Next Article in Journal
Towards the Laboratory Maintenance of Haemagogus janthinomys (Dyar, 1921), the Major Neotropical Vector of Sylvatic Yellow Fever
Next Article in Special Issue
Virally Suppressed People Living with HIV Who Use Opioids Have Diminished Latency Reversal
Previous Article in Journal
Inactivation of Ebola Virus and SARS-CoV-2 in Cell Culture Supernatants and Cell Pellets by Gamma Irradiation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Role of Autophagy in HIV-1 and Drug Abuse-Mediated Neuroinflammaging

1
Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
2
Centre for Excellence in Nanobio Translational Research, Anna University, BIT Campus, Tiruchirappalli 620024, Tamil Nadu, India
3
Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
4
Cedars-Sinai Medical Center, Smidt Heart Institute, Los Angeles, CA 90048, USA
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Viruses 2023, 15(1), 44; https://doi.org/10.3390/v15010044
Submission received: 10 November 2022 / Revised: 15 December 2022 / Accepted: 20 December 2022 / Published: 23 December 2022
(This article belongs to the Special Issue HIV and Drugs of Abuse 2.0)

Abstract

:
Chronic low-grade inflammation remains an essential feature of HIV-1 infection under combined antiretroviral therapy (cART) and contributes to the accelerated cognitive defects and aging in HIV-1 infected populations, indicating cART limitations in suppressing viremia. Interestingly, ~50% of the HIV-1 infected population on cART that develops cognitive defects is complicated by drug abuse, involving the activation of cells in the central nervous system (CNS) and neurotoxin release, altogether leading to neuroinflammation. Neuroinflammation is the hallmark feature of many neurodegenerative disorders, including HIV-1-associated neurocognitive disorders (HAND). Impaired autophagy has been identified as one of the underlying mechanisms of HAND in treated HIV-1-infected people that also abuse drugs. Several lines of evidence suggest that autophagy regulates CNS cells’ responses and maintains cellular hemostasis. The impairment of autophagy is associated with low-grade chronic inflammation and immune senescence, a known characteristic of pathological aging. Therefore, autophagy impairment due to CNS cells, such as neurons, microglia, astrocytes, and pericytes exposure to HIV-1/HIV-1 proteins, cART, and drug abuse could have combined toxicity, resulting in increased neuroinflammation, which ultimately leads to accelerated aging, referred to as neuroinflammaging. In this review, we focus on the potential role of autophagy in the mechanism of neuroinflammaging in the context of HIV-1 and drug abuse.

Graphical Abstract

1. Introduction

The invention of combined antiretroviral therapy (cART) as a pharmaceutical intervention has improved the average life expectancy of people living with HIV-1 (PLWH), which is comparable to healthy individuals [1,2,3]. This therapeutic intervention has transformed HIV-1/acquired immunodeficiency syndrome (AIDS; formerly considered a death sentence), with its short post-diagnosis average lifespan, into a manageable chronic disorder like diabetes mellitus [4]. Despite the promising prospects of cART, the quality of life offered by this therapy is threatened by the prevalence of mild cognitive decline and loss of memory, commonly referred to as HIV-1-associated neurocognitive disorders (HAND), which afflicts 30–60% of PLWH [1,2,3]. The presence of comorbidities, such as diabetes mellitus, cardiovascular disorders, atherosclerosis, and neurological decline, is prevalent, especially among aged PLWH [5]. According to the Centers for Disease Control and Prevention, the increasing number of individuals diagnosed with HIV-1 infection aged 50 and above doubled in 2009 (~33%) from 2001 (~17%) [4,5]. The expectation for an increased diagnostic rate within this age range has thus created an emergency public health challenge [4,5]. The emergence of physiological changes has been implicated as a factor that increases the risk for accompanying comorbidities in aged HIV-1-infected individuals. This complexity aggravates even PLWH on cART therapy [4]. The contributory role of the physiological alterations employed by HIV-1 infection, chronic effects of residual HIV-1 proteins in low penetrating tissues, pain management dependence, and cART treatment remain largely unknown. Although, the constant immune activation occurring as a result of chronic low-level inflammation observed in PLWH is likely to accelerate aging phenotypes among those individuals [6,7,8,9,10,11,12,13,14,15,16]. HIV-1 has been well-documented to impact the immune system and the brain, which can be observed in about 8–15% development of severe HIV-1-associated dementia, an incidence significantly reduced by cART [4]. The efficacy of cART in reducing viremia, and the increase in the average quality of life of the HIV-infected population, have not prevented the continued presence of chronic low-level inflammation associated with accelerated aging and neurodegeneration [4,5]. The neurotoxic impact of cART and its potential effects on premature aging and inflammation are well-characterized [17].
The comorbidity of substance abuse (cocaine, methamphetamine [meth], opiates), with respect to HAND, has further complicated matters for PLWH [5]. This drug abuse is an essential driving factor of HIV-1 infection worldwide, as both are linked and have been shown to promote premature aging [18,19]. Accumulating evidence points to abuse of drugs in approximately 30% of PLWH afflicted with HAND [20,21]. Furthermore, there are also implications of exacerbated neurological aging in these individuals abusing drugs [20,21]. Recent reports indicate that individuals, including patients on cART therapy, who abuse drugs present higher viral load accelerate HIV-1 progression and AIDS-related opportunistic infection [4]. The progression of HIV-1 infection in individuals who abuse drugs is exacerbated by their non-compliance to the prescribed cART, causing recurrent viral activation [22]. This accelerated HIV-1 progression is due to the accessibility of the blood–brain barrier to the virus, thereby triggering the activation of glial cells and neurotoxins released by drug abuse, leading to the mediation of neuroinflammation [23]. The continued presence of low-level inflammation, especially in the central nervous system (CNS), is the primary driver of accelerated aging in PLWH [4,5]. Recent reports have shown a direct linkage between neuroinflammation and the accelerated aging process based on enhanced activation of glial cells in the aged population [5]. This indicates that many mechanisms, such as upregulated endoplasmic reticulum (ER) stress, reactive oxygen species (ROS), dysregulated toll-like receptor signaling (TLR) [24,25,26], and NOD-like receptor proteins (NLRPs) inflammasome activation, have been implicated as part of the molecular details of HIV-1, drug abuse, and cART effects on the CNS [27]. In addition to their addictive effects, drugs of use such as cocaine, morphine, meth, and alcohol have also been reported to be involved in caspase-mediated cell death, mitochondrial dysfunction, activation of transcription factors like cAMP response element-binding protein, and early genes (c-fos and c-jun), neuronal injury, glial activation, and neuroinflammation [28,29,30,31,32,33,34,35,36,37,38,39]. Additionally, cellular dysfunction and neuroinflammation can result from the dysregulation of autophagy by HIV-1/HIV-1 proteins, drug abuse, and cART [5].
Autophagy is an evolutionarily conversed process characterized by cellular degradation and recycling intracellular macromolecules into their constituent subunits in the lysosome [40,41]. Three morphologically distinct primary types of autophagy (macroautophagy, microautophagy, and chaperone-mediated autophagy) exist, but they all culminate in the delivery of cargo to the lysosome for degradation and recycling [41]. Macroautophagy is characterized by the protrusion or invagination of the membranes of a lysosome to capture and sequester cargo and intact organelles, while microautophagy sequesters cargo away from the lysosome [41,42]. Chaperone-mediated autophagy differs from the two primary types of autophagy by using chaperones to identify cargo proteins that contain a particular pentapeptide motif, which is unfolded and individually translocated across lysosomal membranes [42]. Of the three, macroautophagy is the most studied, as it involves the de novo synthesis of double-membrane vesicles (termed autophagosomes) to sequester cargo proteins/organelles and eventually transport them across the lysosome for degradation [43]. Autophagy is a highly regulated and conserved process that involves multiple autophagy-related proteins, transcriptional modification, post-translational modulations, and complex protein–protein interactions [40]. Autophagy mechanisms involve the phosphorylation of Unc-51-like autophagy activating kinase (ULK1/2) by upstream kinase that regulates its preinitiation complex [5]. This leads to the activation of class III phosphoinositide 3-kinase (PI3K), an autophagy initiation complex via the ULK1/2 phosphorylation, and B-cell lymphoma 2 (Bcl-2) to beclin 1 disruptive binding. This activation generates phosphatidylinositol-3-phosphate at the nucleation site in isolated membranes, causing multiple protein binding to form autophagosomes in the elongation step. The formed autophagosome is further stabilized by other proteins, such as E1 ligase, E2 ligase, and E3 ligase complex [5]. The maturation involves the autophagosome fusion with lysosomes to form autolysosome, degrading sequestered contents and releasing them to the cytoplasm for recycling [40]. The maturation step is characterized by changes in the adaptor protein, p62 expression [5].

