Skip to main content
Open Biology logoLink to Open Biology
. 2020 Dec 23;10(12):200286. doi: 10.1098/rsob.200286

Alzheimer's disease-like perturbations in HIV-mediated neuronal dysfunctions: understanding mechanisms and developing therapeutic strategies

Niraj Kumar Jha 1,, Ankur Sharma 2, Saurabh Kumar Jha 1, Shreesh Ojha 3, Dinesh Kumar Chellappan 4, Gaurav Gupta 5, Kavindra Kumar Kesari 6, Shanu Bhardwaj 7, Shakti D Shukla 8, Murtaza M Tambuwala 9, Janne Ruokolainen 6, Kamal Dua 8,10,11, Sandeep Kumar Singh 12,13,
PMCID: PMC7776571  PMID: 33352062

Abstract

Excessive exposure to toxic substances or chemicals in the environment and various pathogens, including viruses and bacteria, is associated with the onset of numerous brain abnormalities. Among them, pathogens, specifically viruses, elicit persistent inflammation that plays a major role in Alzheimer's disease (AD) as well as dementia. AD is the most common brain disorder that affects thought, speech, memory and ability to execute daily routines. It is also manifested by progressive synaptic impairment and neurodegeneration, which eventually leads to dementia following the accumulation of Aβ and hyperphosphorylated Tau. Numerous factors contribute to the pathogenesis of AD, including neuroinflammation associated with pathogens, and specifically viruses. The human immunodeficiency virus (HIV) is often linked with HIV-associated neurocognitive disorders (HAND) following permeation through the blood–brain barrier (BBB) and induction of persistent neuroinflammation. Further, HIV infections also exhibited the ability to modulate numerous AD-associated factors such as BBB regulators, members of stress-related pathways as well as the amyloid and Tau pathways that lead to the formation of amyloid plaques or neurofibrillary tangles accumulation. Studies regarding the role of HIV in HAND and AD are still in infancy, and potential link or mechanism between both is not yet established. Thus, in the present article, we attempt to discuss various molecular mechanisms that contribute to the basic understanding of the role of HIV-associated neuroinflammation in AD and HAND. Further, using numerous growth factors and drugs, we also present possible therapeutic strategies to curb the neuroinflammatory changes and its associated sequels.

Keywords: HIV, Alzheimer's disease, HAND, microglia, neuroinflammation, neurotherapeutics

1. Introduction

Alzheimer's disease (AD) is the most common neurological complication, which mainly manifests progressive synaptic impairment and neurodegeneration, following excessive formation and accumulation of amyloid-beta (Aβ) [1,2]. Aβ deposits and hyperphosphorylated Tau (pTau), which interfere with the neuronal organization and their function, play a considerable role in AD progression [3]. Aβ-pathology often involves a variety of signals that interrupt the homeostasis of neurons [4]. Currently, there are no definite data that can demonstrate a causative relationship between neuronal damage following human immunodeficiency virus (HIV) infections and the onset of AD. However, available literature indicates that there are some common factors and pathways modulated in HIV+ and AD patients, thus suggestive of some similarities in these two pathologies. Among numerous pathways, neuroinflammation is shown closely related to these disorders and is considered a crucial factor in their development and progression. It has been reported that HIV regulatory proteins such as trans-activator of transcription (Tat), envelope glycoprotein (Gp120), viral protein R (Vpr) and negative factor (Nef) can directly influence the central nervous system (CNS) and activate neuroinflammatory pathways followed by neuronal injury and dysfunction. Additionally, abnormal Aβ deposition, a pathological hallmark of AD has been reported in the individuals suffering from HIV infection. Though the abnormalities associated with Aβ burden are more frequent in the AD brain than HIV-infected individuals, it has been predominantly observed in younger HIV-infected individuals [5,6].

Additionally, blood–brain barrier (BBB) dysfunction associated with HIV-1 infection is considered another cause of neuroinflammation in AD. HIV infiltrates macrophages in the CNS by crossing the BBB. The disrupted BBB in HIV patients has been correlated with toxic Aβ aggregation and other abnormalities resulting from a failure to sort out Aβ peptides [7]. The virus-induced fusion of macrophages causes the formation of giant cells and activation of astrocytes which eventually causes injuries to different components of the brain. The most affected areas are the subcortical structures along with the limbic structures and basal ganglia, and the verotoxins, including HIV proteins Gp41, Gp120, Tat, Vpr and Nef, are accountable for such damage. HIV proteins also may cause axonal damage and breakdown of white matter. These injuries cause a decrease in volume of the brain structures such as the caudate nucleus and basal ganglia, resulting in atrophy of the brain volume and decline in cognition [810].

HIV also leads to HIV-associated neurocognitive disorders (HAND), since it has a propensity to cross the BBB and cause neuroinflammation [1114]. HAND exhibits a spectrum of cognitive deficits and typically affects information processing speed, attention, learning and recall memory among other cognitive functions [15]. HAND also has implications for adherence to antiretroviral (ARV) treatment since it affects prospective memory [16]. The exact route in which HIV causes HAND is not yet well known, although HIV replication (potential mechanisms) in the CNS, principally in the basal ganglia and the adjacent subcortical white matter, is where HIV infection is typically observed [17,18].

Among different cell types in the CNS, neurons have the minimal susceptibility to HIV infection; thus the neuronal impairment is reasonably speculated to result from an infection of neighbouring cells like microglia and macrophages, which exert immune functions in the brain. These infected cells result in the production of viral proteins that have the ability to affect the synapse where communication between neurons occurs. Also, the same viral proteins can induce uninfected macrophages, astrocytes and microglial cells that results in the production of neurotoxins and a variety of inflammatory molecules, causing further damage to neurons [19]. Further, the inflammatory molecules and neurotoxins trigger NMDA receptors and may cause additional damage to the neurons following aggregation of calcium (Ca2+) in the neurons which activate the formation of excessive free radicals that contribute to oxidative damage. Among other factors, methamphetamine use or abuse and co-infection with hepatitis C virus (HCV) may aggravate damage caused by HIV, involving activation of macrophages and microglial cells [19].

Like amyloid plaques, neurofibrillary tangles (NFTs) consisting of pTau also occur in people suffering from HIV, particularly in aged individuals [20]. The elevated levels of Tau have been reported to occur at earlier ages in individuals suffering from HIV than in healthy individuals. In HIV-infected individuals, tau phosphorylation results from viral proteins and pro-inflammatory cytokines that may impair amyloidosis and precede the development of tau tangles [21]. Higher expression of pTau in HIV individuals is also correlated with ARV treatment [20]. Many comorbid conditions like chronic substance abuse independent of the direct consequences of HIV also lead to HIV transmission, responsiveness and cognitive difficulties [22]. It is apparent that the linkage and causative mechanisms between neuroinflammation, HIV-CNS neuroinfections, HAND and AD are still not completely understood. Therefore, it is important to understand the fundamental molecular linkage among these pathologies, which may help in understanding pathogenesis and developing therapeutics targeting the pathogenesis events along with additional help in diagnosis and prognosis. In the purview of this, herein we summarize various underlying mechanisms which contribute to HIV-associated neuroinflammation in HAND and AD using synoptic tables and schemes. Additionally, numerous possible therapeutic strategies are also presented, which may have the potential to curb these complications and improve quality of life.

2. Human cells involved in HIV-associated neuronal damage

HIV-1 interacts with different cell types (table 1) in the CNS, including resident macrophages, neurons and astrocytes that are reported to be involved in neuronal damage [12,26,27]. In the CNS, resident macrophages, neurons and astrocytes are the primary cell targets for HIV infection. In neurodegenerative processes, the roles of macrophages are crucial due to their resistance and sustenance against the cytopathic effects of HIV-1 [19,2833]. In the CNS, there are four major types of macrophages: choroid-plexus macrophages, meningeal macrophages, perivascular macrophages and microglia [23,34]. Out of these, perivascular macrophages and microglia are believed to play a crucial role in neuronal damage following the release of inflammatory cytokines [23]. Additionally, viral proteins and neurotoxins also take part in the inflammatory processes, provoking apoptosis and differentiation of astrocytes, and impairing normal neurogenesis [12,24,25]. Further, microglial resident cells play a fundamental role in the pathogenesis of HAND, leading to degenerative changes involving numerous mechanisms. The glial cells upon HIV infection release factors and toxins that aggravate neurons and astrocytes [12,35,36]. Astrocytes are neuroectodermal-derived cells, which support the function and metabolism of neurons, ionic homeostasis into the CNS, control of the state of the neuronal synapses by the uptake of neurotransmitters and tissue repair. These are the important components of the BBB and also regulate the immune responses in the brain [3739]. In addition, astrocytes can facilitate the virus to persist in the CNS, which aids in maintaining low replication of HIV and establishing a latent infection [40]. Furthermore, in HIV-infected cells, viral factors may enhance the release of other chemoattractants that recruit microglia and monocytes, resulting in aggravation of the neuronal damage. Further, cellular factors like interleukin-1β (IL-1β), interferon gamma (IFN-γ) or tumour necrosis factor alpha (TNF-α) have the potential to activate and reactivate viral replication in latently infected cells [19,4145].

Table 1.

The role of human cells in HIV-mediated neuronal damage [12,2325].

neuronal cell associated effects types of infection
neuron enhances P53 expression restricted
enhances caspase activation
enhances intracellular Ca2+ release
microglia induces viral replication productive
provokes the release of viral proteins including, gp120, Tat and Vpr
increases neurotoxins production and also induces the expression of inflammatory mediators, such as PDGF and QUIN
astrocyte enhances the production of neurotoxins restricted
downregulates the glutamate uptake
enhances BBB permeability
enhances intracellular release of glutamate and Ca2+
evokes the migration of monocytes into the brain
perivascular macrophage triggers viral replication productive
increases neurotoxins production and induces the expression of inflammatory mediators, such as PDGF and QUIN
provokes the release of viral proteins including gp120, Tat and Vpr
oligodendrocyte enhances cellular apoptosis restricted
enhances intracellular Ca2+ levels
curtails myelin synthesis

3. The direct and indirect mechanisms of HIV induced-neuronal injury

3.1. Direct mechanisms

HIV-1 infects CNS involving three different mechanisms (figure 1). In the first mechanism, the virus can directly infect endothelial cells which express the chemokine receptors (CCR3, CXCR4, DC-SIGN) engaged in HIV-1 entry [40,46]. In the second mechanism, the virus may directly cross the impaired BBB due to increased permeability [45,47]. In the third mechanism, according to the ‘Trojan horse’ hypothesis, HIV-1 infected monocytes, perivascular macrophages and leucocytes cross the BBB and release viral particles, which infect resident cells like microglia and lead to persistent infection. This one is believed to be the main mechanism for entry of HIV into the brain, similar to other retroviruses and lentiviruses [40].

Figure 1.

Figure 1.

Schematic showing the entry mechanisms of HIV-1 into the CNS and its associated effects on neuronal cells that contribute to neuronal damage and death. (1) HIV-1 can enter through infected T-cells or monocytes that migrate from the bloodstream to the CNS according to the ‘Trojan horse’ hypothesis. (2) The increase in viral proteins and pro-inflammatory cytokines can impair the BBB (epithelial cells) permeability to make virus entry easier. Besides, using infected epithelial cells, virus can reach the other side through a transcytosis process. (3) Reactive astrocytes can provoke epithelial cell apoptosis, leading to the modification of BBB permeability through the release of viral proteins such as Tat. (4) The viral protein Tat has a direct effect on neurons and oligodendrocytes, which cause increased damage and neuronal death. Finally, chronic activation of activated (5) microglia and (6) macrophages causes an increase in the levels of neurotoxins, proinflammatory cytokines, RNA and ROS.

Several observations advocate that cells like monocytes are infected before leaving the bone marrow [48]. Particularly, proviral DNA has been observed in these cells with no presence of viral proteins, which facilitated dissemination of the HIV-1 infection [48,49]. An increase in a subset of monocytes, including (CD14lowCD16high), plays a significant role in HIV-1 infection [34,5054]. These cells display intermediate traits between the differentiated cells (dendritic cells and macrophage) and monocytes [51,53]. Owing to the lower activity of the host restriction factors than the CD14highCD16low cells, the cells are more liable to HIV replication following eased permeation through BBB [4952,54]. Furthermore, viral proteins released into the CNS are believed to induce BBB impairment by enhancing apoptosis and promoting the invasion of HIV as well as other viruses in the different components of the brain [45,5557].

3.2. The indirect mechanisms

In addition to direct mechanisms, HIV-associated neurological complications and neuroinflammation also involve indirect mechanisms such as the infiltration of infected monocytes and lymphocytes in the CNS, release of viral and cellular factors from these infected cells, and infection of the resident cells caused by viral particles released from infected cells or infiltrating into the CNS [58]. The cells (specifically T-cells and monocytes) infected with HIV play a crucial role in the release of pro-inflammatory cytokines that stimulate microglia and astrocytes. The activated microglia and astrocytes along with perivascular macrophages are engaged in releasing inflammatory and neurotoxic mediators, including quinolinic acid (QUIN), nitrogen oxide and platelet-derived growth factor (PDGF), that further lead to neuronal dysfunction and death [45,59].

Despite treatment with ARV agents, a previous study has reported that the level of cytokines such as CCL3, IL-8, CCL2, IFN-γ, CXCL10 and IL-6 was found to be higher in HIV-1 infected individuals in comparison with the uninfected individuals. The higher expressions of cytokines are indicative of uninterrupted neuroinflammation that is accountable for promoting HAND-associated encephalopathy [60]. Recently, Vera et al. [61] reported the presence of neuroinflammatory markers in neuro-asymptomatic HIV-infected patients, despite the effective control of viraemia. The translocation of the virus from the gut to the bloodstream is believed to cause extensive inflammation and altered integrity of white matter, and this reasonably suggests the role of the brain–gut axis in the pathogenesis of HAND [62].

4. Detailed mechanisms of neuroinflammation caused by HIV in the brain

HIV is known to play a key role in depleting cluster of differentiation 4 (CD4) cells, and robustly hampering the immune responses. Subsequently, it may rise to opportunistic infections and cause acquired immunodeficiency syndrome (AIDS). HIV is occasionally known as a neurotropic virus, although lacking expression of its main receptor CD4 in neurons; it cannot directly damage the neuronal tissues [63]. Nevertheless, recent phylogenetic analyses showed that HIV could easily access the CNS during primary infection (within the first two weeks), where it can replicate locally and get compartmentalized [64]. Thereafter, virus replication leads to neurotoxicity that is correlated with impaired sensory, cognitive and motor function in patients suffering from HIV, and these neuronal abnormalities are collectively termed HAND [1114]. These conditions are further categorized into three groups, based on the severity of the symptoms, namely, HIV-associated dementia (HAD), mild neurocognitive disorder (MND) and asymptomatic neurocognitive impairment (ANI) [15]. Patients suffering from these complications exhibit an array of clinical symptoms which may range from cognitive and motor impairment to altered mood and behavioural changes to dementia. The asymptomatic, ANI-HIV+ patients have been reported to display greater risk to develop cognitive dysfunctions in comparison with normal patients, and these are considered to reflect the primary stages of AD [65,66]. The incidences of HAND have been found to reduce with the successful establishment of combination antiretroviral therapy (cART) [12,67]. However, despite the availability of cART, the occurrence of HAND is drastically increasing nowadays, generally due to cardiovascular risk factors, increased life expectancy of patients, exposure to environmental hazards and neuroinflammatory changes. Recently, it has been reported that patients diagnosed with HAND with mild/ severe cognitive loss suffer from low quality of life, along with relatively shorter lifespan [68]. Before the introduction of cART, HAD was reported in 15–20% of HIV+ patients and was considered a focal risk factor [69,70]. However, following the establishment of this therapy, the total fraction of HAND patients did not show any discrepancy, but the distribution of the classes show alteration with an increase in MND and ANI and a decrease in HAD [12]. Evidence from recent studies shows that neuronal manifestations are becoming more common in the ageing HIV+ population [14,71,72]. The data from many clinical trials show poor prediction on the influence of cART on cognitive dysfunction due to BBB restricted lower penetration of the drugs into the CNS. Additionally, some ARV medicines can cause neurotoxicity and are believed to be linked with a poor prediction on the influence of cART on cognitive impairment. Given the available scenario, HAD is also considered as one of the most common forms of dementia in people of less than 40 years of age [14,71,72].

As described previously, HIV uses a mechanism called a ‘Trojan horse’ to enter the CNS, and this mechanism consists of the passage of infected monocytes through the BBB (figure 1) [5,73]. Recently, it has been shown in several clinical studies that CD14+CD16+ monocytes are competent to easily transmigrate through the BBB, and their high numbers are also reported in HIV-infected patients [5,73]. HIV, once it enters the brain, can damage many cell types, including perivascular macrophages, microglia and potentially adult neural precursors due to the presence of CD4 receptor on these cells [74,75]. Moreover, HIV replication can also be seen in astrocytes in a restrictive manner [76]. Due to these reasons, the brain is sometimes classified as a sanctuary and may serve as a reservoir for HIV [77]. The direct and indirect influences of HIV infection in the brain cause astrocytes and microglia-induced release of cytokines, chemokines and free radicals that result in neuronal dysfunction [12]. In addition, BBB disruption caused by HIV also contributes to further entry/exit of viral proteins and virions.

