacco and gp120/antiretroviralsNicotine and SQV or SQV/r decreased ZO-1 [119]. Tobacco smoke extracts and gp120 decreased occludin, ZO-1, and TEER [121].
Tobacco and SQV or gp120, or tobacco smoke extracts and gp120, increased BBB permeability [119,121].
Nicotine and SQV or SQV/r increased ROS and downregulated Notch-4 [119].
Tobacco smoke extracts and gp120 increased ROS, NF-κB, disrupted mitochondrial function, decreased NRF-2 and EC viability [121].
MOR and TatMOR and/or Tat decreased ZO-1, JAM-2, occludin, TEER, and increased P-gp [130].
MOR and/or Tat increased BBB permeability and leukocytes’ transendothelial migration [130].
MOR and/or Tat activated MLCK, increased pro-inflammatory cytokines, and intracellular calcium release [130].
Table 3. Interactions between substances of abuse and HIV, viral proteins, and antiretroviral drugs on the BBB integrity and function.
Table 3. Interactions between substances of abuse and HIV, viral proteins, and antiretroviral drugs on the BBB integrity and function.
InteractionEffects on the BBB
Cocaine + HIV AdditiveUpregulation of ALCAM [101]
Cocaine + TatSynergisticDecreased ZO-1, JAM-2, and TEER, increased BBB permeability, and monocyte transmigration across BBB [102].
Meth + TatSynergisticDecreased ZO-1, occludin, and JAM-A [110,111,115].
Decreased CAT, SOD, and GPx, induced oxidative stress, and increased TRPM2 [111].
Decreased P-gp [110]; decreased claudin-5, GLUT-1, GLUT-3, and cell viability [115].
Meth + gp120SynergisticDecreased ZO-1, JAM-2, claudin-3, claudin-5, and TEER; increased leukocytes’ transmigration across the BBB [113].
Meth + gp120 or Tat SynergisticDecreased ZO-1, occludin, glutathione, and GPx, increased ROS, protein carbonyls, lipid peroxidation [81].
Ethanol + gp120 SynergisticDecreased claudin-3, occludin, ZO-1, JAM-2, and TRPM7 [116].
Ethanol + Tat SynergisticIncreased macrophage migration across BBB [117].
Nicotine + SQR/r AdditiveDecreased Notch-4, and ZO-1, increased ROS, and BBB disruption [119].
Tobacco smoke extracts + gp120 SynergisticDecreased ZO-1, occludin, and NRF-2; increased ROS, NF-κB, and BBB permeability; decreased TEER, and cell viability [121].
Morphine + Tat SynergisticReduced TEER; increased JAM-2, P-gp, and migration of non-infected and HIV-1 infected leukocytes across the BBB [130].
Fentanyl + Tat AdditiveDecreased claudin-5 [126].
Abbreviations (Table 1, Table 2 and Table 3): ALCAM: activated leukocyte cell adhesion molecule; AP-1: activator protein-1; BBB: blood–brain barrier; CAT: catalase; CCL5: chemokine ligand-5; CNS: central nervous system; CREB: cAMP response element-binding protein; ECs: endothelial cells; ELAM-1: endothelial leucocyte adhesion molecule-1; ER: endoplasmic reticulum; ERK: extracellular signal regulated kinase; FAK: focal adhesion kinase; gp120: glycoprotein 120; GLUT: glucose transporter; GPx: glutathione peroxidase; GR: glutathione reductase; GSH: glutathione; ICAM-1: intercellular adhesion molecule-1; IFN: interferon; IP-10: IFN-gamma-inducible protein-10; IL: interleukin; IRAK-1/4: IL-1 receptor-associated kinase-1/4; JAM: junctional adhesion molecule; JNK: C-jun N-terminal kinase; LDH: lactate dehydrogenase; MAPK: mitogen-activated protein kinase; MCP-1: monocyte chemoattractant protein-1/CCL2; MCP-3: monocyte chemoattractant protein-3/CCL7; MDA: malondialdehyde; MOR: morphine; MEK: mitogen-activated protein kinase; MIP: macrophage inflammatory protein; MLCK: myosin light chain kinase; MKK: mitogen-activated protein kinase kinase; MMP: matrix metalloproteinase; NO: nitric oxide; NOTCH-4: neurogenic locus notch homolog 4; NOX: NADPH oxidase; NRF2: Nuclear factor erythroid 2-related factor 2; PAI-1: plasminogen activator inhibitor-1; PARP: poly (ADP-ribose) polymerase; P-gp: P-glycoprotein; PDGF-β: platelet-derived growth factor β; PDK1: phosphoinositide-dependent kinase-1; PECAM-1: platelet endothelial cell adhesion molecule-1; PI3K: phosphoinositide 3-kinase; PKA: protein kinase-A; PKC: protein kinase-C; p-eNOS: phosphorylated endothelial nitric oxide synthase; p-DRP1: phosphorylated dynamin-related protein-1; PYK2: Protein tyrosine kinase-2 beta; ROCK: Rho-associated protein kinase; ROS: reactive oxygen species; SOD: superoxide dismutase; SQV/r: saquinavir/ritonavir; SP-1: specificity protein-1 transcription factor; STAT: signal transducer and activator of transcription; Tat: Trans-activator of transcription; TEER: transendothelial electrical resistance; TIMP: tissue inhibitor of metalloproteinase; TNF: tumor necrosis factor; TRPM: transient receptor potential melastin; UPR: unfolded protein response; VCAM-1: vascular cell adhesion molecule-1; VEGFR-2: vascular endothelial growth factor receptor-2; ZO: zonula occludens.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Annadurai, N.; Kanmogne, G.D. Structural and Functional Dysregulation of the Brain Endothelium in HIV Infection and Substance Abuse. Cells 2024, 13, 1415. https://doi.org/10.3390/cells13171415

AMA Style

Annadurai N, Kanmogne GD. Structural and Functional Dysregulation of the Brain Endothelium in HIV Infection and Substance Abuse. Cells. 2024; 13(17):1415. https://doi.org/10.3390/cells13171415

Chicago/Turabian Style

Annadurai, Narendran, and Georgette D. Kanmogne. 2024. "Structural and Functional Dysregulation of the Brain Endothelium in HIV Infection and Substance Abuse" Cells 13, no. 17: 1415. https://doi.org/10.3390/cells13171415

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Article metric data becomes available approximately 24 hours after publication online.
Back to TopTop