2. Interplay of Autophagy, Inflammation, and Aging–HIV and Drug Abuse

Autophagy is an inherent cellular defense mechanism that is highly regulated and involved in degrading cytoplasmic components using lysosomes. The autophagosome and lysosome play the central role in autophagy, in which the autophagosome, a double-membrane organelle, engulfs cellular components/debris and transports it to a lysosome to break down using lysosomal enzymes [44]. In presenting debris to a lysosome, an autophagosome is not selective—it engulfs cellular organelles, protein aggregates, damaged proteins, and disease-causing pathogens [45].
Autophagy is made operational in a cell by fasting (deprivation of energy to cell), which makes the autophagosome release the engulfed components to provide whatever they have for the survival of cells in these deprived conditions [46]. This process of autophagy is highly coordinated and demands a fine-tuning balance when operational inside the cell, as any dysregulation in it causes cancer, infectious diseases, accelerated aging, and neurodegenerative disorders [47,48,49]. Figure 1 describes the interplay among drug abuse, HIV-1/HIV-1 proteins, and cART in the context of autophagy, cellular activation, and neuroinflammation.

2.1. Role of Autophagy in HIV and Drug Abuse

Autophagy has a profound role in maintaining nutrient levels inside the cells and in infectious diseases like HIV-1. HIV-1 cross-talks with autophagy in that the viral envelope causes autophagy-dependent cell death by binding to the C-X-C chemokine receptor type 4 (CXCR-4) on T cells [50]. Additionally, autophagy assists HIV-1 replication by playing a role in CD4+ T cells and macrophages and its involvement in HIV-1 pathogenesis [51]. There is repression of autophagy in CD4+ T cells infected with HIV-1; however, autophagy is highly activated in infected monocytes/macrophages [51]. Autophagy has been shown to counter the effect of HIV-1 Tat in CD4+ T lymphocytes by causing the degradation of HIV-1 viral particles [52]. HIV-1 has evolved to avoid autophagy-mediated degradation, for which many reasons remain unknown. One of the reasons this occurs is that the HIV-1 Nef interacts with Beclin-1, which in turn blocks the maturation of monocytes/macrophages to cause autophagy, and the HIV-1 avoids getting degraded [53]. Another mechanism the HIV-1 utilizes to evade the autophagy-mediated degradation is activating the mammalian target of rapamycin (MTOR) in dendritic cells, making CD4+ T cells more likely to become infected with the HIV-1. Both mechanisms mentioned above used by HIV-1 could be potential therapeutic targets for drug discoveries, and many other possible means of HIV-1-camouflaging cellular autophagy could be discovered in the future [54]. Drug abuse involving methamphetamine, cocaine, morphine, and alcohol has been reported to compromise the immune system, putting abusers significantly at higher risk of HIV-1 infection [55]. In addition, abusers are highly likely to acquire an HIV-1 infection because their mental illness acts as a precursor for aberrant behaviors, including unprotected sex and needle sharing, which increases the risk of getting HIV-1. There is an increase in autophagy in drug abuse subjects, confirmed by the presence of multi-membrane vacuoles in various cell types [56], which is a hallmark of autophagy. Hence, autophagy, HIV-1 infection, and drug abuse seem to go hand in hand.

2.2. Role of Inflammation in HIV-1 and Drug Abuse

During systemic inflammation, an increase in C-reactive protein (CRP) and serum amyloid A (SAA) are two critical indicators of inflammation known as acute phase proteins. The crucial link between inflammation, HIV-1, and drug abuse is that CRP and SAA affect inflammatory cytokines and immune dysfunction (which trigger HIV-1-associated neuropathogenesis progression) [57,58,59]. The acute-phase protein CRP binds strongly to phosphocholine residues and causes a shift in plasma lipoproteins constituents [60]. On the other hand, SAA induces the proinflammatory cytokines tumor necrosis factor-α (TNFα), interleukin (IL) 6, and IL4 [61]. Additionally, reports have shown that increased CRP and SAA in HIV-1 infection contribute to immune suppression and progression [62,63,64]. Furthermore, it has also been reported that drugs of abuse (methamphetamine, cocaine, alcohol) and HIV-1 infection have an increased risk of developing chronic neuroinflammation, enhanced immune dysfunction (predisposition to HIV-1 infection), and cognitive impairment [65,66,67,68,69,70]. This has been demonstrated in studies on various CNS cells (neurons, glial cells, and pericytes) exposed to HIV-1/HIV-1 proteins and drugs of abuse, which led to increased expression of proinflammatory cytokines (such as TNFα, IL1β, and IL6) in both the intracellular and extracellular compartments [24,25,30,38,65,71,72,73]. Additionally, it has also been reported that exposure of neurons and glia to HIV-1 proteins and drugs of abuse, such as cocaine, resulted in dysregulated autophagy, which was characterized by increased formation of autophagosomes, the defective fusion of the autophagosome with the lysosomes as well as defective auto/mitophagy [24,25,30,38,65,71,72,73]. Notably, the ability of the CNS cells to mediate crosstalk involving the extracellular vesicles also promotes progressive neuroinflammation and neuronal aging in the context of HIV-1 and drugs of abuse [74,75,76,77,78,79]. Cellular crosstalk propagating neuroinflammation is also mediated by proinflammatory cytokines released by the donor cells, which, in turn, can impact the functioning of the recipient cells [76,77,78,79,80].

2.3. Role of Aging in HIV-1 and Drug Abuse

Aging has been associated with drug abuse, and PLWH has further high usage of taking substances of abuse, including alcohol, tobacco, methamphetamine, and other drugs [81]. Aged subjects’ health is impacted by comorbid conditions as the drugs they abuse could easily interact with other medications they use for survival and cause loss of effectiveness of the medication, including cART in aged HIV-1 patients [82]. In addition, drug abuse in aged HIV-1 subjects demonstrates a significantly lower ability to cause viral suppression (ability to combat viral infection), leading to further aggravation of poor health, which becomes one of the causes of their mortality [83]. Therefore, an essential connection among aging, HIV-1, and drug abuse is that advanced age increases susceptibility to HAND in PLWH, who, later in their cognitive impairment, tend to use drugs more and therefore lose capacity to fight secondary infections [84]. It has been also reported that aged people who are active in sex, perform different (anal) sexual practices, and have unprotected sex, puts them in a high-risk group of contracting an HIV-1 infection [85]. What further aggravates it is that these active, aged, sex subjects are also users of abusive drugs, which significantly complicates aging.