Numerous HIV regulatory proteins including Tat, Gp120, Vpr and Nef can have direct influences on the nervous system, and these viral proteins are accountable for triggering neuroinflammatory pathways that cause neuronal dysfunction (table 2 and figure 2). The main source of these viral proteins can be infected non-neuronal cells, although these also shed from virions [91,92]. Some viral proteins such as Vpr and Tat are consistently found in the cerebrospinal fluid (CSF) [91,93,94]. Further, the envelope protein Gp120 has been demonstrated to trigger the release of TNF-α and IL-1β, as well as glutamate, which elicits neuronal apoptosis, as evidenced by numerous ex vivo and in vivo studies [95,96]. Similarly, Tat has been found to potentiate glutamate overactivation of N-methyl-d-aspartate receptor (NMDA) receptors and release of cytokines from astrocytes, and potentiate neuronal apoptosis as well [9799]. Interestingly, Tat and Gp120-induced apoptosis also accounts for higher Ca2+ levels when coupled with excitotoxicity events and activated by glutamate deposition in the extracellular spaces. Patients suffering from HIV often have increased levels of glutamate in the CSF, and this correlates well with both the extent of brain atrophy and severity of dementia [100]. Similar to Tat and Gp120, the protein Nef can also trigger cytotoxic effects, though the exact mechanism played by this protein is yet to be investigated [101].

Table 2.

The roles of HIV regulatory proteins on neuronal damage.

HIV regulatory protein pathological implications on brain references
Tat induces the expression of GAC, GFAP, IL-1β and MCP-1/CCL2 [7881]
regulates cellular gene expression [82]
enhances the expression of GLUT1 in the hippocampus and cortex; also, enhances leucocyte infiltration [78,82]
upregulates the expression of Cx43 human gene [83]
decreases SYN expression; also reduces GABA in the cortex. [82,84]
interacts with CDK9 and Cyclin T1 [85]
Gp120 activates the release of inflammatory cytokines and toxic substances and accumulation of AβPP [86,87]
decreases the expression of MAP2, LC3 and beclin-1 [88]
Vpr promotes pro-apoptotic and cell-cycle proteins [57]
induces the release of matrix metalloproteinases (neurotoxins) [57]
provokes the release of IL-1β, TNF-α and IL-8 in macrophages [57]
Nef enhances the apoptosis of MVEC; also, enhances the sensitivity of astrocytes to H2O2 [55,89]
provokes astrogliosis and astroglial activation [90]

Figure 2.

Figure 2.

The scheme shows pathological implications of HIV regulatory proteins in neuronal damage. MVEC, microvascular endothelial cells; SYN, synaptophysin; GABA, gamma-aminobutyric acid; GLUT-1, glucose transporter-1.

Furthermore, by regulating microtubule stability, the Vpr induced aggregation of neuronal mitochondria and disrupted axonal transport [102]. In the meantime, it is considered that if the viral load is not checked, there will be a high probability of neuronal dysfunction. Interestingly, HIV-associated neurodegeneration cannot be correlated fully with cognitive deficits, as observed during the early phases of AD [103,104]. In recent studies, cognitive impairments in HAD patients had demonstrated a better correlation with synaptic dysfunction than neurodegeneration, which is further accompanied by synaptic loss, degeneration of axons and astrocytosis [105,106]. More studies are required to demonstrate whether neurocognitive deficits are still observed in patients even when the viral load is well under control. Some cohorts demonstrated that in HIV+ viraemic subjects, there is still a high occurrence of HAND, while others suggested that cognition is usually not impaired in individuals with no detectable viraemia [66,107,108]. This can possibly be explained by some possible mechanisms including (i) toxicity of ARVs, (ii) neuroinflammation, (iii) lack of proper cART penetration across the BBB, (iv) increased longevity of infected people and (v) restricted low-noise viral replication [109,110]. Further, constant orchestrated inflammatory events may open up the possibility to understand the linkage between HIV and AD-associated neurodegenerative conditions.

5. Mechanisms linking HIV-derived neuronal damage in the AD brain

With the introduction of cART, AIDS has become a chronic disease. A substantial number of HIV+ patients over 50–55 years of age are prone to age-related diseases [111]. The plaques formed by extracellular Aβ peptide deposits have been reported in patients, specifically before the cART era. Additionally, accelerated ageing such as immunosenescence is considered an integral part of the natural history of HIV infection. Specifically, HAND makes an impact on an already age-compromised organ and facilitates the occurring rate of neurodegenerative conditions. With reference to AD, concerns have been raised on the potential ties between HIV-CNS infection through various findings highlighting the modulation of amyloid and Tau pathways. Many symptoms correlated with AD pathomechanisms were observed in HIV+ individuals. Moreover, similar observations reported in the preclinical models represent neuro-AIDS and mimic neuronal dysfunction in HIV (table 3) [112133]. It has been reported that CSF features of HIV+ patients, present in HAND, resemble the sign and symptoms akin to the early and late stages of AD. For instance, Aβ1–42 levels were found considerably altered in the CSF of HAND patients [134]. However, when comparing CSF with HAND, late-stage AD and age-matched controls, reduced Aβ1–42 levels were observed in HIV+ individuals suffering from neuronal complications [134]. In particular, HIV+ patients without neurological manifestations may have a similar range of Aβ1–42 levels as reported in non-dementia controls.

Table 3.

Summary of common neurotoxic mechanisms of AD, observed between experimental models and HIV+ patients.

pathological hallmarks/symptoms observations in in vivo and in vitro HIV models methods used observations in HIV+ patients methods used references
neurodegeneration reduction in NeuN western blotting loss in cortical grey and white matter of the brain histologic post-mortem study [112114]
altered neurogenesis NA almost 20–50% neuronal damage in the frontal cortex NA [115,116]
neuroinflammation increased expression of microglial Iba1 and astrocytes GFAP histology and western blotting peripheral macrophages invasion and chemokines release cause massive gliosis NA [112,113,117]
oxidative stress increased expression of HIF-1, CYP2E1, NAPDH oxydase, IkB and iNOS western blotting higher ROS production Impaired mitochondrial dynamics and glucose metabolism NA [113,118]
cognitive and learning deficits deficits in Learning and cognition morris water maze test diminished memory performances NA [11,119]
atypical Tau phosphorylation higher expression of p-Ser396, p-Thr181, p-Ser404 and p-Thr231 western blotting increase in CSF total and phosphorylated Tau ELISA [20,112,113,120]
increased expression of GSK-3β contents and CDK5 western blotting frontal cortex display increased expression of GSK3b, CDK5 and p35 NA [112,113,121]
AβPP and Aβ synthesis misprocessing higher amyloid plaques generation congo red staining higher level of CSF Aβ1–42 ELISA [113,120]
increase of C99 fragment western blotting existence of amyloid plaques in brain NA [113,122,123]
higher expression of Aβ1–42 eLISA NA NA [124]
activation of neuronal cell death pathways and apoptosis increase of caspase-3, Bax, pJNK/JNK, Erk contents western blotting increased apoptosis TUNEL assay [99,113,125,126]
increased apoptosis tUNEL assay increased JNK/ERK contents and activities kinases assay and western blotting [99,125]
HPA axis deregulation higher expression of AVP, CRF mRNA and hypothalamic CRF NA impaired cytokine production, modification of glucocorticoid sensitivity and glucocorticoid resistance NA [127,128]
adrenal insufficiency, elevated plasma GC NA [129,130]
blood–brain barrier (BBB) HIV infection leads to increase leucocytes transmigration through metalloproteinases upregulation and downregulation of TJs proteins NA HAD patients show increased CSF/plasma albumin ratio NA [76,131]
excitotoxicity astrocytes cause increase in glutamate release and decrease in glutamate re-uptake NA increased levels of CSF glutamate ELISA [100,132,133]

Mounting evidence indicates that HIV protein/particle exposure to the brain directly or indirectly influences the regulation of amyloid and Tau signalling pathways [113,135137]. Recently, neurodegeneration has been noted in murine models of HIV (Gp120 transgenic mice and HIV-1 transgenic rats). It demonstrates increases in oxidative stress, gliosis, apoptosis, abnormal Aβ formation and phosphorylation of Tau. Further, the viral proteins like Tat affect Aβ synthesis, involving numerous mechanisms, including an increase in Aβ synthesis by deregulating structure and function of endolysosomes [135]. Similar to Tat, recombinant Gp120 injected primary hippocampal cells have demonstrated the promotion of Aβ1–42 secretion [138]. Also, Tat derived from a lentiviral vector exhibited expression in the hippocampus of transgenic mice (AβPP/PS1) and demonstrated an increase in Aβ1–42 formation along with a rise in the volume of amyloid plaques [124]. On the other hand, it causes a rise in Aβ aggregation by inhibiting its mediating degradation enzyme, Neprilysin. Moreover, it also enhances BACE1 expression and synthesis of the C99 fragment to accelerate the production of Aβ [113,135,139]. The increased expressions of BACE1 (commonly observed with AD) have been reported in HIV+ patients [140].

Recently, it has been reported that Tat protein in primary hippocampal neuronal cultures forms complexes with toxic Aβ peptides and potentiates a damaging effect by the formation of pores in the membrane [140]. In HIV-1 transgenic rats, the number and volume of amyloid plaques have been reported to be considerably elevated in the cerebral cortex due to an increase in amyloid C-terminal fragment C99 levels (greater than 5-fold) in the brain of HIV-1 transgenic rats [113]. Likewise, HIV-1 infected cells released p17 (HIV-1 matrix protein) which showed participation in Aβ-induced neuronal toxicity ascribed to misfolding and aggregation even when protease inhibitors (PI) are used [141]. When p17 was injected into the mouse hippocampus, it was observed to colocalize with plaques, phosphorylated Tau and fibril-like structures. In the same study, p17 was further demonstrated to be associated with increased Aβ production and impairment of cognitive function in experimental tests [141]. Recently, the regulatory effect of Gag polyprotein on AβPP metabolism has been demonstrated in macrophages and microglia. The Gag enhances Aβ load and associated neurotoxicity by triggering the activity of secretases. AβPP, on the other hand, mediates antiviral actions by sequestering Gag polyprotein in lipid rafts and limiting the release of HIV-1 [142]. To understand the balance between these two mechanisms (envision and restriction), and the impact on toxic Aβ peptide production, further studies are warranted.

The role of Tau protein in HAND pathogenesis is yet to be understood well. However, cognitive abnormalities accompanied by neuronal death and gliosis as a result of Tau hyperphosphorylation have been reported in transgenic mice (10-month-old Gp120 transgenic mice) [112]. Over-activation of glycogen synthase kinase 3β (GSK-3β) is believed to play a key role in such impairment as it is the main enzyme involved in Tau phosphorylation. Similarly, higher expression of cyclin-dependent kinase 5 (Cdk5), another important enzyme involved in Tau phosphorylation, has also been shown in HIV-1 transgenic rats along with raised levels of pTau (p-Thr181, p-Thr231 and p-Ser396), particularly in the hippocampal components [113]. Observations of experimental models therefore demonstrate the linkage between raised pTau and irregular NFTs in HIV+ patients with HAND [20,112,120].

6. Correlation between BBB, HIV and AD pathogenesis

BBB dysfunction is often associated with the pathogenesis of various neurodegenerative conditions, including HAND [143,144]. In AD, the micro-vessel disruption has been shown to be consistent with disease onset and progression [145147]. The occurrence of impaired BBB is shown to be associated with Aβ aggregation in several animal models as well as in patients suffering from AD [148150]. The BBB impairment arising from HIV-1 infection is probably accountable for the transmission of the virions from the vascular compartments. Additionally, it also proved to boost recruitment of immune cells and facilitates CNS infection by many opportunistic microbes [131,143,144]. The interaction between BBB and HIV-1 may occur in the neurovascular unit (NVU) cells by engaging viral proteins. Some studies have shown that by dysregulating gap junctions, HIV-infected astrocytes can damage BBB integrity and impair brain homeostasis [76]. Numerous viral proteins, including Tat, Gp120, Vpr and Nef, have been found to be associated with deregulated molecular and cellular pathways, and impairing the repair mechanisms, leading to BBB dysfunction [5]. The direct regulatory effect of Tat protein on endothelium has also been shown through multiple cellular routes, such as inhibition of the Ras pathways, culminating in reduced tight junction (TJ) protein expression and BBB dysfunction [151153]. These effects, mainly triggered by toxic Aβ accumulation in the brain, highlight a direct involvement of HIV proteins in Aβ–BBB interaction. Most importantly, Tat also regulates the expression of various Aβ associated receptors and transporters, which are engaged in the bidirectional movement of peptides across BBB. Recently, it has been shown that extracellular Tat induces receptor for advanced glycation endproducts (RAGE) activity and results in the activation of Ras/MAPK signalling cascade and agglomeration of Aβ [7,152]. In addition, it also reduces the clearance of Aβ across the endothelial cells and inhibits the synthesis of low-density lipoprotein receptor-related protein-1 (LRP-1) [152]. Similar to Tat, Gp120 has shown to alter BBB dynamics by regulating protein kinase C (PKC) and JAK/STAT signalling. Gp120 also increases monocyte migration, through which it enhances the number of HIV-infected monocytes that can cross the BBB to enter the CNS [5,154,155]. On the contrary, recombinant Gp120 administration showed injury in CNS micro-vessels that reveal that Gp120 may directly alter the function of endothelial cells in the brain and influence BBB dynamics [156]. These mechanisms ultimately lead to the diminished clearance of Aβ from the interstitial fluid and thus culminate in Aβ deposition, as well as accumulation in the brain. In this context, it is imperative to reasonably speculate and articulate the intriguing role of the BBB in AD and HAND pathogenesis [150].

7. Pathological hallmarks of AD: possible role of HIV

7.1. Amyloid beta (Aβ)

Atypical Aβ build-up is an important trait of AD reported in HIV-infected individuals [120,123]. Abnormalities associated with Aβ burden are more frequent in the AD brain than HIV, predominantly in the younger HIV-infected individuals. Ageing is considered as a potential risk for Aβ aggregation in HIV-infected individuals, although recent studies advocate that HIV and ageing both can influence Aβ aggregation independently, as well as together [136]. It has been shown that in HIV-infected individuals, the plaques are typically dispersed, and accumulation of Aβ generally occurs in brain somas and extracellular plaques as well as axonal tracks [120,123,157]. However, in AD, the plaques are of neurotic occurrence, predominantly in the extracellular spaces [158]. Some neuropathological findings demonstrate that Aβ aggregates in HIV cases preferentially in the basal ganglia, frontal lobe and hippocampus [123,159]. Though the site of Aβ deposition may show a discrepancy in AD brain, it usually tends to arise primarily in neocortical areas [158]. There are numerous studies that highlight the connection between long-term cART usage and aggregation of Aβ [123,159]. Accumulated Aβ may also exist without cognitive impairments in older adults; however, it is widespread and ubiquitous in the AD brain, and it is not a central feature of normal cognitive ageing [160]. The Aβ accumulation develops gradually with reduced neurotoxicity in similar brain areas with healthy ageing as in AD [161]. Though Aβ is strongly linked with AD, substantial evidence is still limited in context to HAND, where Aβ assists as a driving force.

7.2. Hyperphosphorylated Tau (pTau)

Tau is a microtubule-associated protein (MAP) that is accountable for maintaining a normal neuronal network. Hyperphosphorylation of Tau leads to its dissociation from microtubules and the dissociated tau forms paired helical filaments (PHFs) that eventually aggregate and generate NFTs. NFTs consisting of pTau are another characteristic trait of AD, specifically in people suffering from HIV [3,162,163]. The elevated level of Tau has been reported to occur at earlier ages in individuals suffering from HIV than in healthy individuals [20]. Even though pTau contents were found to be irrelevant to the viral levels in the brain, but pTau is often correlated with the activation of microglia [21]. In HIV cases, tau phosphorylation may be initiated by viral proteins as well as pro-inflammatory cytokines that cause amyloidosis and precede the growth of tau tangles [11]. Higher expression of pTau has also been shown to be correlated with ARV treatment [20]. It has been observed that relative to HIV, pTau usually forms in the entorhinal cortex and hippocampus, and later expands to adjacent areas, which represents the phenomenon observed during natural ageing and AD [20,164].

7.3. BBB impairment

The BBB is a biochemical barrier that helps in protecting CNS from potentially damaging substances, including neurotoxins and drugs. It also protects the neural tissues from variations in blood composition and neurotoxins [162]. The permeability of the BBB is altered in HIV infection, which permits effusion or leakage of toxic elements, such as infected macrophages from blood to the brain parenchyma. HIV has been reported to influence neuronal endocytosis, which further serves as a key player in impairing the integrity of BBB associated microvascular endothelial cells [165]. Further, upregulation of adhesion molecules and HIV-induced damage of the tight cell junctions facilitate BBB passage [6]. The disrupted BBB has also been correlated with toxic Aβ aggregation in HIV-infected individuals as other abnormalities arise from functional failure to sort out the Aβ peptides [7]. The increased intracellular Aβ agglomeration in microvascular endothelial cells has also been shown during HIV infection in an in vitro study [166]. The disrupted BBB, which is linked with AD pathogenesis, serves both as a reason and mediator of cerebral Aβ deposition affecting BBB permeability and Aβ agglomeration involving a common pathophysiological mechanism in AD and HIV cases [7,167].