3. Microglia, Autophagy, and Aging

Microglia and brain resident macrophages play a significant role in the innate immune system of the CNS and orchestrate inflammatory responses in the brain due to damage in the CNS. Microglia regulate the release of both pro- or anti-inflammatory cytokines and are involved in the phagocytosis of debris in the CNS [86,87]. Microglia are also known to play diverse roles in maintaining brain health and functions, including pruning of synaptic structures, remodeling of the perisynaptic environment, neuronal plasticity, modulation of neurogenesis, and the release of neurotropic soluble factors, as well as cytokines in the brain [88,89,90]. Based on the nature of the activating stimulus, the microglia respond differently, and release cytokines and chemokines related to either pro- or anti-inflammation. Activation of microglia to the “classic M1 polarization” releases proinflammatory mediators and contributes to neuronal and other glial cell damage. In contrast, the activation of the alternative M2 polarization limits inflammation by yielding anti-inflammatory factors and supports neurons via secreting neuronal growth factors [87,91,92]. The activation of proinflammatory response-as well as chronic altered neuroinflammatory status, including changes in microglial morphology, phenotype, and activity-have been implicated as a potential mechanism of neurodegeneration and progression of age-associated neurodegenerative disease, including Parkinson’s disease (PD), Alzheimer’s disease (AD), and amyotrophic lateral sclerosis (ALS) [93,94,95,96]. Several lines of evidence have confirmed that the autophagy pathway regulates innate and adaptive responses and the outcome of inflammatory reactions, phagocytosis, and antigen presentation in macrophages and microglia [97,98,99]. Intriguingly, it has been documented that impaired autophagy flux in macrophages is well associated with low-grade chronic inflammation and immune senescence, known characteristic features of aging [100,101,102].
Microglia activation-mediated low-grade chronic inflammation has been known to be associated with the progression of “inflammaging” in the CNS and the peripheral tissues [101,103,104,105]. Intriguingly, microglia derived from the brains of aged mice displayed increased basal levels of proinflammatory cytokines, including TNFα, IL1β, and IL6 [106]. Furthermore, studies from different models, including humans, rodents, canines, and non-human primates, showed age-associated microglial priming and an increased inflammatory profile of the CNS, such as major histocompatibility complex II, the cluster of differentiation molecule (CD) 11b, scavenger receptor CD68, CD11c integrins, TLRs and co-stimulatory molecule CD86 in the aged brain [95,107,108,109,110,111,112,113]. Intriguingly, the microglia in aged brain dogs, gerbils, and mice showed an activated morphology of a de-ramified structure, along with an increased ionized calcium-binding adaptor protein-1 (Iba1), a protein expressed on the surface of microglia in young adults [114,115]. Autophagy dysfunction was significantly associated with microglial activation and neuroinflammation [38,92,116,117], which contributed to neuronal damage and the progression of age-related neurodegenerative diseases [98,103,118]. A recent study reported that modulation of the autophagy pathway regulates the polarization of microglia [92]. TNFα exposure directed the microglia towards the M1 phenotype, as evidenced by increased M1 marker (inducible nitric oxide synthase [iNOS], IL1β, and IL6) expression and the reduction of M2 marker (Arginase1, and IL10), which is associated with impaired autophagy flux. It has been documented that those microglia exposed to TNFα promoted the microglia polarization towards M2 upon upregulation of autophagy, either via serum deprivation or pharmacological activators, such as rapamycin and resveratrol. On the other hand, pharmacological or genetic autophagy inhibitors using 3-methyladenine (3-MA) or autophagy-related (ATG) 5 siRNA consistently aggravated the M1 polarization in microglia challenged with TNFα [92]. Recent studies from our group have confirmed that either cocaine or HIV-1 Tat stimulates microglial activation and neuroinflammation due to increased mitochondrial dysfunction (along with lysosomal dysfunction), which results in defective clearance of damaged mitochondria via the mitophagy (selective mitochondrial autophagy) pathway [38,117]. Another study from our group also documented that an antiretroviral drug cause increased microglial activation via increased permeabilization of lysosomes and impaired autophagy flux [119]. A recent study also showed that lipopolysaccharide (LPS)-induced neuroinflammation in N9 microglial cells associated with the inhibition of autophagic flux through activating the PI3KI/AKT/MTOR pathway; however, enhanced microglial autophagy downregulates LPS-induced neuroinflammation [120].
Apart from an inflammatory response, microglia also phagocytize the different types of brain-derived cargo, including neuronal and myelin debris, apoptotic cells, protein deposits, and other substrates. Autophagy plays a significant role in the clearance of those engulfed cargo via lysosomal fusion and degradation [86,98,121]. Studies have shown that increased accumulation of misplaced or altered molecules, resulting from damaged and dead cells and organelles (cell debris) due to defective auto/mitophagy, could activate receptors of an innate immune system and neuroinflammation, in turn accelerating the aging process [122,123,124,125]. A recent study reported that microglia-mediated protection of neurons involved synucleinphagy, where the α-synuclein released from neurons was removed by microglia. Synucleinphagy is mediated via TLR4/NF-κB/p62 signaling pathway in microglial cells of mice expressing human α-synuclein in the neurons [126]. Another study by Pomilio et al. [127] documented that prolonged or chronic exposure of microglia to β-amyloid (Aβ) peptides resulted in impaired autophagy in microglial cells both in vitro and in vivo, as well as in AD patients, in turn, contributing to the progression of the disease. It has also been reported that increased mitophagy using nicotinamide adenine dinucleotide supplementation diminished expression of insoluble Aβ1–42 and Aβ1–40 forms via increased microglial phagocytosis of extracellular Aβ plaques, in turn, ameliorating neuroinflammation and cognitive impairment in an APP/PS1 mouse model. Furthermore, this study also documented that the induction of mitophagy and reversal of cognitive impairment involved the PTEN-induced kinase-1 (PINK-1) and parkin-dependent pathways [128]. There is also evidence that alleviating impaired autophagy flux via pharmacological or genetic approaches prevented microglia activation and neuroinflammation [38,71,116,117,129,130], ultimately preventing neuroinflammation.