7.4. CSF markers

The phosphorylated Tau and Aβ concentrations in CSF also correspond with their levels in the brain, though for toxic Aβ an opposite correlation exists, indicating a problem that is associated with its Aβ clearance. The higher expression of pTau and reduced Aβ level have been reported in the CSF of individuals suffering from symptomatic HIV, representing the phenomenon observed in AD. However, this finding lacks consistency principally for total Tau and pTau [120,168]. In a study, reduced CSF Aβ, but not accelerated pTau, was observed in an individual suffering with HAND [169]. Conversely, accelerated CSF pTau was also noted in asymptomatic HIV patients as compared to the normal controls [170]. Further, this finding also indicates raised levels of CSF pTau in HIV-infected older people suffering from HAND. In view of this finding, it is seen that similarities exist between HIV+ individuals and AD brain with reference to CSF Aβ and Tau, although larger disturbances have been observed consistently during AD in older people, predominantly in comparison with young adults manifesting neuro-asymptomatic HIV.

8. Risk factors and pathophysiological mechanisms of AD induced by HIV

8.1. Genetic predisposition

The apolipoproteins, in particular ε4 allele of apolipoprotein-E (ApoEε4), is known to be one of the major risk factors for AD, which is correlated with elevated Aβ agglomeration, diminished neurocognitive activity, decreased brain volumes and enhanced systemic progression of HIV infection [171173]. ApoEε4 susceptibility to HIV infection has been shown to be enhanced in vitro [173]. The greater expression of ApoEε4 was shown to be correlated with decreased cognition in HIV cases when compared with age-matched seronegative ApoEε4+ individuals, though many studies did not find a meaningful correlation between ApoEε4 and HAND [172,174]. Another isoform, ApoEμ4, has been shown to display a more stable association with cognitive functioning in AD than in HIV cases, as evidenced by the fact that carriers with two alleles may have up to 85–90% probability of developing AD by the age of 80. Many risk factors associated with developing AD have also been reported with the ApoEε4 risk alleles [171]. Although HIV may influence neurological structure and function, aggravated by pre-existing genetic factors, and then eventually lead to neurodegeneration or cognitive dysfunction following epigenetic changes [175].

8.2. Cerebral metabolism

Emerging evidence shows that HIV infection in individuals causes disturbances in cerebral metabolism, which significantly contributes to the development of brain defects and progression of neurocognitive deficit [6,176,177]. In HIV infection, there is mitochondrial dysfunction followed by oxidative stress via overproduction of reactive oxygen species (ROS), the release of neuroinflammatory markers, neuroimmune dysfunction, susceptibility to drug toxicities and development of HAND [6,177,178]. ROS is considered as the main cause of brain ageing due to oxidative changes as well as cellular damage that affects the aged brain along with impaired insulin signalling [179,180]. Further, glutamate overproduction, enhanced neuroinflammation and Ca2+ overload is associated with mitochondrial dysfunction, and all these contribute to the neurotoxicity [181]. Likewise, perturbations in brain mitochondrial activity, oxygen utilization capacity and carbohydrate metabolism have also been implicated in AD [182,183]. Additionally, the occurrence of oxidative stress at an early stage of AD promotes and facilitates the formation of Aβ-plaques and tau tangles [182].

8.3. Neuroinflammation

The dispersal of HIV takes place between infected monocytes to uninfected cerebral microglia and astrocytes, where it activates inflammatory immune responses by releasing cytokines, chemokines and ROS. Chronic and sustained neuroinflammation caused by prolonged glial and astrocyte activation has been reported to culminate in neuronal death and exhibit correlation with brain defects associated with HIV infections [6,177,178]. The positron emission tomography (PET) results have also shown functional changes due to regional microglial activation, consistent with autopsy findings that demonstrate frontal cortical aggregation of oxidative damage of macromolecules initiated by ROS in AIDS patients [184,185]. Enhanced glial expression has been observed in asymptomatic neuro cases of HIV with substantial activation of frontal and parietal components among people with HAD. This demonstrates that excessive glial activation and neuroinflammation attribute to cognitive impairment [186]. PET results also indicated that the systemic stimulation of microglia occurs in AD, often in conjunction with cognitive impairment [187]. Aβ aggregation also contributes to astrocyte activation as well as the onset of inflammatory reactions and related immunological responses. In addition to Aβ accumulation, NFTs induced neuronal degeneration also provokes neuroinflammation [167].

8.4. Neurotoxicity

An orchestrated reaction of excitotoxicity and apoptosis, which maintains immunological and inflammatory responses to the virus is potentially accountable for HIV-related brain dysfunction [6,177,180]. It has been found that depletion of T-cells and apoptosis are influenced directly by HIV gene expression, whereas indirectly by apoptosis in the uninfected cells. Tat, Gp120 and complementary proteins (such as Fas) are among the substances that have been implicated in HIV-associated neurotoxicity. Tat and Gp120 disrupt the uptake of glutamate by astrocytes, leading to glutamate excitotoxicity and trigger neuroinflammation and apoptosis. Further, they also result in Ca2+ accumulation and have neurotoxic effects of a related kind. Moreover, Tat can promote astrocytosis and neuronal death and associate with AβPP to enhance Aβ production [124]. Most importantly, viral structures and regulatory proteins also contribute to cerebral mitochondrial damage and BBB dysfunction following overproduction of ROS that causes oxidative injury [178,188].

Neurotoxicity may also result from numerous ARV drugs used to treat HIV cases, such as nucleoside analogue reverse transcriptase inhibitors. Some ARV drugs that penetrate the BBB and enter the brain efficiently than others possess more potential to cope with HIV-associated brain dysfunction [189]. In recent trials, cART-treated HIV patients exhibited a higher concentration of cerebral Aβ as well as pTau than cART-naive patients [20,123]. There have been contradictory results, but it seems unlikely that cART tends to be the major reason for brain dysfunction in most cases [169,190]. Nevertheless, further studies are required on cART-related neurotoxicity; specifically provided ongoing usage of cART in people of old age suffering from HIV and the probability of emergence of many medications which are under the different stages of clinical development. The inflammation and infection of other organ systems outside of the brain, including liver, gut and vascular systems may also represent indirect neurotoxicity. For instance, HIV causes leaky gut syndrome by damaging and impairing the permeability of the intestinal lining, allowing microbes and toxins to enter the blood and reach systemic circulation, which eventually causes neuroinflammation [191]. Further, in response to HIV, hepatic ceramides were correlated with various components of the metabolic syndrome, apoptosis and neurodegeneration [192].

8.5. Vascular and metabolic comorbidities

Numerous comorbidities like chronic substance abuse, often independent of the direct consequences of HIV, lead to HIV transmission, responsiveness and cognitive difficulties [22]. HCV also aggravates HIV-associated neurocognitive damage following similar mechanisms [193,194]. Further, vascular and metabolic conditions such as metabolic syndrome, diabetes mellitus, vascular injury and obesity are in parallel rise with chronically HIV-infected people age, and there are indications that HIV permits them to improve and flourish [195,196]. These conditions can also have an adverse effect on neurocognitive function [197,198]. For instance, impaired glucose metabolism which results in hyperglycaemia and hyperinsulinaemia provokes ROS production, tau hyperphosphorylation, Aβ accumulation and brain microangiopathy, and altogether these contribute towards a reduction in Aβ degradation and clearance [197]. Hence, vascular, neurological dysfunction may be a significant component of HAND caused by HIV, along with the development of vascular comorbidities. However, it is still challenging to identify the specific effect of vascular cognitive dysfunction to HAND. It should also be underlined that vascular risk factors are strongly dominant in aged individuals and there is a strong indication that these risk factors can be correlated with vascular, neurological impairment, even though there are no distinct cerebrovascular events [199]. Further, the epidemiological studies also suggest that these conditions raise the possibility of progression of AD and increase vascular risk in both HIV and AD individuals, and are correlated with higher Aβ burden [198,200202]. Additionally, the flexible complexity of vascular and metabolic risk factors may essentially represent therapeutic targets in order to prevent or curtail cognitive impairments in HIV-infected individuals.

9. Possible mechanisms linking HAND, synaptic degeneration and AD

As described previously, HIV-1 infection of the CNS initiates from the transmigration of HIV-1-infected peripheral blood monocytic cells/macrophages across the BBB. Subsequently, microglia and astrocytes become infected and reactivated. The immune-activated and HIV-1-infected microglia/macrophages release viral proteins (e.g. gp120, Tat, Nef and Vpr), chemokines (e.g. MCP1, CXCL12), cytokines (e.g. IL-1β, TNF-α, IL-6) and other neurotoxic factors. In addition, infected/reactivated astrocytes can also release neurotoxic substances and pathogenically increase synaptic activity with increased transmitter release and impaired glutamate re-uptake. The released neurotoxins and extracellular glutamate can cause excessive Ca2+ influx, perturbations of energy metabolism and ROS production, leading to the disruption of normal neuronal function. Most importantly, the released viral proteins, cytokines, chemokines and free radicals can trigger more glial cells and macrophages. These damaged neurons may mark the abnormal synapses with some kind of ‘eat-me’ signals, which can be recognized and eliminated by microglia and/or astrocytes through phagocytotic pathways such as the MerTK, Megf10 and APOE pathway in astrocytes and the complementary and FKN/CX3CR1 pathways in microglia. Further, all these mechanisms can contribute to AD-like characteristics, including Tau phosphorylation, Aβ production, oxidative stress and excitotoxicity, and also influence neuron integrity and CNS homeostasis. It is also observed that HIV+ patients present high glucocorticoid (cortisol) levels, characteristic of a hypothalamic–pituitary–adrenal (HPA) axis deregulation. Glucocorticoids (GC) and their receptors are highly engaged in the etiology of AD. Further, GC and their receptors may modulate/potentiate the development of HAND and potentially AD. The dysregulation of the HPA axis is observed both in HIV+ individuals and rodent models. GC overexposure, along with viral proteins or not, is able to induce the enhancement of Tau phosphorylation, Aβ production, oxidative stress, excitotoxicity, neuroinflammation and apoptosis. Through these numerous pathways, HIV-1 causes synaptic deficits and neurodegeneration, thus leading to cognitive impairment and behavioural deficits, and could also explain the establishment of HAND in HIV+ patients, and potentially the onset of AD. All these processes lead to neurodegeneration and synaptic deficits/degeneration, and are potentially responsible for cognitive decline observed in HAND patients, all of which could progressively favour the development of AD (figure 3) [203,204].

Figure 3.

Figure 3.

Schematic showing possible linkage between HAND, synaptic degeneration and AD.

10. Therapeutics strategies to combat HIV-mediated neuronal damage

In the above sections, we comprehensively discussed various underlying interconnected mechanisms between HIV, neuroinflammation, HAND and AD. Understanding the underlying mechanisms will help explore various possible therapeutic strategies and agents, which may be able to combat these complications. Unfortunately, there are no medications identified so far, and very few studies are available on therapeutic aspects. Neuroprotective therapies are designed with a targeted approach to ameliorate damage and improve survival as well as the function of neurons. The mechanisms associated with neuroprotection are classically aimed to diminish the extent of neuronal damage in HIV-1-induced neuronal dysfunction. It can be considered that agents that regulate inflammatory and/or cell death pathways and favourably modulate neurotransmitter function may provide opportunities for pharmacological manipulation during HIV-1 brain infections, although previous studies which focused on anti-inflammatory mechanisms have not demonstrated promising results in attenuating endogenous inflammation and considerable neuroprotection. As a result, a number of studies have recently been conducted to reduce neurotoxicity by blocking or modulating the actions of viral proteins, augmenting the protective action of neurotrophins and growth factors, or curtailing neuroinflammation triggered by HIV-1-infected microglia and macrophages (figure 4). For instance, the neuroprotective role of brain-derived neurotrophic factor (BDNF) has recently been observed in HIV-1-mediated neurotoxicity. It appears a potent neurotrophic agent for HIV-1 associated neuronal injury, which confers neuroprotection via inhibiting caspase-3 activation and HIV-1 Gp120 mediated neuronal apoptosis [205]. Moreover, BDNF is also found to reduce the levels of CXC chemokine receptor- 4 (CXCR4) and inhibit neuronal apoptosis by blocking the neurotoxic effects of SDF-1α, a ligand for CXCR4. The SDF-1-mediated apoptosis is quantitatively akin to that provoked by Gp120. CXCR4 activation can contribute to the cell death of a different kind of neuronal population. Consequently, BDNF-mediated neuroprotection occurs by reducing CXCR4 level that ultimately leads to the reduced activation of this receptor during HIV-1 neuropathogenesis [205]. Recently, activation of nuclear factor kappa beta (NF-κβ) mediating nerve growth factor (NGF) and BDNF and rise in Bcl-2 expression has also been reported to promote neuronal survival in HIV-1 associated neurodegeneration [206,207]. Additionally, BDNF has also been reported to prevent glutamate-induced excitotoxicity through modulation of NMDA receptors in HIV-1 patients [208]. Similarly, erythropoetin (Epo), a neurotrophin, can also confer neuroprotection against HIV [209]. A higher dose of Epo for a long duration showed better neuroprotective effect against HIV-1 transmission from mother to infant [210]. It can also protect cortical neurons against apoptosis by targeting HIV-1 Gp120 [211]. These observations suggest that Epo can be considered as a potential therapeutic agent for the treatment of HAD [212]. Recently, the promising role of recombinant human NGF (rhNGF) has shown to improve the symptoms associated with both HIV-related neuropathy and diabetic polyneuropathy. Substantial evidence demonstrates that NGF signalling may also prevent glutamate-induced neurotoxicity caused by ischemic injury. However, in HIV-1-induced neuronal damage, especially in the peripheral nervous system, NGF may have significant therapeutic effects [213215]. Activation of the insulin-like growth factor I (IGF-I) system is another potential approach to treat HAD, as it exhibited neuroprotective action against neurotoxins [216218]. Activating IGF-I-stimulated signalling components may offer a potential therapeutic approach to protect susceptible neurons in HAD patients. Earlier, impaired IGF-I responses were reported during the course of HIV infection [216218]. In HIV-infected patients, reduced levels of serum IGF-I have been observed particularly in children failure to thrive and individuals displaying wasting syndrome [216]. Reduction in the levels of IGF-I in CNS may aggravate neuronal apoptosis in the course of HIV infection [218]. Thus, it can be reasonably argued that activation of the IGF-I system or increased utilization of IGF-I-activated pathways may signify a promising treatment approach to rescue neurons susceptible or vulnerable to injury in HAD patients. Similarly, higher expression of fibroblast growth factor I FGF-I can also rescue the CNS from the neurotoxic effects of HIV. Altered expression of FGF-I and GSK-3β in susceptible neurons can be considered crucially important for the pathogenesis of HAD and emergence of therapeutic strategies [219,220].

Figure 4.

Figure 4.

Proposed protective role of neurotrophins, growth factors and drugs in combating HIV-induced neuronal damage. A neurotrophic agent such as BDNF confers neuroprotection via inhibiting caspase-3 activation and HIV-1 gp120 mediated neuronal apoptosis. BDNF can also curtail the levels of CXCR4 and prevent neuronal apoptosis by blocking the neurotoxic effects of SDF-1α. NGF and BDNF mediated activation of NF-κβ and upregulation of Bcl-2 may act as another way to promote neuronal survival in HIV-1 associated neurodegeneration. In addition, NGF signalling components can also prevent glutamate-induced neurotoxicity induced by ischaemic injury. Similarly, a neurotrophin like EPO has potential to protect cortical neurons against apoptosis by targeting HIV-1 gp120. Since impaired IGF-I and FGF responses were recorded during the course of HIV infection in several studies, therefore higher expression of these factors can also rescue the CNS from the neurotoxic effects of HIV. The altered expression of FGF-I and GSK-3β in susceptible neurons are now regarded as crucial during HAD pathogenesis. Further, drugs like memantine can be used to prevent neurotoxicity induced by Tat and gp120 viral proteins. Similarly, using inhibitors/drugs like valproate and lithium for GSK-3β could have therapeutic importance in HAD patients, since higher expression of GSK-3β induces apoptosis and it has been found to be associated with HIV-1 protein-mediated neurotoxicity. Finally, activated astrocytes-induced MCP-1 production positively influences neuroprotection through caspase-1 blockade. On the contrary, MCP-1 associated inflammatory reaction contributes to HIV-1- associated neurological illness. MCP-1 can protect human mixed cultures of neurons and astrocytes from Tat or NMDA-induced apoptosis by downregulating the extracellular glutamate expression, and in neurons by modulating Tat and NMDAR1 expression. These strategies together can be helpful in preventing HIV-induced neuronal damage.

Furthermore, the Tat and Gp120 mediated neurotoxicity can be fully blocked by memantine, an NMDA antagonist used well in the treatment of dementia [221,222]. It also ameliorates hippocampal synaptic transmission in the SCID mouse model of HIV-1-associated neurologic diseases [223]. Recently, the use of inhibitors of GSK-3β in the brain suggested that regulation of GSK-3β activity in neurons may be vital for neuroprotection. Higher expressions of GSK-3β induced apoptosis and showed association with HIV-1 protein-mediated neurotoxicity [224,225]. As a consequence, pharmacological agents like valproate and lithium identified to inhibit GSK- 3β activity could be valuable for therapeutic benefits in HAD patients.

The neuroprotective role of monocyte chemoattractant protein 1 (MCP-1) has recently been observed in HIV and HAD patients [226,227]. Activated astrocytes-induced MCP-1 production positively influences neuroprotection through the caspase-1 blockade. On the contrary, MCP-1 associated inflammatory reactions contribute to HIV-1-associated neurological ailments [226,227]. MCP-1 can protect mixed cultures of neurons and astrocytes from Tat or NMDA-induced apoptosis by downregulating the extracellular glutamate expression, along with modulating Tat and NMDAR1 expression [228]. In the case of HAD, MCP-1 may exert a protective as well as a degenerative role as it is coupled with monocyte recruitment and inflammation into the CNS [229]. The intricate balance between neuroinflammation and neuroprotection could be vital in triggering the initial as well as the ongoing response of the CNS to injury. Taken together, potential approaches to amplify the biologic effects of these factors or intensify their expression may support an advantageous role against this type of neurodegeneration.

11. Antiretroviral drugs: potential therapeutic agent for the treatment of HIV-induced neuronal damage

More recently, drugs used in highly active antiretroviral therapy (HAART) have shown improvement in cognitive functions, including all cognitive paradigms. The cognitive improvement is also correlated with an increase in CD4 count with a concomitant reduction in viral load [230]. The ability of ARV drugs to penetrate CNS supports the basis of its therapeutic success, as is evident in various reports. In order to reduce viral load, it is important that the drug should achieve a high concentration in the CSF following its ability to cross the BBB. Letendre et al. [231] examined the CNS penetrability of ARV drugs and ranked the ARV drugs for penetration based on scores assigned as 0 (low), 0.5 (intermediate) or 1 (high). This ranking system was based on drug concentrations in CSF, effectiveness in CNS and chemical properties in the clinical studies. The calculation for CNS penetration effectiveness (CPE) rank was determined by summing the individual penetration ranks for each ARV in the regime. For instance, combinations of efavirenz, zidovudine and lamivudine scored high for CPE [232]. Drugs like abacavir displayed low CPE score and rank; this was correlated well with higher viral load in the CSF [232]. Moreover, a small study involving 37 individuals demonstrated greater cognitive improvement with higher drug penetrability [233]. Similarly, another study looked at both HIV patients with cognitive impairment and patients with cognitive impairment without HIV, and it showed a worsening of cognitive. The ARV drugs with high penetrability can be neurotoxic too; thus, it is advised to suspect ARV drug neurotoxicity when cognitive improvement is not observed or detected with ARV treatment [234].

In the last few decades, appreciable progress has been made in the area of ARV therapy related to improved neurological clinical outcomes for HIV-1 patients. An immediate first-line treatment regimen for all new diagnosed HIV-1 infected patients is recommended by international guidelines for reducing the neurological complications associated with HIV-1infected patients [235,236]. Current ARV therapy is highly efficient in controlling HIV-1; still, viral replication can be found in the CSF among some patients. It has been found that ARVs reach different areas of CSF with significant variability due to the different expression profiles of cellular drug transporters and the concentrations of few ARVs do not the exceed inhibitory concentration for wild-type HIV replication in CSF [237,238] (table 4). The main limitation to achieve the HIV-1 eradication from the brain is the suboptimal concentrations of ARV within this site. Factors like molecular weight, blood protein binding and lipophilicity influence the concentration of drug in the brain tissue [231,241243]. For instance, while entry and integrase inhibitors are able to reach the CNS, the nucleoside/nucleotide reverse transcriptase inhibitors and non-nucleoside reverse transcriptase inhibitors can only partially cross the BBB. Conversely, the majority of PIs are characterized by a medium/low permeability to the BBB [5,239,244,245]. Furthermore, some cellular transporters like P-gp, MRP4 and MRP5 have the ability to reduce the intracellular concentration of ARV drugs which ultimately favours both the emergence of drug-resistant viruses and their productive infections to other cells [46,56,240,246].

Table 4.

Class, name and CNS penetration of the antiretroviral drugs [239,240].

class of drug name of the drug CNS penetration
protease inhibitor tipranavir low
fosamprenavir medium
atazanavir medium
saquinavir low
nelfinavir low
lopinavir medium
ritonavir low
darunavir medium
indinavir medium
amprenavir medium
nucleoside reverse transcriptase inhibitor tenofovir disoproxil fumarate low
abacavir medium
didanosine medium
emtricitabine medium
stavudine medium
lamivudine medium
zidovudine high
entry/fusion inhibitors maraviroc high
enfuvirtide low
non-nucleoside reverse transcriptase inhibitor etravirine low
delavirdine high
nevirapine high
efavirenz medium
integrase strand transfer inhibitor raltegravir medium
elvitegravir medium

New strategies like the usage of a hypertonic solution of urea or mannitol [48,49] are currently used to increase the concentrations of ARV within site. This deed can be achieved by inhibiting the drug efflux transport, while nanoparticles and cell-mediated nanoART may confer other key advantages, such as improved blood half-life and bioavailability, precise delivery and higher aqueous stability [231]. Different types of nanoparticles that have been identified for improving the concentration of ARV are listed below:

  • 1.

    Lipid nanoparticles have the ability to easily cross the BBB [247,248].

  • 2.

    Polymeric nanoparticles are able to exploit the interaction with low-density lipoproteins receptors on the surface of endothelial cells [239,249].

  • 3.

    Inorganic nanoparticles such as small size silica with the addition of polyethylene glycol (PEG) [250].

  • 4.

    Gold nanoparticles conjugated with cell-penetrating peptides [251].

It has been recently reported that poly(dl-lactide-co-glycolide) nanoparticles and other nanoparticles increase the peak concentrations of lopinavir, ritonavir and efavirenz (these drugs are characterized by a low penetration into CNS) [239,252]. Recently, a CPE that depends on pharmacokinetics' features of various ARV drugs was proposed to estimate the efficacy of ARV treatment in the CSF [238]. However, some contradictory results of this CPE on clinical outcomes in HIV-1 infected patients have been reported in some of the studies [169,232,234]. These observations reflect that further studies are required to prescribe ARV therapy and that the regimens characterized by high CPE scores must be carefully chosen. It has been demonstrated that in the presence of high CPE, there is an acceleration of neurological disorders [253,254]. For instance, PIs are shown to induce oxidative stress in neuronal cells, while the NNRTI efavirenz caused toxicity in the cortical neuronal cultures of fetal rats [253255]. Still in vivo studies are needed to confirm the neurotoxicity profiles of these drugs for potential applications.

Further, various reports highlighted the use of psychiatric medication for mood disorders like depression. Many subtypes of antidepressants, including tricyclic antidepressants, serotonin–norepinephrine re-uptake inhibitors and selective serotonin re-uptake inhibitors, have been found useful in providing moderate symptomatic relief [256,257]. Psychostimulants may also be useful for apathy and fatigue [258]. Psychotic and manic symptoms are less reported in the case of HIV+ individuals, though a small-scale study with psychosis demonstrated a higher occurrence of extrapyramidal symptoms [259]. Numerous drugs such as mood stabilizers (like lithium) may have concurrent neurotoxic effects, and carbamazepine may stimulate the same CYP enzyme system which participates in the metabolism of ARV drugs, and therefore may cause drug–drug interactions [260,261]. However, on a pharmacological basis, many agents including memantine, nimodipine, selegiline, pentoxifylline and peptide T can be considered neuroprotective, although among these numerous agents, only selegiline appears to exhibit potential benefits [262].

12. Conclusion

Based on the available literature, it can be concluded that HIV-associated synaptic loss and aetiology of AD and HAND is an interconnected and orchestrated consequence of numerous neuropathogenic processes triggered by HIV-1. Interactions between HIV-1 and the host cells are believed to play a vital role in the pathogenesis of these abnormalities. Several viral proteins (Tat, Gp120, Nef and Vpr), which are released from infected cells in the nervous system, may impart induction of synaptic injury and pathogenesis of AD. In addition, these proteins are likely to act in conjunction and cause synaptotoxicity when released from infected cells in the CNS. Further, AD-associated numerous factors such as BBB regulators, members of the stress-related pathways as well as the amyloid and Tau pathways appear to augment amyloid plaques deposition or NFT accumulation following HIV neuroinfections. Additionally, the HPA axis dysregulation also showed that when associated with HIV infection, it is conducive of generating an environment where BBB disruption, neuroinflammation, oxidative stress, excitotoxicity and Aβ burden are exacerbated. This, combined with other factors (environmental/genetic), may provide a new insight for understanding the pathogenesis, diagnosis and therapeutics of brain disorders including AD and HAND. The scenario of replication-independent production of HIV-1 protein is apparently counterintuitive, and the underlying molecular mechanism is yet largely remained unexplored. It is therefore imperative to explore more in this field. Considering the need for therapeutics against HIV neuroinfection, unfortunately, still, there is an urgent need for evidence-based medications to be identified that would be able to combat these complications. Many studies have recently shown a reduction in neurotoxicity via modulating the actions of viral proteins, augmenting the protective action of neurotrophins and growth factors, or curtailing neuroinflammation triggered by HIV-1-infected microglia and macrophages. The mechanisms associated with neuroprotection are classically aimed to diminish the extent of neuronal damage in HIV-1-induced synaptic dysfunction. Agents that regulate inflammatory and/or cell death pathways and favourably modulate neurotransmitter function may provide opportunities for pharmacological manipulation during HIV-1 brain infections. Altogether, in the near future, it could be of paramount significance to explore the molecular mechanisms of HIV neuroinfection and develop therapeutic strategies.

Data accessibility

This article has no additional data.

Authors' contributions

A.S., S.K.J., S.O., K.D., D.K.C., G.G. and K.K.K. initiated the draft of the review. N.K.J. and S.K.S. edited the review and performed proofreading and final editing. All authors read and approved the final manuscript. S.B., S.D.S., M.M.T. and J.R. contributed in revision of the review.

Competing interests

We declare we have no competing interests.

Funding

We received no funding for this study.