4. Astrocytes, Autophagy, and Aging

Astrocytes are the most abundant CNS cell type and have a multitude of functions in the brain, including maintenance of cellular homeostasis. Additionally, autophagy in astrocytes is believed to play a vital role in the pathogenesis of aging and neurodegenerative diseases. Several reports have shown that autophagy can influence inflammation, oxidative stress, and the aging process in different cell types, and interrelation among the different factors have been well-studied, but in relation to astrocytes are small. It has been reported that activation of astrocytes is an early event in PLWH [131] and in animal models of NeuroHIV [132]. Earlier studies showed that activation of the autophagic signaling pathway was required for HIV-1 replication, and an inhibition in this pathway resulted in attenuation of viral replication [133,134]. This autophagy dysregulation is considered essential for HIV-1 replication and can also be associated with HAND [135]. HIV-1 proteins have also been shown to regulate astrocytic functions via direct modulation of autophagy pathways [136]. For instance, HIV-1 gp120 has been shown to play a pro-survival role against astrocyte cell death via induction of the autophagy pathway [136]. It has also been observed that HIV-1 Tat can induce autophagy by upregulation of a Bcl-2-associated athanogene 3 (BAG3) and NF-κB signaling pathway [137]. Abundant expression of the HIV-1 viral protein Nef was observed in HIV-1-infected astrocytes [138], and this Nef protein was shown to induce neurocognitive impairments in rats [139,140]. Overexpression of Nef in human astrocytes resulted in the expression of key molecules of the autophagy pathway, e.g., ATG8, LC3II, and p62 [138]. Exposure of astrocytes with autophagy inhibitors resulted in significant release of Nef in the extracellular vesicles (EVs) derived from primary human fetal astrocytes, which, upon being uptaken by the neurons, resulted in inhibition of neuronal action potential and therefore induced neurotoxicity [141]. Some other reports have shown that HIV-1 suppresses autophagy by disrupting the conversion of LC-I to LC3-II and induces toxicity in astrocytes [134]. Another study has shown that mitophagy is crucial for cell death resistance in HIV-1 productively infected astrocytes, but its impairment may favor inflammasome-mediated cell death in abortively infected cells [142]. These studies show the importance of the autophagic pathway in astrocytes with HIV-1 or viral proteins.
Drugs of abuse, such as opiates, can potentiate HIV-1 infectivity. Opioids have been shown to enhance HIV-1 replication and interact with opioid receptors in astrocytes, inducing the release of astrocyte-derived proinflammatory factors and potentiate neurodegeneration in HIV-1 patients on opiates [143,144], and can result in cognitive impairment [145]. Drugs of abuse, such as cocaine and methamphetamine, have also been implicated in potentiating HIV-1-induced neuropathogenesis [146,147,148]. Periyasamy et al. reported that cocaine-mediated dysregulated autophagy led to astrocytosis, which resulted in increased expression of proinflammatory mediators, such as TNF, IL1β, and IL6, leading to neuroinflammation [25]. Another study has shown that cocaine-induced autophagy led to type II programmed cell death in astrocytes [149]. Methamphetamine was also shown to induce autophagy in astrocytes via the opioid receptor and metabotropic glutamate receptor subtype 5 receptor-mediated signaling, whereas inhibition of autophagy exacerbated methamphetamine-induced apoptotic cell death [136]. Co-treatment of astrocytes with HIV-1 and morphine resulted in increased expression of p62, indicating blockage in the fusion of autophagosomes with lysosomes, resulting in the induction of inflammation [150]. Studies by Sil et al. demonstrated that morphine exposure to human astrocytes results in the induction of an ER stress-induced autophagy pathway, which leads to astrocytosis and neuroinflammation [151]. All these studies showed that HIV-1 alone, or in combination with different drugs of abuse, can induce autophagy-induced neuroinflammation in astrocytes, which can contribute to the process of inflammaging.
Aging is characterized by loss of regenerative capacity (which leads to morbid conditions) and has been considered a comorbid condition in HIV-1 and substance abuse disorders [4,5]. Studies have shown that structural and metabolic parameters of the brain decline at relatively younger ages in HIV-1-infected apolipoprotein E4 (ApoE4) carriers, leading to cognitive deficit and inflammation [152,153]. However, neuropsychological studies on HIV-1-infected cohorts have produced conflicting results regarding the impact of ApoE4 or the Apoε4 allele on the course of cognition. During normal aging, ApoE4 contributes to cognitive impairment by transcriptional repression for some genes related to cognitive reserve, and the same effect is observed in aging HIV-1 populations [154,155]. Studies on serum from HIV-1-infected patients have demonstrated the association between inflammatory markers, such as C-X-C motif chemokine ligand 1 and transforming growth factor (TGF) α, and increased telomere length (indicative of aging). Furthermore, IL-10RA on neurons was associated with decreased telomere length [156], thus indicating inflammaging. Studies have shown that antiretroviral therapy induces cell cycle arrest, senescence-associated beta-galactosidase, and the cell cycle inhibitor p21 in astrocytes. Further studies have also shown that anti-retroviral-induced astrocytic senescence can be associated with oxidative stress and metabolic changes that could play a role in the development of HAND [157].
Several drugs of abuse have been shown to potentiate HIV-1 infectivity and inflammation via the autophagy pathway, so their role in HIV-1-induced aging will be essential to be considered. It was reported that self-administration of cocaine reduced the uptake of neuron-derived EVs (NDEVs) by the astrocytes, primarily in the motor cortex, which can be effectively reversed by extinction training [158]. One study demonstrated that HIV-1 and methamphetamine induce senescence of primary human astrocytes, as evaluated by induction of senescence markers (β-galactosidase and p16), senescence-associated morphologic changes, as well as cell cycle arrest via the β-catenin pathway. HIV-1 and methamphetamine-mediated astrocyte senescence was also demonstrated in three small animal models (the humanized mouse model of HIV/NSG-huPBMCs, HIV-transgenic rats, and in a methamphetamine administration rat model). Senescent astrocytes, in turn, caused neuronal toxicity. Thus, the β-catenin pathway can be a potential therapeutic target to overcome astrocyte senescence [159]. Drugs of abuse like cocaine, morphine, and methamphetamine can induce inflammation as a result of the constant and consistent release of proinflammatory cytokines [25,151,160,161]. They can also potentiate inflammatory status in HIV-1 conditions [162,163], which can contribute to the process of inflammaging and will be an essential aspect to study in addicted HIV-1 patients undergoing premature aging. Autophagy has been closely associated with aging, i.e., the autophagic pathway was shown to decline during aging [164]. Inhibition of the target of rapamycin complex 1 (TORC1), which is a known negative regulator of autophagy, enhances lifespan [165]. The exercise was shown to downregulate TORC1 activity and upregulation of autophagy in different tissues and thus may represent practical means to promote longevity [165]. Recently, it was reported that there were reductions in autophagic flux in C. elegans, which corroborates with organismal lifetime since there was an age-dependent accumulation of autophagosomes, thus decreasing autophagic degradation [166]. Therefore, autophagy is linked with aging. There are scanty reports on HIV-1 and drug-mediated autophagy linked to aging, but further investigation on the role of autophagy in aging will open promising avenues for anti-aging drug development.