References

  • 1.Jha NK, Jha SK, Sharma R, Kumar D, Ambasta RK, Kumar P. 2018. Hypoxia-induced signaling activation in neurodegenerative diseases: targets for new therapeutic strategies. J. Alzheimers Dis. 62, 15–38. ( 10.3233/JAD-170589) [DOI] [PubMed] [Google Scholar]
  • 2.Jha NK, Jha SK, Kar R, Ambasta RK, Kumar P. 2014. Role of oxidative stress, ER stress and ubiquitin proteasome system in neurodegeneration. MOJ Cell Sci. Rep. 1, 38–44. ( 10.15406/mojcsr.2014.01.00010) [DOI] [Google Scholar]
  • 3.Jha NK, Jha SK, Kumar D, Kejriwal N, Sharma R, Ambasta RK, Kumar P. 2015. Impact of insulin degrading enzyme and Neprilysin in Alzheimer's disease biology: characterization of putative cognates for therapeutic applications. J. Alzheimers Dis. 48, 891–917. ( 10.3233/JAD-150379) [DOI] [PubMed] [Google Scholar]
  • 4.Jha SK, Jha NK, Kumar D, Ambasta RK, Kumar P. 2017. Linking mitochondrial dysfunction, metabolic syndrome and stress signaling in neurodegeneration. Biochim. Biophys. Acta Mol. Basis Dis. 1863, 1132–1146. ( 10.1016/j.bbadis.2016.06.015) [DOI] [PubMed] [Google Scholar]
  • 5.Zhang YL, Ouyang YB, Liu LG, Chen DX. 2015. Blood-brain barrier and neuro-AIDS. Eur. Rev. Med. Pharmacol. Sci. 19, 4927–4939. [PubMed] [Google Scholar]
  • 6.Anderson E, Zink W, Xiong H, Gendelman HE. 2002. HIV-1-associated dementia: a metabolic encephalopathy perpetrated by virus-infected and immunecompetent mononuclear phagocytes. J. Acquir. Immune Defic. Syndr. 31, S43–S54. ( 10.1097/00126334-200210012-00004) [DOI] [PubMed] [Google Scholar]
  • 7.Andras IE, Toborek M. 2013. Amyloid beta accumulation in HIV-1-infected brain: the role of the blood brain barrier. IUBMB Life 65, 43–49. ( 10.1002/iub.1106) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Ghafouri M, Amini S, Khalili K, Sawaya BE. 2006. HIV-1 associated dementia: symptoms and causes. Retrovirology 3, 28 ( 10.1186/1742-4690-3-28) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Lawrence DM, Major EO. 2002. HIV-1 and the brain: connections between HIV-1-associated dementia, neuropathology and neuroimmunology. Microbes Infect. 4, 301–308. ( 10.1016/S1286-4579(02)01542-3) [DOI] [PubMed] [Google Scholar]
  • 10.Tucker KA, Robertson KR, Lin W, Smith JK, An H, Chen Y, Aylward SR, Hall CD. 2004. Neuroimaging in human immunodeficiency virus infection. J. Neuroimmunol. 157, 153–162. ( 10.1016/j.jneuroim.2004.08.036) [DOI] [PubMed] [Google Scholar]
  • 11.Becker JT, et al. 2009. Vascular risk factors, HIV serostatus, and cognitive dysfunction in gay and bisexual men. Neurology 73, 1292–1299. ( 10.1212/WNL.0b013e3181bd10e7) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.González-Scarano F, Martín-García J. 2005. The neuropathogenesis of AIDS. Nat. Rev. Immunol. 5, 69–81. ( 10.1038/nri1527) [DOI] [PubMed] [Google Scholar]
  • 13.Clifford DB, Ances BM. 2013. HIV-associated neurocognitive disorder. Lancet Infect. Dis. 13, 976–986. ( 10.1016/S1473-3099(13)70269-X) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Thakur KT, Boubour A, Saylor D, Das M, Bearden DR, Birbeck GL. 2018. Global HIV neurology: a comprehensive review. AIDS 33, 163–184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Antinori A, et al. 2007. Updated research nosology for HIV-associated neurocognitive disorders. Neurology 69, 1789–1799. ( 10.1212/01.WNL.0000287431.88658.8b) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Blackstone K, Moore D, Woods S, Morgan E, Franklin D, Ellis R. 2012. The CHARTER Group. How ‘asymptomatic’ Is HIV-associated asymptomatic neurocognitive impairment? In 19th Conf. on Retroviruses and Opportunistic Infections, Seattle, WA, Abstract 497. [Google Scholar]
  • 17.Boska MD, et al. 2004. Advances in neuroimaging for HIV-1 associated neurological dysfunction: clues to the diagnosis, pathogenesis and therapeutic monitoring. Curr. HIV Res. 2, 61–78. ( 10.2174/1570162043485095) [DOI] [PubMed] [Google Scholar]
  • 18.Aylward EH, Brettschneider PD, McArthur JC, Harris GJ, Schlaepfer TE, Henderer JD, Barta PE, Tien AY, Pearlson GD. 1995. Magnetic resonance imaging measurement of gray matter volume reductions in HIV dementia. Am. J. Psychiatry 152, 987–994. ( 10.1176/ajp.152.7.987) [DOI] [PubMed] [Google Scholar]
  • 19.Kaul M, Garden GA, Lipton SA. 2001. Pathways to neuronal injury and apoptosis in HIV-associated dementia. Nature 410, 988–994. ( 10.1038/35073667) [DOI] [PubMed] [Google Scholar]
  • 20.Anthony IC, Ramage SN, Carnie FW, Simmonds P, Bell JE. 2006. Accelerated Tau deposition in the brains of individuals infected with human immunodeficiency virus-1 before and after the advent of highly active anti-retroviral therapy. Acta Neuropathol. 111, 529–538. ( 10.1007/s00401-006-0037-0) [DOI] [PubMed] [Google Scholar]
  • 21.Smith DB, Simmonds P, Bell JE. 2014. Brain viral burden, neuroinflammation and neurodegeneration in HAART-treated HIV positive injecting drug users. J. Neurovirol. 20, 28–38. ( 10.1007/s13365-013-0225-3) [DOI] [PubMed] [Google Scholar]
  • 22.Grassi MP, Clerici F, Perin C, Zocchetti C, Borella M, Cargnel A, Mangoni A. 1995. HIV infection and drug use: influence on cognitive function. AIDS 9, 165–170. ( 10.1097/00002030-199509020-00008) [DOI] [PubMed] [Google Scholar]
  • 23.Williams KC, Hickey WF. 2002. Central nervous system damage, monocytes and macrophages, and neurological disorders in AIDS. Annu. Rev. Neurosci. 25, 537–562. ( 10.1146/annurev.neuro.25.112701.142822) [DOI] [PubMed] [Google Scholar]
  • 24.Peng H, Sun L, Jia B, Lan X, Zhu B, Wu Y, Zheng J. 2011. HIV-1-infected and immune-activated macrophages induce astrocytic differentiation of human cortical neural progenitor cells via the STAT3 pathway. PLoS ONE 6, e19439 ( 10.1371/journal.pone.0019439) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Hauser KF, Fitting S, Dever SM, Podhaizer EM, Knapp PE. 2012. Opiate drug use and the pathophysiology of neuroAIDS. Curr. HIV Res. 10, 435–452. ( 10.2174/157016212802138779) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Thompson KA, Cherry CL, Bell JE, McLean CA. 2011. Brain cell reservoirs of latent virus in presymptomatic HIV-infected individuals. Am. J. Pathol. 179, 1623–1629. ( 10.1016/j.ajpath.2011.06.039) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Liu Y, Liu H, Kim BO, Gattone VH, Li J, Nath A, Blum J, He JJ. 2004. CD4-independent infection of astrocytes by human immunodeficiency virus type 1: requirement for the human mannose receptor. J. Virol. 78, 4120–4133. ( 10.1128/JVI.78.8.4120-4133.2004) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Coiras M, López-Huertas MR, Pérez-Olmeda M, Alcamí J. 2009. Understanding HIV-1 latency provides clues for the eradication of long-term reservoirs. Nat. Rev. Microbiol. 7, 798–812. ( 10.1038/nrmicro2223) [DOI] [PubMed] [Google Scholar]
  • 29.Cassol E, Alfano M, Biswas P, Poli G. 2006. Monocyte-derived macrophages and myeloid cell lines as targets of HIV-1 replication and persistence. J. Leukoc. Biol. 80, 1018–1130. ( 10.1189/jlb.0306150) [DOI] [PubMed] [Google Scholar]
  • 30.Aquaro S, Bagnarelli P, Guenci T, De Luca A, Clementi M, Balestra E, Caliò R, Perno CF. 2002. Long-term survival and virus production in human primary macrophages infected by human immunodeficiency virus. J. Med. Virol. 68, 479–488. ( 10.1002/jmv.10245) [DOI] [PubMed] [Google Scholar]
  • 31.Koppensteiner H, Brack-Werner R, Schindler M. 2012. Macrophages and their relevance in human immunodeficiency virus type I infection. Retrovirology 9, 82 ( 10.1186/1742-4690-9-82) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Orenstein J, Fox C, Wahl S. 1997. Macrophages as a source of HIV during opportunistic infections. Science 276, 1857–1861. ( 10.1126/science.276.5320.1857) [DOI] [PubMed] [Google Scholar]
  • 33.Coiras M. 2010. HIV-1 latency and eradication of long-term viral reservoirs. Discov. Med. 9, 185–191. [PubMed] [Google Scholar]
  • 34.Le Douce V, Herbein G, Rohr O, Schwartz C. 2010. Molecular mechanisms of HIV-1 persistence in the monocyte-macrophage lineage. Retrovirology 7, 32 ( 10.1186/1742-4690-7-32) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Zhang D, Hu X, Qian L, O'Callaghan JP, Hong J-S. 2010. Astrogliosisi in CNS pathologies: is there a role for microglia? Mol. Neurobiol. 41, 232–241. ( 10.1007/s12035-010-8098-4) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Farina C, Aloisi F, Meinl E. 2007. Astrocytes are active players in cerebral innate immunity. Trends Immunol. 28, 138–145. ( 10.1016/j.it.2007.01.005) [DOI] [PubMed] [Google Scholar]
  • 37.Aikaterini A. 2008. Cellular reservoirs of HIV-1 and their role in viral persistence. Curr. HIV Res. 6, 388–400. ( 10.2174/157016208785861195) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Dong Y, Benveniste EN. 2001. Immune function of astrocytes. Glia 190, 180–190. ( 10.1002/glia.1107) [DOI] [PubMed] [Google Scholar]
  • 39.Narasipura SD, Henderson LJ, Fu SW, Chen L, Kashanchi F, Al-Harthi L. 2012. Role of β-catenin and TCF/LEF family members in transcriptional activity of HIV in astrocytes. J. Virol. 86, 1911–1921. ( 10.1128/JVI.06266-11) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Albright AV, Soldan SS, González-Scarano F. 2003. Pathogenesis of human immunodeficiency virus-induced neurological disease. J. Neurovirol. 9, 222–227. ( 10.1080/13550280390194073) [DOI] [PubMed] [Google Scholar]
  • 41.Aquaro S, Svicher V, Ronga L, Perno CF, Pollicita M. 2008. HIV-1-associated dementia during HAART therapy. Recent Pat. CNS Drug Discov. 3, S23–S33. [DOI] [PubMed] [Google Scholar]
  • 42.Kohleisen B, Shumay E, Sutter G, Foerster R, Brack-Werner R, Nuesse M, Erfle V. 1999. Stable expression of HIV-1 Nef induces changes in growth properties and activation state of human astrocytes. AIDS 13, 2331–2341. ( 10.1097/00002030-199912030-00004) [DOI] [PubMed] [Google Scholar]
  • 43.Wei L, Henderson LJ, Major EO, Al-Harthi L. 2011. IFN-γ mediates enhancement of HIV replication in astrocytes by inducing an antagonist of the β-catenin pathway (DKK1) in a STAT 3-dependent manner. J. Immunol. 186, 6771–6778. ( 10.4049/jimmunol.1100099) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Mamik MK, Banerjee S, Walseth TF, Hirte R, Tang L, Borgmann K, Ghorpade A. 2011. HIV-1 and IL-1β regulate astrocytic CD38 through mitogen-activated protein kinases and nuclear factor-κB signaling mechanisms. J. Neuroinflammation 8, 145 ( 10.1186/1742-2094-8-145) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Trujillo R, Jaramillo-Rangel G, Ortega-Martinez M, Penalva De Oliveira AC, Vidal JE, Bryant J, Gallo RC. 2005. International NeuroAIDS: prospects of HIV-1 associated neurological complications. Cell Res. 15, 962–969. ( 10.1038/sj.cr.7290374) [DOI] [PubMed] [Google Scholar]
  • 46.Mukhtar M, Harley S, Chen P, BouHamdan M, Patel C, Acheampong E, Pomerantz RJ. 2002. PrimaryIsolated human brain microvascular endothelial cells express diverse HIV/SIV-associated chemokine coreceptors and DC-SIGN and L-SIGN. Virology 297, 78–88. ( 10.1006/viro.2002.1376) [DOI] [PubMed] [Google Scholar]
  • 47.Hazleton JE, Berman JW, Eugenin EA. 2010. Novel mechanisms of central nervous system damage in HIVinfection. HIV/AIDS 2, 39–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Bergamaschi A, Pancino G. 2010. Host hindrance to HIV-1 replication in monocytes and macrophages. Retrovirology 7, 31 ( 10.1186/1742-4690-7-31) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Coleman CM, Wu L. 2009. HIV interactions with monocytes and dendritic cells: viral latency and reservoirs. Retrovirology 6, 51 ( 10.1186/1742-4690-6-51) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Strazza M, Pirrone V, Wigdahl B, Nonnemacher MR. 2011. Breaking down the barrier: the effects of HIV-1 on the blood-brain barrier. Brain Res. 1399, 96–115. ( 10.1016/j.brainres.2011.05.015) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Gavegnano C, Schinazi RF. 2010. Antiretroviral therapy in macrophages: implication for HIV eradication. Antivir. Chem. Chemother. 20, 63–78. ( 10.3851/IMP1374) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Meléndez LM, Colon K, Rivera L, Rodriguez-Franco E, Toro-Nieves D. 2011. Proteomic analysis of HIV-infected macrophages. J. Neuroimmune Pharmacol. 6, 89–106. ( 10.1007/s11481-010-9253-4) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Crowe S, Zhu T, Muller WA. 2003. The contribution of monocyte infection and trafficking to viral persistence,and maintenance of the viral reservoir in HIV infection. J. Leukoc. Biol. 74, 635–641. ( 10.1189/jlb.0503204) [DOI] [PubMed] [Google Scholar]
  • 54.Ellery PJ, et al. 2007. The CD16+ monocyte subset is more harbors HIV-1 in vivo. J. Immunol. 178, 6581–6589. ( 10.4049/jimmunol.178.10.6581) [DOI] [PubMed] [Google Scholar]
  • 55.Khan NA, Di Cello F, Stins M, Kim KS. 2007. Gp120-mediated cytotoxicity of human brain microvascularendothelial cells is dependent on p38 mitogen-activated protein kinase activation. J. Neurovirol. 13, 242–251. ( 10.1080/13550280701286531) [DOI] [PubMed] [Google Scholar]
  • 56.Acheampong E, Parveen Z, Muthoga LW, Kalayeh M, Mukhatar M, Pomerantz RJ. 2005. Human immunodeficiency virus type 1 Nef potently induces apoptosis in primary human brain microvascular endothelial cells via the activation of caspases. J. Virol. 79, 4257–4269. ( 10.1128/JVI.79.7.4257-4269.2005) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Kim AY, Chung RT. 2009. Coinfection with HIV-1 and HCV-a one-two punch. Gastroenterology 137, 795–814. ( 10.1053/j.gastro.2009.06.040) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Guha D, Nagilla P, Redinger C, Srinivasan A, Schatten GP, Ayyavoo V. 2012. Neuronal apoptosis by HIV-1Vpr: contribution of proinflammatory molecular networks from infected target cells. J. Neuroinflammation 9, 138 ( 10.1186/1742-2094-9-138) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Hong S, Banks WA. 2015. Role of the immune system in HIV-associated neuroinflammation and neurocognitiveImplications. Brain Behav. Immun. 45, 1–12. ( 10.1016/j.bbi.2014.10.008) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Palacio M, Álvarez S, Muñoz-fernández MÁ. 2012. HIV-1 infection and neurocognitive impairment in the current era. Rev. Med. Virol. 22, 33–45. ( 10.1002/rmv.711) [DOI] [PubMed] [Google Scholar]
  • 61.Vera JH, Guo Q, Rabiner I, Matthews P, Gunn R, Winston A. 2014. Neuroinflammation in asymptomatic HIV-infected subjects on effective cART. In Proc. of the Conf. on Retroviruses and Opportunistic Infections (CROI), Boston, MA, USA, 3–6 March 2014 Abstract 486LB. [Google Scholar]
  • 62.Vera JH, et al. 2015. Microbial translocation is associated with neuroinflammation in HIV-infected subjects on ART. In Proc. of the 22nd Conf. on Retroviruses and Opportunistic Infections, Seattle, WA, USA, 23–26 February 2015 Abstract 477. [Google Scholar]
  • 63.Peudenier S, Héry C, Ng KH, Tardieu M. 1991. HIV receptors within the brain: a study of CD4 and MHC-II on human neurons, astrocytes and microglial cells. Res. Virol. 142, 145–149. ( 10.1016/0923-2516(91)90051-4) [DOI] [PubMed] [Google Scholar]
  • 64.Sturdevant CB, Joseph SB, Schnell G, Price RW, Swanstrom R, Spudich S. 2015. Compartmentalized replication of R5T cell-tropic HIV-1 in the central nervous system early in the course of infection. PLoS Pathog. 1, e1004720 ( 10.1371/journal.ppat.1004720) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Ellis R, Langford D, Masliah E. 2007. HIV and antiretroviral therapy in the brain: neuronal injury and repair. Nat. Rev. Neurosci. 8, 33–44. ( 10.1038/nrn2040) [DOI] [PubMed] [Google Scholar]
  • 66.McArthur JC, Steiner J, Sacktor N, Nath A. 2010. Human immunodeficiency virus-associated neurocognitive disorders: mind the gap. Ann. Neurol. 67, 699–714. ( 10.1002/ana.22053) [DOI] [PubMed] [Google Scholar]
  • 67.Maschke M, Kastrup O, Esser S, Ross B, Hengge U, Hufnagel A. 2000. Incidence and prevalence of neurological disorders associated with HIV since the introduction of highly active antiretroviral therapy (HAART). J. Neurol. Neurosurg. Psychiatry 69, 376–380. ( 10.1136/jnnp.69.3.376) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Heaton RK, et al. 2011. HIV-associated neurocognitive disorders before and during the era of combination antiretroviral therapy: differences in rates, nature, and predictors. J. Neurovirol. 17, 3–16. ( 10.1007/s13365-010-0006-1) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.McArthur JC, Haughey N, Gartner S, Conant K, Pardo C, Nath A, Sacktor N. 2003. Human immunodeficiency virus-associated dementia: an evolving disease. J. Neurovirol. 9, 205–221. ( 10.1080/13550280390194109) [DOI] [PubMed] [Google Scholar]
  • 70.McArthur JC, et al. 1993. Dementia in AIDS patients: incidence and risk factors. Multicenter AIDS Cohort study. Neurology 43, 2245–2252. ( 10.1212/wnl.43.11.2245) [DOI] [PubMed] [Google Scholar]
  • 71.Soontornniyomkij V, Umlauf A, Soontornniyomkij B, Gouaux B, Ellis RJ, Levine AJ, Moore DJ, Letendre SL. et al. 2018. Association of antiretroviral therapy with brain aging changes among HIV-infected adults. AIDS 32, 2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Janssen R. 1992. Epidemiology of human immunodeficiency virus infection and the neurologic complications of the infection. Semin. Neurol. 12, 10–17. ( 10.1055/s-2008-1041152) [DOI] [PubMed] [Google Scholar]
  • 73.Williams DW, Anastos K, Morgello S, Berman JW. 2015. JAMA and ALCAM are therapeutic targets to inhibit diapedesis across the BBB of CD14CCD16C monocytes in HIV-infected individuals. J. Leukoc. Biol. 97, 401–412. ( 10.1189/jlb.5A0714-347R) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Cosenza MA, Zhao ML, Si Q, Lee SC. 2002. Human brain parenchymal microglia express CD14 and CD45 and are productively infected by HIV-1 in HIV-1 encephalitis. Brain Pathol. 12, 442–455. ( 10.1111/j.1750-3639.2002.tb00461.x) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Rothenaigner I, Kramer S, Ziegler M, Wolff H, Kleinschmidt A, Brack-werner R. 2007. Long-term HIV-1 infection of neural progenitor populations. AIDS 21, 2271–2281. ( 10.1097/QAD.0b013e3282f12f27) [DOI] [PubMed] [Google Scholar]
  • 76.Eugenin EA, Clements JE, Zink MC, Berman JW. 2011. Human immunodeficiency virus infection of human astrocytes disrupts blood-brain barrier integrity by a gap junction-dependent mechanism. J. Neurosci. 31, 9456–9465. ( 10.1523/JNEUROSCI.1460-11.2011) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Hellmuth J, Valcour V, Spudich S. 2015. CNS reservoirs for HIV: implications for eradication. J. Virus Erad. 1, 67–71. ( 10.1016/S2055-6640(20)30489-1) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Nutile-McMenemy N, Elfenbein A, Deleo JA. 2007. Minocycline decreases in vitro microglial motility, beta1-integrin, and Kv1.3 channel expression. J. Neurochem. 103, 2035–2046. ( 10.1111/j.1471-4159.2007.04889.x) [DOI] [PubMed] [Google Scholar]
  • 79.Lu S-M, et al. 2011. HIV-1 Tat-induced microgliosis and synaptic damage via interactions between peripheral and central myeloid cells. PLoS ONE 6, e23915 ( 10.1371/journal.pone.0023915) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Nath A, Conant K, Chen P, Scott C, Major EO. 1999. Transient exposure to HIV-1 Tat protein results in cytokine production in macrophages and astrocytes. A hit and run phenomenon. J. Biol. Chem. 274, 17 098–17 102. ( 10.1074/jbc.274.24.17098) [DOI] [PubMed] [Google Scholar]
  • 81.Zucchini S, et al. 2013. Increased excitability in tat-transgenic mice: role of tatin HIV-related neurological disorders. Neurobiol. Dis. 55, 110–119. ( 10.1016/j.nbd.2013.02.004) [DOI] [PubMed] [Google Scholar]
  • 82.Berman J, Carvallo L, Buckner CM, Luers A, Prevedel L, Bennett MV, Eugenin EA. 2016. HIV-tat alters Connexin43 expression and trafficking in human astrocytes: role in NeuroAIDS. J. Neuroinflammation 13, 54 ( 10.1186/s12974-016-0510-1) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Fan Y, He JJ. 2016. HIV-1 Tat induces unfolded protein response and endoplasmic reticulum stress in astrocytes and causes neurotoxicity through GFAP activation and aggregation. J. Biol. Chem. 291, 22 819–22 829. ( 10.1074/jbc.M116.731828) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Shin AH, Thayer SA. 2013. Human immunodeficiency virus-1 protein Tat induces excitotoxic loss of presynapticterminals in hippocampal cultures. Mol. Cell. Neurosci. 54, 22–29. ( 10.1016/j.mcn.2012.12.005) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Bai L, Zhu X, Ma T, Wang J, Wang F, Zhang S. 2013. The p38 MAPK NF-κB pathway, not the ERK pathway, is involved in exogenous HIV-1 Tat-induced apoptotic cell death in retinal pigment epithelial cells. Int. J. Biochem. Cell Biol. 31, 1–8. [DOI] [PubMed] [Google Scholar]
  • 86.Midde N, Gomez A, Zhu J. 2012. HIV-1 Tat decreases dopamine transporter cell surface expression and vescicular monoamine transporter-2 function in Rat striatal synaptosomes. J. Neuroimmune Pharmacol. 7, 629–639. ( 10.1007/s11481-012-9369-9) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Chen L, Liu J, Xu C, Keblesh J, Zang W, Xiong H. 2011. HIV-1gp120 induces neuronal apoptosis through enhancement of 4-aminopyridine-senstive outward K+ currents. PLoS ONE 6, e25994 ( 10.1371/journal.pone.0025994) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.El-Hage N, Podhaizer EM, Sturgill J, Hauser KF. 2011. Toll-like receptor expression and activation in astroglia: differential regulation by HIV-1 Tat, gp120, and morphine. Immunol. Investig. 40, 498–522. ( 10.3109/08820139.2011.561904) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Lamers S, Fogel G, Singer EJ, Salemi M, Nolan DJ, Huysentruyt LC, McGrath MS. 2012. HIV-1 Nef in macrophage-mediated disease pathogenesis. Int. Rev. Immunol. 31, 432–450. ( 10.3109/08830185.2012.737073) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Masanetz S, Lehmann MH. 2011. HIV-1 Nef increases astrocyte sensitivity towards exogenous hydrogenperoxide. Virol. J. 8, 35 ( 10.1186/1743-422X-8-35) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Nath A. 2002. Human immunodeficiency virus (HIV) proteins in neuropathogenesis of HIV dementia. J. Infect. Dis. 7609, 193–198. ( 10.1086/344528) [DOI] [PubMed] [Google Scholar]
  • 92.Churchill MJ, Cowley DJ, Wesselingh SL, Gorry PR, Gray LR. 2015. HIV-1 transcriptional regulation in the central nervous system and implications for HIV cure research. J. Neurovirol. 21, 290–300. ( 10.1007/s13365-014-0271-5) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Levy DN, Refaeli Y, MacGregor RR, Weiner DB. 1994. Serum Vpr regulates productive infection and latency of human immunodeficiency virus type 1. Proc. Natl Acad. Sci. USA 91, 10 873–10 877. ( 10.1073/pnas.91.23.10873) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Hudson L, Liu J, Nath A, Jones M, Raghavan R, Narayan O, Male D, Everall I. 2000. Detection of the human immunodeficiency virus regulatory protein Tat in CNS tissues. J. Neurovirol. 6, 145–155. ( 10.3109/13550280009013158) [DOI] [PubMed] [Google Scholar]
  • 95.Garden GA, et al. 2002. Caspase cascades in human immunodeficiency virus-associated neurodegeneration. J. Neurosci. 22, 4015–4024. ( 10.1523/jneurosci.22-10-04015.2002) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Bachis A, Biggio F, Major EO, Mocchetti I. 2009. M- and T-tropic HIVs promote apoptosis in rat neurons. J. Neuroimmune Pharmacol. 4, 150–160. ( 10.1007/s11481-008-9141-3) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Haughey NJ, Nath A, Mattson MP, Slevin JT, Geiger JD. 2001. HIV-1 Tat through phosphorylation of NMDA receptors potentiates glutamate excitotoxicity. J. Neurochem. 78, 457–467. ( 10.1046/j.1471-4159.2001.00396.x) [DOI] [PubMed] [Google Scholar]
  • 98.King JE, Eugenin EA, Buckner CM, Berman JW. 2006. HIV tat and neurotoxicity. Microbes Infect. 8, 1347–1357. ( 10.1016/j.micinf.2005.11.014) [DOI] [PubMed] [Google Scholar]
  • 99.Kruman II, Nath A, Mattson MP. 1998. HIV-1 protein Tat induces apoptosis of hippocampal neurons by a mechanism involving caspase activation, calcium overload and oxidative stress. Exp. Neurol. 154, 276–288. ( 10.1006/exnr.1998.6958) [DOI] [PubMed] [Google Scholar]
  • 100.Ferrarese C, Aliprandi A, Tremolizzo L, Stanzani L, De Micheli A, Dolara A, Frattola L. 2001. Increased glutamate in CSF and plasma of patients with HIV dementia. Neurology 57, 671–675. ( 10.1212/wnl.57.4.671) [DOI] [PubMed] [Google Scholar]
  • 101.Sami SA, Cicalese S, Ahooyi TM, Khalili K, Amini S, Sariyer IK. 2017. HIV-1 Nef is released in extracellular vesicles derived from astrocytes: evidence for Nef-mediated neurotoxicity. Cell Death Dis. 8, e2542 ( 10.1038/cddis.2016.467) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Wang Y, Santerre M, Tempera I, Martin K, Mukerjee R, Sawaya BE. 2017. HIV-1 Vpr disrupts mitochondria axonal transport and accelerates neuronal aging. Neuropharmacology 117, 364–375. ( 10.1016/j.neuropharm.2017.02.008) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Bateman RJ, et al. 2012. Clinical and biomarker changes in dominantly inherited Alzheimer's disease. N. Engl. J. Med. 367, 795–804. ( 10.1056/NEJMoa1202753) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Marcello E, Epis R, Saraceno C, Di Luca M. 2012. Synaptic dysfunction in Alzheimer's disease. Adv. Exp. Med. Biol. 970, 573–601. ( 10.1007/978-3-7091-0932-8_25) [DOI] [PubMed] [Google Scholar]
  • 105.Adle-Biassette H, Chrétien F, Wingertsmann L, Héry C, Ereau T, Scaravilli F, Tardieu M, Gray F. 1999. Neuronal apoptosis does not correlate with dementia in HIV infection but is related to microglial activation and axonal damage. Neuropathol. Appl. Neurobiol. 25, 123–133. ( 10.1046/j.1365-2990.1999.00167.x) [DOI] [PubMed] [Google Scholar]
  • 106.Avdoshina V, Bachis A, Mocchetti I. 2013. Synaptic dysfunction in human immunodeficiency virus type-1-positive subjects: inflammation or impaired neuronal plasticity? J. Intern. Med. 273, 454–465. ( 10.1111/joim.12050) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Lopardo GD, Bissio E, Iannella Mdel C, Crespo AD, Garone DB, Cassetti LI. 2009. Good neurocognitive performance measured by the international HIV dementia scale in early HIV-1 infection. J. Acquir. Immune Defic. Syndr. 52, 488–492. ( 10.1097/QAI.0b013e3181b06348) [DOI] [PubMed] [Google Scholar]
  • 108.Cysique LAJ, Maruff P, Brew BJ. 2006. Variable benefit in neuropsychological function in HIV-infected HAART-treated patients. Neurology 66, 1447–1450. ( 10.1212/01.wnl.0000210477.63851.d3) [DOI] [PubMed] [Google Scholar]
  • 109.Alfahad T, Nath A. 2013. Retroviruses and amyotrophic lateral sclerosis. Antiviral Res. 99, 180–187. ( 10.1016/j.antiviral.2013.05.006) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Chakradhar S. 2018. A tale of two diseases: aging HIV patients inspire a closer look at Alzheimer's disease. Nat. Med. 24, 376–377. ( 10.1038/nm0418-376) [DOI] [PubMed] [Google Scholar]
  • 111.Milanini B, Valcour V. 2017. Differentiating HIV-associated neurocognitive disorders from Alzheimer's disease: an emerging issue in geriatric NeuroHIV. Curr. HIV/AIDS Rep. 14, 123–132. ( 10.1007/s11904-017-0361-0) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Kang YJ, Digicaylioglu M, Russo R, Kaul M, Achim CL, Fletcher L, Masliah E, Lipton SA. 2010. Erythropoietin plus insulin-like growth factor-I protects against neuronal damage in a murine model of human immunodeficiency virus-associated neurocognitive disorders. Ann. Neurol. 68, 342–352. ( 10.1002/ana.22070) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Cho YE, Lee MH, Song BJ. 2017. Neuronal cell death and degeneration through increased nitroxidative stress and tau phosphorylation in HIV-1 transgenic rats. PLoS ONE 12, e0169945 ( 10.1371/journal.pone.0169945) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Masliah E, Achim CL, Ge N, DeTeresa R, Terry RD, Wiley CA. 1992. Spectrum of human immunodeficiency virus-associated neocortical damage. Ann. Neurol. 32, 321–329. ( 10.1002/ana.410320304) [DOI] [PubMed] [Google Scholar]
  • 115.Mishra M, Taneja M, Malik S, Khalique H, Seth P. 2010. Human immunodeficiency virus type 1 Tat modulates proliferation and differentiation of human neural precursor cells: implication in NeuroAIDS. J. Neurovirol. 16, 355–367. ( 10.3109/13550284.2010.513028) [DOI] [PubMed] [Google Scholar]
  • 116.Ketzler S, Weis S, Haug H, Budka H. 1990. Loss of neurons in the frontal cortex in AIDS brains. Acta Neuropathol. 80, 92–94. ( 10.1007/bf00294228) [DOI] [PubMed] [Google Scholar]
  • 117.Peters PJ, Bhattacharya J, Hibbitts S, Dittmar MT, Simmons G, Bell J, Simmonds P, Clapham PR. 2004. Biological analysis of human immunodeficiency virus type 1 R5 envelopes amplified from brain and lymph node tissues of AIDS patients with neuropathology reveals two distinct tropism phenotypes and identifies envelopes in the brain that confer an enhanced tropism and fusigenicity for macrophages. J. Virol. 78, 6915–6926. ( 10.1128/JVI.78.13.6915-6926.2004) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Vignoli AL, Martini I, Haglid KG, Silvestroni L, Augusti-Tocco G, Biagioni S. 2000. Neuronal glycolytic pathway impairment induced by HIV envelope glycoprotein gp120. Mol. Cell. Biochem. 215, 73–80. ( 10.1023/A:1026590916661) [DOI] [PubMed] [Google Scholar]
  • 119.Vigorito M, LaShomb AL, Chang SL. 2007. Spatial learning and memory in HIV-1 transgenic rats. J. Neuroimmune Pharmacol. 2, 319–328. ( 10.1007/s11481-007-9078-y) [DOI] [PubMed] [Google Scholar]
  • 120.Brew BJ, Pemberton L, Blennow K, Wallin A, Hagberg L. 2005. CSF amyloid b42 and tau levels correlate with AIDS dementia complex. Neurology 65, 1490–1492. ( 10.1212/01.wnl.0000183293.95787.b7) [DOI] [PubMed] [Google Scholar]
  • 121.Patrick C, Crews L, Desplats P, Dumaop W, Rockenstein E, Achim CL, Everall IP, Masliah E. 2011. Increased CDK5 expression in HIV encephalitis contributes to neurodegeneration via tau phosphorylation and is reversed with Roscovitine. Am. J. Pathol. 178, 1646–1661. ( 10.1016/j.ajpath.2010.12.033) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Esiri MM, Biddolph SC, Morris CS. 1998. Prevalence of Alzheimer plaques in AIDS. J. Neurol. Neurosurg. Psychiatry 65, 29–33. ( 10.1136/jnnp.65.1.29) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Green DA, Masliah E, Vinters HV, Beizai P, Moore DJ, Achim CL. 2005. Brain deposition of beta-amyloid is a common pathologic feature in HIV positive patients. AIDS 19, 407–411. ( 10.1097/01.aids.0000161770.06158.5c) [DOI] [PubMed] [Google Scholar]
  • 124.Kim J, Yoon JH, Kim YS. 2013. HIV-1 Tat interacts with and regulates the localization and processing of amyloid precursor protein. PLoS ONE 8, e77972 ( 10.1371/journal.pone.0077972) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Lannuzel A, Tardieu M, Hery C, Barnier JV, Vincent JD, Guibert B, Van Tan H, Gray F. 1997. Human immunodeficiency virus type 1 and its coat protein gp120 induce apoptosis and activate JNK and ERK mitogen-activated protein kinases in human neurons. Ann. Neurol. 42, 847–856. ( 10.1002/ana.410420605) [DOI] [PubMed] [Google Scholar]
  • 126.Kaul M, Lipton SA. 1999. Chemokines and activated macrophages in HIV gp120-induced neuronal apoptosis. Proc. Natl Acad. Sci. USA 96, 8212–8216. ( 10.1073/pnas.96.14.8212) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Costa A, Nappi RE, Polatti F, Poma A, Grossman AB, Nappi G. 2000. Stimulating effect of HIV-1 coat protein gp120 on corticotropin releasing hormone and arginine vasopressin in the rat hypothalamus: involvement of nitric oxide. Exp. Neurol. 166, 376–384. ( 10.1006/exnr.2000.7502) [DOI] [PubMed] [Google Scholar]
  • 128.Chrousos GP, Zapanti ED. 2014. Hypothalamic-pituitary-adrenal axis in HIV infection and disease. Endocrinol. Metab. Clin. North Am. 43, 791–806. ( 10.1016/j.ecl.2014.06.002) [DOI] [PubMed] [Google Scholar]
  • 129.Christeff N, Gherbi N, Mammes O, Dalle MT, Gharakhanian S, Lortholary O, Melchior JC, Nunez EA. 1997. Serum cortisol and DHEA concentrations during HIV infection. Psychoneuroendocrinology 22, S11–S18. ( 10.1016/s0306-4530(97)00015-2) [DOI] [PubMed] [Google Scholar]
  • 130.Kino T. 2000. AIDS/HPA axis. In Endotext (eds De Groot LJ, Chrousos G, Dungan K). South Dartmouth, MA: MDText.com Inc; See www.endotext.org [Google Scholar]
  • 131.Anesten B, Yilmaz A, Hagberg L, Zetterberg H, Nilsson S, Brew BJ, Fuchs D, Price RW, Gisslén M. 2016. Blood-brain barrier integrity, intrathecal immunoactivation and neuronal injury in HIV. Neurol. Neuroimmunol. Neuroinflammation 3, e300 ( 10.1212/nxi.0000000000000300) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Dreyer EB, Lipton SA. 1995. The coat protein gp120 of HIV-1 inhibits astrocyte uptake of excitatory amino acids via macrophage arachidonic acid. Eur. J. Neurosci. 7, 2502–2507. ( 10.1111/j.1460-9568.1995.tb01048.x) [DOI] [PubMed] [Google Scholar]