5. Neurons, Autophagy, and Aging

Aging leads to neuronal alterations–such as changes in neuronal morphology, neuronal damage, and dysregulation of neuronal signaling pathways–which also contribute to neuroinflammation and aging. Emerging studies have demonstrated that neuronal autophagy plays an essential role in neuroinflammation, aging, and HIV-1 dementia [167,168,169]. Atg7 is an E1-like enzyme required for autophagosome biogenesis [170]. To understand the role of neuronal autophagy in aging and age-related disease, Rudnick et al. generated motor neuron-specific autophagy-deficient mice by crossing Atg7flox/flox mice to ChAT-Cre mice (Atg7 cKO) [171]. Interestingly, inhibition of motor neuron autophagy significantly extends lifespan in Atg7 cKO mice compared with Atg7 cWT mice [171]. A study from the same group demonstrated that TANK-Binding Kinase 1 (TBK1) is active in ubiquitinated inclusions in motor neurons and is directly involved in the autophagy pathway that targets protein aggregates for clearance [116]. These findings thus indicate that autophagy plays both beneficial and detrimental roles in aging and age-related diseases.
In transgenic mice expressing exon 1 of the human HD (Huntington’s disease) gene carrying a CAG repeat expansion, Petersén et al. demonstrated that the mutant striatal neurons exhibited elevated cell death and increased autophagic granules and electron-dense lysosomes in response to dopamine treatment compared with wild-type neurons [172]. These findings suggest that the combinatorial effect of mutant neurons in the Huntington gene and dopamine-induced oxyradical stress promotes autophagy, which could, in turn, lead to cell death underlying age-related diseases. In line with these findings, Purkinje neuron death has also been characterized by excessive autophagic-lysosomal vacuolation [173]. Florez-McClure et al. demonstrated that trophic factor withdrawal induces a marked increase in the sizes of autophagic and lysosomal vacuoles, resulting in a nonapoptotic death pathway in Purkinje neurons [173]. Moreover, antisense oligonucleotides to the p75 neurotrophin receptor (p75ntr) decrease autophagy and inhibit trophic factor withdrawal-induced Purkinje neuron death [173]. The authors further demonstrated that the p75ntr supports Purkinje neuron survival in the presence of trophic factors but promotes autophagy and death of Purkinje neurons in the absence of trophic stimuli [173]. Emerging studies suggest that HIV+ve/HD patients exhibit an earlier onset of HD symptoms than HIV−ve/HD patients [174,175,176]. Studies also demonstrated that in these patients, dysregulation of neuronal autophagy was associated with HIV-1 dementia and encephalitis [167,168]. As the brain is a target of various drugs of abuse, including morphine, which results in various cellular signaling alterations [177,178], it is not surprising that increased neuronal loss was found in HIV-1 Tat and morphine-administered Becn1+/+ mice but not in Becn1+/− mice [169,179]. These findings also suggest a role of autophagy in HIV-1 Tat and morphine-mediated neuronal damage. However, whether and how neuronal autophagy is involved in the earlier onset of HD in HIV+ve/HD patients on drugs warrants further investigation.
High-mobility group box 1 (HMGB1) is a nuclear protein and can be released into the extracellular space to act as an inflammatory mediator [180,181]. Zhu et al. demonstrated that extracellular HMGB1 induces autophagy in primary spinal neurons underlying spinal root avulsion [182]. This was further demonstrated that HMGB1-induced autophagy increases the survival of primary spinal neurons under oxygen-glucose deprivation conditions [182]. These findings suggest a beneficial role of neuronal autophagy. However, in another study, Yin et al. demonstrated that inhibiting autophagy by salidroside protected neonatal neurons from glutamate-induced apoptotic cell death [183]. A study by Tripathi et al. also showed that impairment of autophagy in human motor neurons induced by reactive astrocytes secreted TGF-β1 and led to ALS-like axonal and cytoplasmic protein aggregation in the neurons [184]. Similarly, Lang et al. found that dopaminergic neurons significantly enhanced autophagy in PD mice [185]. The authors further demonstrated that the upregulation of long non-coding RNA HOTAIR promoted autophagy in mid-brain dopaminergic neurons of the substantia nigra compacta in PD mice [185].
To examine whether motor neuron autophagy plays a role in amyotrophic lateral sclerosis (ALS), Tian et al. generated a double transgenic (DTg) mouse model by crossing green fluorescent protein (GFP)-fused LC3 transgenic (LC3-Tg) mice with G93A mutant human Cu/Zn superoxide dismutase (mSOD1) transgenic (mSOD1-Tg) mice [186]. Imaging analysis revealed a solid increased autophagic signal in both the spinal motor neurons and surrounding glial cells [186]. The authors also found that the number of neurons displaying autophagy in the DTg mice reduced at the end symptomatic stage, with decreased numbers of living motor neurons [186]. These results suggest that autophagy-mediated motor neuronal death could be involved in the progress of ALS.
Hypothalamic autophagy reduces during aging [187]. Loss of autophagy in hypothalamic proopiomelanocortin (POMC) neurons have been shown to impair the generation of α melanocyte-stimulating hormone (α-MSH) and lipolysis [188]. These data suggest that POMC neuronal autophagy plays an essential role in maintaining energy balance, whereas reduced autophagy in POMC neurons could lead to metabolic disturbances observed during aging [188]. Mice with specific deletion of autophagy-related 7 (Atg7), an essential autophagy gene, in hypothalamic POMC neurons (Atg7ΔPOMC mice) exhibited increased food intake and decreased energy expenditure with a concomitant increase in body weight [189]. These findings thus demonstrated the critical role of autophagy in POMC neurons in controlling appetite balance and energy homeostasis [189]. Increasing evidence suggests that autophagy plays a vital role in neuroinflammaging. Interestingly, studies have revealed diverse roles for autophagy in neurons (depending on the type of neurons), the status of the disease, and the stage of age. Therefore, outcomes of investigation on neuronal autophagy could pave the way for the future development of neuronal autophagy-targeted strategies in the prevention and treatment of neuroinflammaging.

6. The Role of Autophagy in Aging-Associated Blood–Brain barrier (BBB) Disruption

BBB is a tight barrier critical for preventing pathogens and small molecules from entering the brain. It is composed of endothelial cells, pericytes, and end-feet of astrocytes [190]. Disruption of the BBB contributes to the pathogenesis of many CNS diseases, including PD [191], AD [192], HAND [193], etc. Moreover, the breakdown of BBB was considered an early hallmark in the aging brain [190] and recognized as an early biomarker of cognitive impairment in AD [194]. As conveyed in previous sections, autophagy plays a crucial role in cellular/organismic aging, age-related dysfunction, and disease. Impairment of vascular autophagy could contribute to the development of oxidative stress, inflammation, and vascular dysfunction with aging through the loss of ability to remove dysfunctional proteins/organelles and to recycle damaged biomolecules that interfere with normal cellular function. Although evidence suggests that autophagy declines with aging in numerous tissues [195,196], its contribution to the development of vascular dysfunction with aging, mechanisms involved, and aging-associated neurological disease are largely unknown. We will provide a brief overview of previous findings on the role of autophagy underlying age-associated BBB dysfunction.

6.1. Endothelial Cells

The tight junction (TJ) complexes are molecules that interact in the extracellular junctional space and act as anchors between endothelial cells [197], maintaining the barrier functions of BBB. Tight junctions are integral membrane proteins, including claudins, occludin, junctional adhesion molecules (JAM)-A, -B, -C, and scaffolding proteins like zonula occludens (ZO-1, ZO-2, and ZO-3) [198]. During aging, it has been demonstrated that the expression of BBB tight junction proteins, occludin-1, and ZO-1, were significantly attenuated in aged mice compared to young mice [199]. Additionally, the number of tight junctions decreasing with advanced age was also reported in rats, resulting in increased BBB permeability [200]. The permeability of BBB is closely associated with the expression levels of these tight junction proteins. The function of tight junctions’ regulating permeability is based on two pathways: (1) the “pore” pathway that can carry small uncharged solutes and specific ions and (2) the “leak” pathway that can carry non-charged larger molecules [201]. The TJ membrane proteins, such as claudins (such as claudin-2), have been shown to play a crucial role in regulating the cation-selective pore pathway, while the barrier-forming protein occludin is known to regulate the leak pathway. Induction of autophagy has been shown to result in the degradation of claudin-2, leading to the enhancement of the TJ barrier function with increased transepithelial resistance. In a recent study, exposure of primary human brain microvascular endothelial cells to HIV-1 Tat-induced colocalization of LC3B puncta with ZO-1 and HIV-1 Tat-mediated downregulation of ZO-1 was attenuated by pretreating the cells with pharmacological inhibitors of autophagy, as well as by silencing ATG5. Collectively, these findings underpin the role of autophagy in the HIV-1 Tat-mediated downregulation of ZO-1, which, in turn, leads to increased permeability of BBB [202].