  • 133.Belmadani A, Zou JY, Schipma MJ, Neafsey EJ, Collins MA. 2001. Ethanol pre-exposure suppresses HIV-1 glycoprotein 120-induced neuronal degeneration by abrogating endogenous glutamate/Ca2+-mediated neurotoxicity. Neuroscience 104, 769–781. ( 10.1016/s0306-4522(01)00139-7) [DOI] [PubMed] [Google Scholar]
  • 134.Clifford DB, Fagan AM, Holtzman DM, Morris JC, Teshome M, Shah AR, Kauwe JSK. 2009. CSF biomarkers of Alzheimer disease in HIV-associated neurologic disease. Neurology 73, 1982–1987. ( 10.1212/WNL.0b013e3181c5b445) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Chen X, Hui L, Geiger NH, Haughey NJ, Geiger JD. 2013. Endolysosome involvement in HIV-1 transactivator protein-induced neuronal amyloid beta production. Neurobiol. Aging 34, 2370–2378. ( 10.1016/j.neurobiolaging.2013.04.015) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Ortega M, Ances BM. 2014. Role of HIV in amyloid metabolism. J. Neuroimmune Pharmacol. 9, 483–491. ( 10.1007/s11481-014-9546-0) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Hategan A, et al. 2017. HIV Tat protein and amyloid-b peptide form multifibrillar structures that cause neurotoxicity. Nat. Struct. Mol. Biol. 24, 379–386. ( 10.1038/nsmb.3379) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Aksenov MY, Aksenova MV, Mactutus CF, Booze RM. 2010. HIV-1 protein-mediated amyloidogenesis in rat hippocampal cell cultures. Neurosci. Lett. 475, 174–178. ( 10.1016/j.neulet.2010.03.073) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Daily A, Nath A, Hersh LB. 2006. Tat peptides inhibit neprilysin. J. Neurovirol. 12, 153–160. ( 10.1080/13550280600760677) [DOI] [PubMed] [Google Scholar]
  • 140.Stern AL, et al. 2018. BACE1 mediates HIV-associated and excitotoxic neuronal damage through an APP-dependent mechanism. J. Neurosci. 38, 4288–4300. ( 10.1523/JNEUROSCI.1280-17.2018) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Zeinolabediny Y, et al. 2017. HIV-1 matrix protein p17 misfolding forms toxic amyloidogenic assemblies that induce neurocognitive disorders. Sci. Rep. 7, 10313 ( 10.1038/s41598-017-10875-0) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Chai Q, Jovasevic V, Malikov V, Sabo Y, Morham S, Walsh D, Naghavi MH. 2017. HIV-1 counteracts an innate restriction by amyloid precursor protein resulting in neurodegeneration. Nat. Commun. 8, 1522 ( 10.1038/s41467-017-01795-8) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Atluri VSR, Hidalgo M, Samikkannu T, Kurapati KRV, Jayant RD, Sagar V, Nair MPN. 2015. Effect of human immunodeficiency virus on blood–brain barrier integrity and function: an update. Front. Cell. Neurosci. 9, 212 ( 10.3389/fncel.2015.00212) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Zhao Z, Nelson AR, Betsholtz C, Zlokovic BV. 2015. Establishment and dysfunction of the blood-brain barrier. Cell 163, 1064–1078. ( 10.1016/j.cell.2015.10.067) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Johnson NA, Jahng GH, Weiner MW, Miller BL, Chui HC, Jagust WJ, Gorno-Tempini ML, Schuff N. 2005. Pattern of cerebral hypoperfusion in Alzheimer disease and mild cognitive impairment measured with arterial spin-labeling MR imaging: initial experience. Radiology 234, 851–859. ( 10.1148/radiol.2343040197) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Marchesi VT. 2011. Alzheimer's dementia begins as a disease of small blood vessels, damaged by oxidative-induced inflammation and dysregulated amyloid metabolism: implications for early detection and therapy. FASEB J. 25, 5–13. ( 10.1096/fj.11-0102ufm) [DOI] [PubMed] [Google Scholar]
  • 147.Rosenberg GA. 2014. Blood-brain barrier permeability in aging and Alzheimers disease. J. Prev. Alzheimers Dis. 1, 138–139. ( 10.14283/jpad.2014.25) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Gosselet F, Saint-Pol J, Candela P, Fenart L. 2013. Amyloid-b peptides, Alzheimer's disease and the blood-brain barrier. Curr. Alzheimer Res. 10, 1015–1033. ( 10.2174/15672050113106660174) [DOI] [PubMed] [Google Scholar]
  • 149.Montagne A, Zhao Z, Zlokovic BV. 2017. Alzheimer's disease: a matter of blood-brain barrier dysfunction? J. Exp. Med. 214, 3151–3169. ( 10.1084/jem.20171406) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Yamazaki Y, Kanekiyo T. 2017. Blood-brain barrier dysfunction and the pathogenesis of Alzheimer's disease. Int. J. Mol. Sci. 18, 1965 ( 10.3390/ijms18091965) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Chen L, Choi JJ, Choi YJ, Hennig B, Toborek M. 2012. HIV-1 Tat-induced cerebrovascular toxicity is enhanced in mice with amyloid deposits. Neurobiol. Aging 33, 1579–1590. ( 10.1016/j.neurobiolaging.2011.06.004) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Chen Y, Huang W, Jiang W, Wu X, Ye B, Zhou X. 2016. HIV-1 Tat regulates occludin and Ab transfer receptor expression in brain endothelial cells via Rho/ROCK signaling pathway. Oxid. Med. Cell. Longev. 2016, 4196572 ( 10.1155/2016/4196572) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Jiang W, Huang W, Chen Y, Zou M, Peng D, Chen D. 2017. HIV-1 transactivator protein induces ZO-1 and neprilysin dysfunction in brain endothelial cells via the ras signaling pathway. Oxid. Med. Cell. Longev. 2017, 3160360 ( 10.1155/2017/3160360) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Kanmogne GD, Schall K, Leibhart J, Knipe B, Gendelman HE, Persidsky Y. 2007. 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. 27, 123–134. ( 10.1038/sj.jcbfm.9600330) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Yang B, Akhter S, Chaudhuri A, Kanmogne GD. 2009. HIV-1 gp120 induces cytokine expression, leukocyte adhesion and transmigration across the blood-brain barrier: modulatory effects of STAT1 signaling. Microvasc. Res. 77, 212–219. ( 10.1016/j.mvr.2008) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Louboutin JP, Reyes BAS, Agrawal L, Maxwell CR, Van Bockstaele EJ, Strayer DS. 2010. Blood-brain barrier abnormalities caused by exposure to HIV-1 gp120–protection by gene delivery of antioxidant enzymes. Neurobiol. Dis. 38, 313–325. ( 10.1016/j.nbd.2010.02.007) [DOI] [PubMed] [Google Scholar]
  • 157.Everall I, et al. 2009. Cliniconeuropathologic correlates of human immunodeficiency virus in the era of antiretroviral therapy. J. Neurovirol. 15, 360–370. ( 10.3109/13550280903131915) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Braak H, Alafuzoff I, Arzberger T, Kretzschmar H, Del Tredici K. 2006. Staging of Alzheimer disease-associated neurofibrillary pathology using paraffin sections and immunocytochemistry. Acta Neuropathol. 112, 389–404. ( 10.1007/s00401-006-0127-z) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Nebuloni M, Pellegrinelli A, Ferri A, Bonetto S, Boldorini R, Vago L, Grassi MP, Costanzi G. 2001. Beta amyloid precursor protein and patterns of HIV p24 immunohistochemistry in different brain areas of AIDS patients. AIDS 15, 571–575. ( 10.1097/00002030-200103300-00005) [DOI] [PubMed] [Google Scholar]
  • 160.Fjell AM, McEvoy L, Holland D, Dale AM, Walhovd KB. 2014. What is normal in normal aging? Effects of aging, amyloid and Alzheimer's disease on the cerebral cortex and the hippocampus. Prog. Neurobiol. 117, 20–40. ( 10.1016/j.pneurobio.2014.02.004) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Piccini A, et al. 2005. beta-amyloid is different in normal aging and in Alzheimer disease. J. Biol. Chem. 280, 34 186–34 192. ( 10.1074/jbc.M501694200) [DOI] [PubMed] [Google Scholar]
  • 162.Jha NK, Jha SK, Kar R, Nand P, Swati K, Goswami VK. 2019. Nuclear factor-kappa β as a therapeutic target for Alzheimer's disease. J. Neurochem. 150, 113–137. ( 10.1111/jnc.14687) [DOI] [PubMed] [Google Scholar]
  • 163.Jha SK, Jha NK, Kumar D, Sharma R, Shrivastava A, Ambasta RK, Kumar P. 2017. Stress-induced synaptic dysfunction and neurotransmitter release in Alzheimer's disease: can neurotransmitters and neuromodulators be potential therapeutic targets? J. Alzheimers Dis. 57, 1017–1039. ( 10.3233/JAD-160623) [DOI] [PubMed] [Google Scholar]
  • 164.Price JL, Davis PB, Morris JC, White DL. 1991. The distribution of tangles, plaques and related immunohistochemical markers in healthy aging and Alzheimer's disease. Neurobiol. Aging 12, 295–312. ( 10.1016/0197-4580(91)90006-6) [DOI] [PubMed] [Google Scholar]
  • 165.Banks WA, Ercal N, Price TO. 2006. The blood–brain barrier in neuroAIDS. Curr. HIV Res. 4, 259–266. ( 10.2174/157016206777709447) [DOI] [PubMed] [Google Scholar]
  • 166.Andras IE, Eum SY, Huang W, Zhong Y, Hennig B, Toborek M. 2010. HIV-1-induced amyloid beta accumulation in brain endothelial cells is attenuated by simvastatin. Mol. Cell. Neurosci. 43, 232–243. ( 10.1016/j.mcn.2009.11.004) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Erickson MA, Banks WA. 2013. Blood–brain barrier dysfunction as a cause and consequence of Alzheimer's disease. J. Cereb. Blood Flow Metab. 33, 1500–1513. ( 10.1038/jcbfm.2013.135) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Gisslen M, et al. 2009. Amyloid and tau cerebrospinal fluid biomarkers in HIV infection. BMC Neurol. 9, 63 ( 10.1186/1471-2377-9-63) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Marra CM, et al. 2009. Impact of combination antiretroviral therapy on cerebrospinal fluid HIV RNA and neurocognitive performance. AIDS 23, 1359–1366. ( 10.1097/QAD.0b013e32832c4152) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Peluso MJ, et al. 2013. Cerebrospinal fluid and neuroimaging biomarker abnormalities suggest early neurological injury in a subset of individuals during primary HIV infection. J. Infect. Dis. 207, 1703–1712. ( 10.1093/infdis/jit088) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Raber J, Huang Y, Ashford JW. 2004. ApoE genotype accounts for the vast majority of AD risk and AD pathology. Neurobiol. Aging 25, 641–650. ( 10.1016/j.neurobiolaging.2003.12.023) [DOI] [PubMed] [Google Scholar]
  • 172.Chang L, Andres M, Sadino J, Jiang CS, Nakama H, Miller E, Ernst T. 2011. Impact of apolipoprotein E epsilon4 and HIV on cognition and brain atrophy: antagonistic pleiotropy and premature brain aging. Neuroimage 58, 1017–1027. ( 10.1016/j.neuroimage.2011.07.010) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Burt TD, et al. 2008. Apolipoprotein (apo) E4 enhances HIV-1 cell entry in vitro, and the APOE epsilon4/epsilon4 genotype accelerates HIV disease progression. Proc. Natl Acad. Sci. USA 105, 8718–8723. ( 10.1073/pnas.0803526105) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Morgan EE, et al. 2013. Apolipoprotein E4 genotype does not increase risk of HIV-associated neurocognitive disorders. J. Neurovirol. 19, 150–156. ( 10.1007/s13365-013-0152-3) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Desplats P, et al. 2013. Molecular and pathologic insights from latent HIV-1 infection in the human brain. Neurology 80, 1415–1423. ( 10.1212/WNL.0b013e31828c2e9e) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Paul RH, Ernst T, Brickman AM, Yiannoutsos CT, Tate DF, Cohen RA, Navia B. 2008. Relative sensitivity of magnetic resonance spectroscopy and quantitative magnetic resonance imaging to cognitive function among nondemented individuals infected with HIV. J. Int. Neuropsychol. Soc. 14, 725–733. ( 10.1017/S1355617708080910) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Minghetti L, Visentin S, Patrizio M, Franchini L, Ajmone-Cat MA, Levi G. 2004. Multiple actions of the human immunodeficiency virus type-1 Tat protein on microglial cell functions. Neurochem. Res. 29, 965–978. ( 10.1023/B:NERE.0000021241.90133.89) [DOI] [PubMed] [Google Scholar]
  • 178.Mollace V, Nottet HS, Clayette P, Turco MC, Muscoli C, Salvemini D, Perno CF. 2001. Oxidative stress and neuroAIDS: triggers, modulators and novel antioxidants. Trends Neurosci. 24, 411–416. ( 10.1016/S0166-2236(00)01819-1) [DOI] [PubMed] [Google Scholar]
  • 179.Harman D, Hendricks S, Eddy DE, Seibold J. 1976. Free radical theory of aging: effect of dietary fat on central nervous system function. J. Am. Geriatr. Soc. 24, 301–307. ( 10.1111/j.1532-5415.1976.tb06800.x) [DOI] [PubMed] [Google Scholar]
  • 180.Mattson MP, Haughey NJ, Nath A. 2005. Cell death in HIV dementia. Cell Death Differ. 12, 893–904. ( 10.1038/sj.cdd.4401577) [DOI] [PubMed] [Google Scholar]
  • 181.Tian C, Sun L, Jia B, Ma K, Curthoys N, Ding J, Zheng J. 2012. Mitochondrial glutaminase release contributes to glutamate-mediated neurotoxicity during human immunodeficiency virus-1 infection. J. Neuroimmune Pharmacol. 7, 619–628. ( 10.1007/s11481-012-9364-1) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Shi Q, Gibson GE. 2007. Oxidative stress and transcriptional regulation in Alzheimer disease. Alzheimer Dis. Assoc. Disord. 21, 276–291. ( 10.1097/WAD.0b013e31815721c3) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Nakamura T, Watanabe A, Fujino T, Hosono T, Michikawa M. 2009. Apolipoprotein E4 (1–272) fragment is associated with mitochondrial proteins and affects mitochondrial function in neuronal cells. Mol. Neurodegener. 4, 35 ( 10.1186/1750-1326-4-35) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Garvey LJ, Pavese N, Politis M, Ramlackhansingh A, Brooks DJ, Taylor-Robinson SD, Winston A. 2014. Increased microglia activation in neurologically asymptomatic HIV infected patients receiving effective ART. AIDS 28, 67–72. ( 10.1097/01.aids.0000432467.54003.f7) [DOI] [PubMed] [Google Scholar]
  • 185.Zhang Y, Wang M, Li H, Zhang H, Shi Y, Wei F, Liu D, Liu K, Chen D. 2012. Accumulation of nuclear and mitochondrial DNA damage in the frontal cortex cells of patients with HIV-associated neurocognitive disorders. Brain Res. 1458, 1–11. ( 10.1016/j.brainres.2012.04.001) [DOI] [PubMed] [Google Scholar]
  • 186.Coughlin JM, et al. 2014. Regional brain distribution of translocator protein using [C]DPA-713 PET in individuals infected with HIV. J. Neurovirol. 20, 219–232. ( 10.1007/s13365-014-0239-5) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Edison P, et al. 2008. Microglia, amyloid, and cognition in Alzheimer's disease: an [11C](R) PK11195-PET and [11C]PIB-PET study. Neurobiol. Dis. 32, 412–419. ( 10.1016/j.nbd.2008.08.001) [DOI] [PubMed] [Google Scholar]
  • 188.Wojda U, Salinska E, Kuznicki J. 2008. Calcium ions in neuronal degeneration. IUBMB Life 60, 575–590. ( 10.1002/iub.91) [DOI] [PubMed] [Google Scholar]
  • 189.Letendre SL, McCutchan JA, Childers ME, Woods SP, Lazzaretto D, Heaton RK, Grant I, Ellis RJ. 2004. Enhancing antiretroviral therapy for human immunodeficiency virus cognitive disorders. Ann. Neurol. 56, 416–423. ( 10.1002/ana.20198) [DOI] [PubMed] [Google Scholar]
  • 190.Harezlak J, et al. 2011. Persistence of HIV-associated cognitive impairment, inflammation, and neuronal injury in era of highly active antiretroviral treatment. AIDS 25, 625–633. ( 10.1097/QAD.0b013e3283427da7) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Brenchley JM, et al. 2006. Microbial translocation is a cause of systemic immune activation in chronic HIV infection. Nat. Med. 12, 1365–1371. ( 10.1038/nm1511) [DOI] [PubMed] [Google Scholar]
  • 192.Lyn-Cook LE, Jr, et al. 2009. Hepatic ceramide may mediate brain insulin resistance and neurodegeneration in type 2 diabetes and non-alcoholic steatohepatitis. J. Alzheimers Dis. 16, 715–729. ( 10.3233/JAD-2009-0984) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Devlin KN, Gongvatana A, Clark US, Chasman JD, Westbrook ML, Tashima KT, Navia B, Cohen RA. 2012. Neurocognitive effects of HIV, hepatitis C, and substance use history. J. Int. Neuropsychol. Soc. 18, 68–78. ( 10.1017/S1355617711001408) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Gongvatana A, et al. 2011. Clinical contributors to cerebral white matter integrity in HIV-infected individuals. J. Neurovirol. 17, 477–486. ( 10.1007/s13365-011-0055-0) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Nix LM, Tien PC. 2014. Metabolic syndrome, diabetes, and cardiovascular risk in HIV. Curr. HIV/AIDS Rep. 11, 271–278. ( 10.1007/s11904-014-0219-7) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.El-Sadr WM, Mullin CM, Carr A, Gibert C, Rappoport C, Visnegarwala F, Grunfeld C, Raghavan SS. 2005. Effects of HIV disease on lipid, glucose and insulin levels: results from a large antiretroviral-naive cohort. HIV Med. 6, 114–121. ( 10.1111/j.1468-1293.2005.00273.x) [DOI] [PubMed] [Google Scholar]
  • 197.Roriz-Filho SJ, Sá-Roriz TM, Rosset I, Camozzato AL, Santos AC, Chaves ML, Moriguti J, Roriz-Cruz M. 2009. (Pre)diabetes, brain aging, and cognition. Biochim. Biophys. Acta 1792, 432–443. ( 10.1016/j.bbadis.2008.12.003) [DOI] [PubMed] [Google Scholar]