6.2. Pericytes

Pericytes, essential constituents of the cerebrovascular unit, play a crucial role in maintaining the integrity of the blood–brain barrier [203]. Several studies have demonstrated pericyte coverage is positively correlated with the integrity of BBB [204] and modulating the influx of immune cells [205,206], controlling blood flow, etc. [203]. Emerging studies have suggested that pericytes may control critical neurovascular functions, and pericyte loss may play a role in age-dependent vascular-mediated neurodegeneration [205]. Additionally, pericytes were found to regulate capillary blood flow in the aging mouse brain, which might be the mechanism underlying aging-associated neurovascular dysfunction [207]. However, the mechanism by which age-dependent pericyte dysfunction regulates blood flow and the mechanisms of aging-associated pericyte loss is still mostly unclear. Recently, Yuan Zhang et al. reported that the knockdown of the sigma-1 receptor (σ-1R) resulted in pericyte loss via increasing autophagy and apoptosis, which, in turn, led to BBB disruption and thus contributed to cerebral infarction [208]. Furthermore, they found YZ001 (σ-1R agonist) -alleviated pericyte loss via inhibition of autophagy, resulting from σ-1R activation, indicating a novel role of σ-1R in mediating pericyte survival through regulating the interplay between apoptosis and autophagy. While pericytes have also been reported to be productively infected with HIV-1 [209], studies are warranted to investigate the role of these cells in regulating autophagy and aging in the context of BBB disruption. Overall, future studies are needed to explore the involvement of the autophagy pathway in aging-mediated pericyte disruption, which could provide a possible therapeutic strategy for aging-associated neurological diseases.

7. Conclusions

Overall, defective or blocked autophagy is an essential target in activating CNS cells such as microglia, astrocytes, and neurons, leading to the continued release of proinflammatory cytokines. Various reports implicate defective autophagy as an upstream signaling pathway in the generation of the inflammatory response, leading to neuroinflammation and neuroinflammaging. Interestingly, membrane phagophore emerge in the first place when specific cellular cargo is targeted for degradation, and alternatively, independent Atg5/Atg7 mechanisms are regulated in the context of HIV-1, and drug abuse remains unknown. Interactions aimed at blocking pathways involved in HIV-1 and a drug abuse-mediated role in defective autophagy could be considered a future therapeutic strategy to dampen the effects of sustained inflammation caused by these CNS assaults. The use of rapamycin or hypoxia-inducible factor-dependent target genes that reduce mTOR activity, which leads to the induction of autophagy, ensures that cells adapt to their changing environment, eliminate toxic episodes, and promote cell viability, can be considered as a potential target for restoring cognitive deficits associated with neurodegenerative disorders and neuroinflammaging. Thus, autophagy could emerge as a potent modulator of neuroinflammaging in HIV-1 and drug abuse-mediated pathologies.