  • 198.Yaffe K. 2007. Metabolic syndrome and cognitive disorders: is the sum greater than its parts? Alzheimer Dis. Assoc. Disord. 21, 167–171. ( 10.1097/WAD.0b013e318065bfd6) [DOI] [PubMed] [Google Scholar]
  • 199.Cohen RA, et al. 2009. Vascular and cognitive functions associated with cardiovascular disease in the elderly. J. Clin. Exp. Neuropsychol. 31, 96–110. ( 10.1080/13803390802014594) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Correia SC, Santos RX, Carvalho C, Cardoso S, Candeias E, Santos MS, Oliveira CR, Moreira PI. 2012. Insulin signaling, glucose metabolism and mitochondria: major players in Alzheimer's disease and diabetes interrelation. Brain Res. 1441, 64–78. ( 10.1016/j.brainres.2011.12.063) [DOI] [PubMed] [Google Scholar]
  • 201.Izycka-Swieszewska E, Zoltowska A, Rzepko R, Gross M, Borowska-Lehman J. 2000. Vasculopathy and amyloid beta reactivity in brains of patients with acquired immune deficiency (AIDS). Folia Neuropathol. 38, 175–182. [PubMed] [Google Scholar]
  • 202.Nelson L, Gard P, Tabet N. 2014. Hypertension and inflammation in Alzheimer's disease: close partners in disease development and progression!. J. Alzheimers Dis. 41, 331–343. ( 10.3233/JAD-140024) [DOI] [PubMed] [Google Scholar]
  • 203.Ru W, Tang SJ. 2017. HIV-associated synaptic degeneration. Mol. Brain 10, 40 ( 10.1186/s13041-017-0321-z) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Canet G, et al. 2018. HIV neuroinfection and Alzheimer's disease: similarities and potential links? Front. Cell. Neurosci. 12, 307 ( 10.3389/fncel.2018.00307) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Bachis A, Major EO, Mocchetti I. 2003. Brain-derived neurotrophic factor inhibits human immunodeficiency virus-1/gp120-mediated cerebellar granule cell death by preventing gp120 internalization. J. Neurosci. 23, 5715–5722. ( 10.1523/JNEUROSCI.23-13-05715.2003) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Macdonald NJ, Perez-Polo JR, Bennett AD, Taglialatela G. 1999. NGF-resistant PC12 cell death induced by arachidonic acid is accompanied by a decrease of active PKC zeta and nuclear factor kappa B. J. Neurosci. Res. 57, 219–226. () [DOI] [PubMed] [Google Scholar]
  • 207.Ramirez SH, Sanchez JF, Dimitri CA, Gelbard HA, Dewhurst S, Maggirwar SB. 2001. Neurotrophins prevent HIV Tat-induced neuronal apoptosis via a nuclear factor-kappaB (NF-kappaB)-dependent mechanism. J. Neurochem. 78, 874–889. ( 10.1046/j.1471-4159.2001.00467.x) [DOI] [PubMed] [Google Scholar]
  • 208.Brandoli C, Sanna A, De Bernardi MA, Follesa P, Brooker G, Mocchetti I. 1998. Brain-derived neurotrophic factor and basic fibroblast growth factor downregulate NMDA receptor function in cerebellar granule cells. J. Neurosci. 18, 7953–7961. ( 10.1523/JNEUROSCI.18-19-07953.1998) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Romsi P, et al. 2002. Potential neuroprotective benefits of erythropoietin during experimental hypothermic circulatory arrest. J. Thorac. Cardiovasc. Surg. 124, 714–723. ( 10.1067/mtc.2002.123704) [DOI] [PubMed] [Google Scholar]
  • 210.Gassmann M, Heinicke K, Soliz J, Ogunshola OO, Marti HH, Hofer T, Grimm C, Heinicke I, Egli B. 2003. Non-erythroid functions of erythropoietin. Adv. Exp. Med. Biol. 543, 323–330. ( 10.1007/978-1-4419-8997-0_22) [DOI] [PubMed] [Google Scholar]
  • 211.Digicaylioglu M, Kaul M, Fletcher L, Dowen R, Lipton SA. 2004. Erythropoietin protects cerebrocortical neurons from HIV-1/gp120-induced damage. Neurorepor 15, 761–763. ( 10.1097/00001756-200404090-00004) [DOI] [PubMed] [Google Scholar]
  • 212.Digicaylioglu M, Lipton SA. 2001. Erythropoietin-mediated neuroprotection involves cross-talk between Jak2 and NF-kappaB signalling cascades. Nature 412, 641–647. ( 10.1038/35088074) [DOI] [PubMed] [Google Scholar]
  • 213.Semkova I, Schilling M, Henrich-Noack P, Rami A, Krieglstein J. 1996. Clenbuterol protects mouse cerebral cortex and rat hippocampus from ischemic damage and attenuates glutamate neurotoxicity in cultured hippocampal neurons by induction of NGF. Brain Res. 717, 44–54. ( 10.1016/0006-8993(95)01567-1) [DOI] [PubMed] [Google Scholar]
  • 214.Semkova I, Krieglstein J. 1999. Neuroprotection mediated via neurotrophic factors and induction of neurotrophic factors. Brain Res. Brain Res. Rev. 30, 176–188. ( 10.1016/S0165-0173(99)00013-2) [DOI] [PubMed] [Google Scholar]
  • 215.Chiaretti A, Piastra M, Polidori G, Di Rocco C, Caresta E, Antonelli A, Amendola T, Aloe L. 2003. Correlation between neurotrophic factor expression and outcome of children with severe traumatic brain injury. Intensive Care Med. 29, 1329–1338. ( 10.1007/s00134-003-1852-6) [DOI] [PubMed] [Google Scholar]
  • 216.Jain S, Golde DW, Bailey R, Geffner ME. 1998. Insulin-like growth factor-I resistance. Endocr. Rev. 19, 625–646. [DOI] [PubMed] [Google Scholar]
  • 217.Rondanelli M, et al. 2002. Insulin-like growth factor I (IGF-I) and IGF-binding protein 3 response to growth hormone is impaired in HIV-infected children. AIDS Res. Hum. Retrovir. 18, 331–339. ( 10.1089/088922202753519106) [DOI] [PubMed] [Google Scholar]
  • 218.Ying WJ, Peruzzi F, Lassak A, Del Valle L, Radhakrishnan S, Rappaport J, Khalili K, Amini S, Reiss K. 2003. Neuroprotective effects of IGF-I against TNFalpha-induced neuronal damage in HIV-associated dementia. Virolog 305, 66–76. ( 10.1006/viro.2002.1690) [DOI] [PubMed] [Google Scholar]
  • 219.Everall IP, Trillo-Pazos G, Bell C, Mallory M, Sanders V, Masliah E. 2001. Amelioration of neurotoxic effects of HIV envelope protein gp120 by fibroblast growth factor: a strategy for neuroprotection. J. Neuropathol. Exp. Neurol. 60, 293–301. ( 10.1093/jnen/60.3.293) [DOI] [PubMed] [Google Scholar]
  • 220.Everall IP, Bell C, Mallory M, Langford D, Adame A, Rockestein E, Masliah E. 2002. Lithium ameliorates HIV-gp120-mediated neurotoxicity. Mol. Cell. Neurosci. 21, 493–501. ( 10.1006/mcne.2002.1196) [DOI] [PubMed] [Google Scholar]
  • 221.Jain KK. 2000. Evaluation of memantine for neuroprotection in dementia. Expert Opin. Investig. Drug 9, 1397–1406. ( 10.1517/13543784.9.6.1397) [DOI] [PubMed] [Google Scholar]
  • 222.Bormann J. 1989. Memantine is a potent blocker of N-methyl-daspartate (NMDA) receptor channels. Eur. J. Pharmacol. 166, 591–592. ( 10.1016/0014-2999(89)90385-3) [DOI] [PubMed] [Google Scholar]
  • 223.Anderson ER, Gendelman HE, Xiong H. 2004. Memantine protects hippocampal neuronal function in murine human immunodeficiency virus type 1 encephalitis. J. Neurosci. 24, 7194–7198. ( 10.1523/JNEUROSCI.1933-04.2004) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.Maggirwar SB, Tong N, Ramirez S, Gelbard HA, Dewhurst S. 1999. HIV-1 Tat-mediated activation of glycogen synthase kinase-3beta contributes to Tat-mediated neurotoxicity. J. Neurochem. 73, 578–586. ( 10.1046/j.1471-4159.1999.0730578.x) [DOI] [PubMed] [Google Scholar]
  • 225.Tong N, Sanchez JF, Maggirwar SB, Ramirez SH, Guo H, Dewhurst S, Gelbard HA. 2001. Activation of glycogen synthase kinase 3 beta (GSK-3beta) by platelet activating factor mediates migration and cell death in cerebellar granule neurons. Eur. J. Neurosci. 13, 1913–1922. ( 10.1046/j.0953-816x.2001.01572.x) [DOI] [PubMed] [Google Scholar]
  • 226.McManus CM, Brosnan CF, Berman JW. 1998. Cytokine induction of MIP-1 alpha and MIP-1 beta in human fetal microglia. J. Immunol. 160, 1449–1455. [PubMed] [Google Scholar]
  • 227.Wang EJ, et al. 2003. Microglia from mice transgenic for a provirus encoding a monocyte- tropic HIV type 1 isolate produce infectious virus and display in vitro and in vivo upregulation of lipopolysaccharide induced chemokine gene expression. AIDS Res. Hum. Retrovir. 19, 755–765. ( 10.1089/088922203769232557) [DOI] [PubMed] [Google Scholar]
  • 228.Eugenin EA, D'Aversa TG, Lopez L, Calderon TM, Berman JW. 2003. MCP-1 (CCL2) protects human neurons and astrocytes from NMDA or HIV-tat-induced apoptosis. J. Neurochem. 85, 1299–1311. ( 10.1046/j.1471-4159.2003.01775.x) [DOI] [PubMed] [Google Scholar]
  • 229.Kelder W, McArthur JC, Nance-Sproson T, McClernon D, Griffin DE. 1998. Beta-chemokines MCP-1 and RANTES are selectively increased in cerebrospinal fluid of patients with human immunodeficiency virus-associated dementia. Ann. Neurol. 44, 831–835. ( 10.1002/ana.410440521) [DOI] [PubMed] [Google Scholar]
  • 230.Tozzi V, et al. 2007. Persistence of neuropsychologic deficits despite long-term highly active antiretroviral therapy in patients with HIV-related neurocognitive impairment: prevalence and risk factors. J. Acquir. Immune Defic. Syndr. 45, 174–182. ( 10.1097/QAI.0b013e318042e1ee) [DOI] [PubMed] [Google Scholar]
  • 231.Letendre S, et al. 2008. Validation of the CNS penetration-effectiveness rank for quantifying antiretroviral penetration into the central nervous system. Arch. Neurol. 65, 65–70. ( 10.1001/archneurol.2007.31) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.Caniglia EC, et al. 2014. HIV-CAUSAL Collaboration. Antiretroviral penetration into the CNS and incidence of AIDS-defining neurologic conditions. Neurology 83, 134–141. ( 10.1212/WNL.0000000000000564) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233.Cysique LA, Vaida F, Letendre S, Gibson S, Cherner M, Woods SP, McCutchan JA, Heaton RK, Ellis RJ. 2009. Dynamics of cognitive change in impaired HIV-positive patients initiating antiretroviral therapy. Neurology 73, 342–348. ( 10.1212/WNL.0b013e3181ab2b3b) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234.Robertson KR, Su Z, Margolis DM, Krambrink A, Havlir DV, Evans S, Skiest DJ, A5170 Study Team. 2010. Neurocognitive effects of treatment interruption in stable HIV-positive patients in an observational cohort. Neurology 74, 1260–1266. ( 10.1212/WNL.0b013e3181d9ed09) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 235.Department of Health and Human Services. 2016. Guidelines for the use of antiretroviral agents in HIV-infected adults and adolescents. See https://aidsinfo.nih.gov/contentfiles/lvguidelines/adultandadolescentgl.pdf (accessed on 16 July 2016). [DOI] [PubMed]
  • 236.Nowacek A, Gendelman H. 2009. NanoART, neuroAIDS and CNS drug delivery. Nanomedicine 4, 557–574. ( 10.2217/nnm.09.38) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237.Bleasby K, et al. 2006. Expression profiles of 50 xenobiotic transporter genes in humans and pre-clinical species: a resource for investigations into drug disposition. Xenobiotica 36, 963–988. ( 10.1080/00498250600861751) [DOI] [PubMed] [Google Scholar]
  • 238.Ene L, Duiculescu D, Ruta SM. 2011. How much do antiretroviral drugs penetrate into the central nervous system? J. Med. Life 4, 432–439. [PMC free article] [PubMed] [Google Scholar]
  • 239.Fiandra L, Capetti A, Sorrentino L, Corsi F. 2017. Nanoformulated antiretrovirals for penetration of the central nervous system: state of the art. J. Neuroimmune Pharmacol. 12, 17–30. ( 10.1007/s11481-016-9716-3) [DOI] [PubMed] [Google Scholar]
  • 240.Saksena NK, Wang B, Zhou L, Soedjono M, Ho YS, Conceicao V. 2010. HIV reservoirs in vivo and new strategies for possible eradication of HIV from the reservoir sites. HIV/AIDS 2, 103–122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241.Varatharajan L, Thomas SA. 2009. The transport of anti-HIV drugs across blood-CNS interfaces: summary of current knowledge and recommendations for further research. Antivir. Res. 82, A99–A109. ( 10.1016/j.antiviral.2008.12.013) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 242.Strazielle N, Ghersi-Egea J-F. 2005. Factors affecting delivery of antiviral drugs to the brain. Rev. Med. Virol. 15, 105–133. ( 10.1002/rmv.454) [DOI] [PubMed] [Google Scholar]
  • 243.Letendre SL, Ellis RJ, Ances BM, McCutchan JA. 2010. Neurologic complications of HIV disease and their treatment. Top HIV Med. 18, 45–55. [PMC free article] [PubMed] [Google Scholar]
  • 244.Best BM, et al. 2009. Low Atazanavir concentrations in cerebrospinal fluid. AIDS 23, 83–87. ( 10.1097/QAD.0b013e328317a702) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 245.De Oliveira FTM, do Olival GS, de Oliveira ACP. 2015. Central nervous system antiretroviral high penetration therapy. J. AIDS Clin. Res. 6, 12. [Google Scholar]
  • 246.Aquaro S, et al. 1997. Inhibition of replication of HIV in primary monocyte/macrophages by different antiviral drugs and comparative efficacy in lymphocytes. J. Leukoc. Biol. 62, 138–143. ( 10.1002/jlb.62.1.138) [DOI] [PubMed] [Google Scholar]
  • 247.Kaur IP, Bhandari R, Bhandari S, Kakkar V. 2008. Potential of solid lipid nanoparticles in brain targeting. J. Control. Release 127, 97–109. ( 10.1016/j.jconrel.2007.12.018) [DOI] [PubMed] [Google Scholar]
  • 248.Lai F, Fadda AM, Sinico C. 2013. Liposomes for brain delivery. Exp. Opin. Drug Deliv. 10, 1003–1022. ( 10.1517/17425247.2013.766714) [DOI] [PubMed] [Google Scholar]
  • 249.Saiyed ZM, Gandhi NH, Nair MPN. 2010. Magnetic nanoformulation of azidothymidine 50-triphosphate for targeted delivery across the blood-brain barrier. Int. J. Nanomed. 5, 157–166. ( 10.2147/IJN.S8905) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 250.Liu D, Lin B, Shao W, Zhu Z, Ji T, Yang C. 2014. In vitro and in vivo studies on the transport of PEGylated silica nanoparticles across the blood–brain barrier. ACS Appl. Mater. Interfaces 6, 2131–2136. ( 10.1021/am405219u) [DOI] [PubMed] [Google Scholar]
  • 251.Cheng Y, et al. 2014. Blood–brain barrier permeable gold nanoparticles: an efficient delivery platform for enhanced malignant glioma therapy and imaging. Small 29, 5137–5150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 252.Borgmann K, Rao K, Labhasetwar V, Ghorpade A. 2011. Efficacy of Tat-conjugated ritonavir-loaded nanoparticles in reducing HIV-1 replication in monocyte-derived macrophages and cytocompatibility with macrophages and human neurons. AIDS Res. Hum. Retrovir. 27, 853–862. ( 10.1089/aid.2010.0295) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 253.Akay C, et al. 2014. Antiretroviral drugs induce oxidative stress and neuronal damage in the central nervous system. J. Neurovirol. 20, 39–53. ( 10.1007/s13365-013-0227-1) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 254.Akay-Espinoza C, Stern AL, Nara RL, Panvelker N, Li J, Jordan-Sciutto K-L. 2017. Differential in vitroneurotoxicity of antiretroviral drugs. In Proceedings of the Conference on Retroviruses and Opportunistic Infections (CROI), Seattle, WA, USA, 13–16 February 2017. [Google Scholar]
  • 255.Robertson K, Liner J, Meeker RB. 2012. Antiretroviral neurotoxicity. J. Neurovirol. 18, 388–399. ( 10.1007/s13365-012-0120-3) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 256.Rabkin JG, Rabkin R, Wagner G. 1994. Effects of fluoxetine on mood and immune status in depressed patients with HIV illness. J. Clin. Psychiatry 55, 92–97. [PubMed] [Google Scholar]
  • 257.Perkins DO, Stern RA, Golden RN, Murphy C, Naftolowitz D, Evans DL. 1994. Mood disorders in HIV infection: prevalence and risk factors in a nonepicenter of the AIDS epidemic. Am. J. Psychiatry 151, 233–236. ( 10.1176/ajp.151.2.299) [DOI] [PubMed] [Google Scholar]
  • 258.Hinkin CH, Castellon SA, Hardy DJ, Farinpour R, Newton T, Singer E. 2001. Methylphenidate improves HIV-1-associated cognitive slowing. J. Neuropsychiatry Clin. Neurosci. 13, 248–254. ( 10.1176/jnp.13.2.248) [DOI] [PubMed] [Google Scholar]
  • 259.Sewell DD, Jeste DV, Atkinson JH, Heaton RK, Hesselink JR, Wiley C, Thal L, Chandler JL, Grant I. 1994. HIV-associated psychosis: a study of 20 cases. San Diego HIV neurobehavioral research center group. Am. J. Psychiatry 151, 237–242. ( 10.1176/ajp.151.2.237) [DOI] [PubMed] [Google Scholar]
  • 260.Parenti DM, Simon GL, Scheib RG, Meyer WA, Sztein MB, Paxton H, DiGioia RA, Schulof RS. 1988. Effect of lithium carbonate in HIV-infected patients with immune dysfunction. J. Acquir. Immune Defic. Syndr (1988). 1, 119–124. [PubMed] [Google Scholar]
  • 261.Halman MH, Worth JL, Sanders KM, Renshaw PF, Murray GB. 1993. Anticonvulsant use in the treatment of manic syndromes in patients with HIV-1 infection. J. Neuropsychiatry Clin. Neurosci. 5, 430–434. ( 10.1176/jnp.5.4.430) [DOI] [PubMed] [Google Scholar]
  • 262.Dubé B, Benton T, Cruess DG, Evans DL. 2005. Neuropsychiatric manifestations of HIV infection and AIDS. J. Psychiatry Neurosci. 30, 237–246. [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

This article has no additional data.


Articles from Open Biology are provided here courtesy of The Royal Society

RESOURCES