Author Contributions

Resources, P.P. and S.B.; writing—original draft preparation, S.S., A.T., A.O., G.H., N.A.K., K.L. and P.P.; writing—review and editing, S.S., A.T., A.O., G.H., N.A.K., K.L., S.B. and P.P.; supervision, S.B. and P.P.; funding acquisition, S.B. and P.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by NIH grants: DA050545, DA044586, DA047156, DA043138, and DA052266. The support by Chronic HIV infection and Aging in NeuroAIDS (CHAIN; MH062261) and Nebraska Center for Substance Abuse Research (NCSAR) are also highly acknowledged.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We would like to thank all members (past and present) of our laboratories for their invaluable contributions and apologize to all those collaborators whose work could not be discussed in this review owing to space limitations. We also thank Amy Sather, Editing/Project Specialist, Department of Pharmacology and Experimental Neuroscience, for the editorial assistance.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. McLaurin, K.A.; Booze, R.M.; Mactutus, C.F. Diagnostic and prognostic biomarkers for HAND. J. Neurovirol. 2019, 25, 686–701. [Google Scholar] [CrossRef] [PubMed]
  2. Paul, R. Neurocognitive Phenotyping of HIV in the Era of Antiretroviral Therapy. Curr. HIV/AIDS Rep. 2019, 16, 230–235. [Google Scholar] [CrossRef] [PubMed]
  3. Alford, K.; Vera, J.H. Cognitive Impairment in people living with HIV in the ART era: A Review. Br. Med. Bull. 2018, 127, 55–68. [Google Scholar] [CrossRef] [PubMed]
  4. Sil, S.; Niu, F.; Chivero, E.T.; Singh, S.; Periyasamy, P.; Buch, S. Role of Inflammasomes in HIV-1 and Drug Abuse Mediated Neuroinflammaging. Cells 2020, 9, 1857. [Google Scholar] [CrossRef]
  5. Guo, M.L.; Buch, S. Neuroinflammation & pre-mature aging in the context of chronic HIV infection and drug abuse: Role of dysregulated autophagy. Brain Res. 2019, 1724, 146446. [Google Scholar] [CrossRef]
  6. Bonnet, F.; Amieva, H.; Marquant, F.; Bernard, C.; Bruyand, M.; Dauchy, F.A.; Mercie, P.; Greib, C.; Richert, L.; Neau, D.; et al. Cognitive disorders in HIV-infected patients: Are they HIV-related? AIDS 2013, 27, 391–400. [Google Scholar] [CrossRef]
  7. Deeks, S.G. HIV infection, inflammation, immunosenescence, and aging. Annu. Rev. Med. 2011, 62, 141–155. [Google Scholar] [CrossRef] [Green Version]
  8. Deeks, S.G.; Tracy, R.; Douek, D.C. Systemic effects of inflammation on health during chronic HIV infection. Immunity 2013, 39, 633–645. [Google Scholar] [CrossRef] [Green Version]
  9. Guaraldi, G.; Prakash, M.; Moecklinghoff, C.; Stellbrink, H.J. Morbidity in older HIV-infected patients: Impact of long-term antiretroviral use. AIDS Rev. 2014, 16, 75–89. [Google Scholar]
  10. Heaton, R.K.; Clifford, D.B.; Franklin, D.R., Jr.; Woods, S.P.; Ake, C.; Vaida, F.; Ellis, R.J.; Letendre, S.L.; Marcotte, T.D.; Atkinson, J.H.; et al. HIV-associated neurocognitive disorders persist in the era of potent antiretroviral therapy: CHARTER Study. Neurology 2010, 75, 2087–2096. [Google Scholar] [CrossRef] [Green Version]
  11. High, K.P.; Brennan-Ing, M.; Clifford, D.B.; Cohen, M.H.; Currier, J.; Deeks, S.G.; Deren, S.; Effros, R.B.; Gebo, K.; Goronzy, J.J.; et al. HIV and aging: State of knowledge and areas of critical need for research. A report to the NIH Office of AIDS Research by the HIV and Aging Working Group. J. Acquir. Immune Defic. Syndr. 2012, 60 (Suppl. S1), S1–S18. [Google Scholar] [CrossRef] [PubMed]
  12. Leng, S.X.; Margolick, J.B. Aging, sex, inflammation, frailty, and CMV and HIV infections. Cell Immunol. 2020, 348, 104024. [Google Scholar] [CrossRef] [PubMed]
  13. Palmer, C.S.; Palchaudhuri, R.; Albargy, H.; Abdel-Mohsen, M.; Crowe, S.M. Exploiting immune cell metabolic machinery for functional HIV cure and the prevention of inflammaging. F1000Research 2018, 7, 125. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Skiest, D.J. The importance of co-morbidity in older HIV-infected patients. AIDS 2003, 17, 1577. [Google Scholar] [CrossRef] [PubMed]
  15. Steele, A.K.; Lee, E.J.; Vestal, B.; Hecht, D.; Dong, Z.; Rapaport, E.; Koeppe, J.; Campbell, T.B.; Wilson, C.C. Contribution of intestinal barrier damage, microbial translocation and HIV-1 infection status to an inflammaging signature. PLoS ONE 2014, 9, e97171. [Google Scholar] [CrossRef]
  16. Sundermann, E.E.; Hussain, M.A.; Moore, D.J.; Horvath, S.; Lin, D.T.S.; Kobor, M.S.; Levine, A.; Group, H. Inflammation-related genes are associated with epigenetic aging in HIV. J. Neurovirol. 2019, 25, 853–865. [Google Scholar] [CrossRef]
  17. Holt, J.L.; Kraft-Terry, S.D.; Chang, L. Neuroimaging studies of the aging HIV-1-infected brain. J. Neurovirol. 2012, 18, 291–302. [Google Scholar] [CrossRef]
  18. Chang, L.; Shukla, D.K. Imaging studies of the HIV-infected brain. Handb. Clin. Neurol. 2018, 152, 229–264. [Google Scholar] [CrossRef]
  19. Vera, J.H.; Ridha, B.; Gilleece, Y.; Amlani, A.; Thorburn, P.; Dizdarevic, S. PET brain imaging in HIV-associated neurocognitive disorders (HAND) in the era of combination antiretroviral therapy. Eur J. Nucl. Med. Mol. Imaging 2017, 44, 895–902. [Google Scholar] [CrossRef] [Green Version]
  20. Campbell, A.N.; Tross, S.; Calsyn, D.A. Substance use disorders and HIV/AIDS prevention and treatment intervention: Research and practice considerations. Soc. Work Public Health 2013, 28, 333–348. [Google Scholar] [CrossRef]
  21. Dahal, S.; Chitti, S.V.; Nair, M.P.; Saxena, S.K. Interactive effects of cocaine on HIV infection: Implication in HIV-associated neurocognitive disorder and neuroAIDS. Front. Microbiol. 2015, 6, 931. [Google Scholar] [CrossRef] [PubMed]
  22. Brooks, R.W.; Robbins, P.D. Treating Age-Related Diseases with Somatic Stem Cells. Adv. Exp. Med. Biol. 2018, 1056, 29–45. [Google Scholar] [CrossRef] [PubMed]
  23. Kumar, S.; Vijayan, M.; Bhatti, J.S.; Reddy, P.H. MicroRNAs as Peripheral Biomarkers in Aging and Age-Related Diseases. Prog Mol. Biol. Transl. Sci. 2017, 146, 47–94. [Google Scholar] [CrossRef]
  24. Periyasamy, P.; Liao, K.; Kook, Y.H.; Niu, F.; Callen, S.E.; Guo, M.L.; Buch, S. Cocaine-Mediated Downregulation of miR-124 Activates Microglia by Targeting KLF4 and TLR4 Signaling. Mol. Neurobiol. 2018, 55, 3196–3210. [Google Scholar] [CrossRef] [PubMed]
  25. Periyasamy, P.; Guo, M.L.; Buch, S. Cocaine induces astrocytosis through ER stress-mediated activation of autophagy. Autophagy 2016, 12, 1310–1329. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Chivero, E.T.; Guo, M.L.; Periyasamy, P.; Liao, K.; Callen, S.E.; Buch, S. HIV-1 Tat Primes and Activates Microglial NLRP3 Inflammasome-Mediated Neuroinflammation. J. Neurosci. 2017, 37, 3599–3609. [Google Scholar] [CrossRef] [Green Version]
  27. Parzych, K.R.; Klionsky, D.J. An overview of autophagy: Morphology, mechanism, and regulation. Antioxid. Redox Signal. 2014, 20, 460–473. [Google Scholar] [CrossRef] [Green Version]
  28. Asuni, G.P.; Speidell, A.; Mocchetti, I. Neuronal apoptosis induced by morphine withdrawal is mediated by the p75 neurotrophin receptor. J. Neurochem. 2021, 158, 169–181. [Google Scholar] [CrossRef]
  29. Calma, I.D.; Persons, A.L.; Napier, T.C. Mitochondrial function influences expression of methamphetamine-induced behavioral sensitization. Sci. Rep. 2021, 11, 24529. [Google Scholar] [CrossRef]
  30. Chivero, E.T.; Thangaraj, A.; Tripathi, A.; Periyasamy, P.; Guo, M.L.; Buch, S. NLRP3 Inflammasome Blockade Reduces Cocaine-Induced Microglial Activation and Neuroinflammation. Mol. Neurobiol. 2021, 58, 2215–2230. [Google Scholar] [CrossRef]
  31. Doke, M.; Jeganathan, V.; McLaughlin, J.P.; Samikkannu, T. HIV-1 Tat and cocaine impact mitochondrial epigenetics: Effects on DNA methylation. Epigenetics 2021, 16, 980–999. [Google Scholar] [CrossRef] [PubMed]
  32. Fan, R.; Shen, Y.; Li, X.; Luo, H.; Zhang, P.; Liu, Y.; Si, Z.; Zhou, W.; Liu, Y. The effect of the NLRP1 inflammasome on methamphetamine-induced cognitive impairment in rats. Drug Alcohol. Depend. 2022, 237, 109537. [Google Scholar] [CrossRef] [PubMed]
  33. Hernandez, J.; Kaun, K.R. Alcohol, neuronal plasticity, and mitochondrial trafficking. Proc. Natl. Acad. Sci. USA 2022, 119, e2208744119. [Google Scholar] [CrossRef] [PubMed]
  34. Hu, P.; Zhu, W.; Zhu, C.; Jin, L.; Guan, Y.; Guan, X. Resveratrol fails to affect cocaine conditioned place preference behavior, but alleviates anxiety-like behaviors in cocaine withdrawn rats. Psychopharmacology 2016, 233, 1279–1287. [Google Scholar] [CrossRef]
  35. Jia, M.; Wang, X.; Zhang, H.; Wang, X.; Ma, H.; Yang, M.; Li, Y.; Cui, C. MicroRNA-132 is involved in morphine dependence via modifying the structural plasticity of the dentate gyrus neurons in rats. Addict. Biol. 2022, 27, e13086. [Google Scholar] [CrossRef]
  36. Lin, M.; Deng, K.; Li, Y.; Wan, J. Morphine enhances LPS-induced macrophage apoptosis through a PPARgamma-dependent mechanism. Exp. Ther. Med. 2021, 22, 714. [Google Scholar] [CrossRef]
  37. Peng, J.; Pan, J.; Wang, H.; Mo, J.; Lan, L.; Peng, Y. Morphine-induced microglial immunosuppression via activation of insufficient mitophagy regulated by NLRX1. J. Neuroinflammation 2022, 19, 87. [Google Scholar] [CrossRef]
  38. Thangaraj, A.; Periyasamy, P.; Guo, M.L.; Chivero, E.T.; Callen, S.; Buch, S. Mitigation of cocaine-mediated mitochondrial damage, defective mitophagy and microglial activation by superoxide dismutase mimetics. Autophagy 2020, 16, 289–312. [Google Scholar] [CrossRef]
  39. Zhang, Z.; Cao, X.; Bao, X.; Zhang, Y.; Xu, Y.; Sha, D. Cocaine- and amphetamine-regulated transcript protects synaptic structures in neurons after ischemic cerebral injury. Neuropeptides 2020, 81, 102023. [Google Scholar] [CrossRef]
  40. Yang, Z.; Klionsky, D.J. Mammalian autophagy: Core molecular machinery and signaling regulation. Curr. Opin. Cell Biol. 2010, 22, 124–131. [Google Scholar] [CrossRef] [Green Version]
  41. Mijaljica, D.; Prescott, M.; Devenish, R.J. Microautophagy in mammalian cells: Revisiting a 40-year-old conundrum. Autophagy 2011, 7, 673–682. [Google Scholar] [CrossRef] [PubMed]
  42. Massey, A.; Kiffin, R.; Cuervo, A.M. Pathophysiology of chaperone-mediated autophagy. Int. J. Biochem. Cell Biol. 2004, 36, 2420–2434. [Google Scholar] [CrossRef] [PubMed]
  43. Yorimitsu, T.; Klionsky, D.J. Autophagy: Molecular machinery for self-eating. Cell Death Differ. 2005, 12 (Suppl. S2), 1542–1552. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Peracchio, C.; Alabiso, O.; Valente, G.; Isidoro, C. Involvement of autophagy in ovarian cancer: A working hypothesis. J. Ovarian Res. 2012, 5, 22. [Google Scholar] [CrossRef] [Green Version]
  45. Feng, Y.; He, D.; Yao, Z.; Klionsky, D.J. The machinery of macroautophagy. Cell Res. 2014, 24, 24–41. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Kroemer, G.; Marino, G.; Levine, B. Autophagy and the integrated stress response. Mol. Cell 2010, 40, 280–293. [Google Scholar] [CrossRef] [Green Version]
  47. Campoy, E.; Colombo, M.I. Autophagy in intracellular bacterial infection. Biochim. Biophys. Acta 2009, 1793, 1465–1477. [Google Scholar] [CrossRef] [Green Version]
  48. Nixon, R.A. The role of autophagy in neurodegenerative disease. Nat. Med. 2013, 19, 983–997. [Google Scholar] [CrossRef]
  49. Yang, Z.J.; Chee, C.E.; Huang, S.; Sinicrope, F.A. The role of autophagy in cancer: Therapeutic implications. Mol. Cancer Ther. 2011, 10, 1533–1541. [Google Scholar] [CrossRef] [Green Version]
  50. Espert, L.; Denizot, M.; Grimaldi, M.; Robert-Hebmann, V.; Gay, B.; Varbanov, M.; Codogno, P.; Biard-Piechaczyk, M. Autophagy is involved in T cell death after binding of HIV-1 envelope proteins to CXCR4. J. Clin. Investig. 2006, 116, 2161–2172. [Google Scholar] [CrossRef]
  51. Espert, L.; Varbanov, M.; Robert-Hebmann, V.; Sagnier, S.; Robbins, I.; Sanchez, F.; Lafont, V.; Biard-Piechaczyk, M. Differential role of autophagy in CD4 T cells and macrophages during X4 and R5 HIV-1 infection. PLoS ONE 2009, 4, e5787. [Google Scholar] [CrossRef] [PubMed]
  52. Sagnier, S.; Daussy, C.F.; Borel, S.; Robert-Hebmann, V.; Faure, M.; Blanchet, F.P.; Beaumelle, B.; Biard-Piechaczyk, M.; Espert, L. Autophagy restricts HIV-1 infection by selectively degrading Tat in CD4+ T lymphocytes. J. Virol. 2015, 89, 615–625. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Kyei, G.B.; Dinkins, C.; Davis, A.S.; Roberts, E.; Singh, S.B.; Dong, C.; Wu, L.; Kominami, E.; Ueno, T.; Yamamoto, A.; et al. Autophagy pathway intersects with HIV-1 biosynthesis and regulates viral yields in macrophages. J. Cell Biol. 2009, 186, 255–268. [Google Scholar] [CrossRef]
  54. Blanchet, F.P.; Moris, A.; Nikolic, D.S.; Lehmann, M.; Cardinaud, S.; Stalder, R.; Garcia, E.; Dinkins, C.; Leuba, F.; Wu, L.; et al. Human immunodeficiency virus-1 inhibition of immunoamphisomes in dendritic cells impairs early innate and adaptive immune responses. Immunity 2010, 32, 654–669. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Friedman, H.; Newton, C.; Klein, T.W. Microbial infections, immunomodulation, and drugs of abuse. Clin. Microbiol. Rev. 2003, 16, 209–219. [Google Scholar] [CrossRef] [Green Version]
  56. Cao, L.; Glazyrin, A.; Kumar, S.; Kumar, A. Role of Autophagy in HIV Pathogenesis and Drug Abuse. Mol. Neurobiol. 2017, 54, 5855–5867. [Google Scholar] [CrossRef]
  57. Kling, M.A.; Alesci, S.; Csako, G.; Costello, R.; Luckenbaugh, D.A.; Bonne, O.; Duncko, R.; Drevets, W.C.; Manji, H.K.; Charney, D.S.; et al. Sustained low-grade pro-inflammatory state in unmedicated, remitted women with major depressive disorder as evidenced by elevated serum levels of the acute phase proteins C-reactive protein and serum amyloid A. Biol. Psychiatry 2007, 62, 309–313. [Google Scholar] [CrossRef] [Green Version]
  58. Nixon, D.E.; Landay, A.L. Biomarkers of immune dysfunction in HIV. Curr. Opin. HIV AIDS 2010, 5, 498–503. [Google Scholar] [CrossRef] [Green Version]
  59. Triant, V.A.; Meigs, J.B.; Grinspoon, S.K. Association of C-reactive protein and HIV infection with acute myocardial infarction. J. Acquir. Immune Defic. Syndr. 2009, 51, 268–273. [Google Scholar] [CrossRef] [Green Version]
  60. Agrawal, A.; Simpson, M.J.; Black, S.; Carey, M.P.; Samols, D. A C-reactive protein mutant that does not bind to phosphocholine and pneumococcal C-polysaccharide. J. Immunol. 2002, 169, 3217–3222. [Google Scholar] [CrossRef] [Green Version]
  61. Malle, E.; Steinmetz, A.; Raynes, J.G. Serum amyloid A (SAA): An acute phase protein and apolipoprotein. Atherosclerosis 1993, 102, 131–146. [Google Scholar] [CrossRef] [PubMed]
  62. Kramer, H.B.; Lavender, K.J.; Qin, L.; Stacey, A.R.; Liu, M.K.; di Gleria, K.; Simmons, A.; Gasper-Smith, N.; Haynes, B.F.; McMichael, A.J.; et al. Elevation of intact and proteolytic fragments of acute phase proteins constitutes the earliest systemic antiviral response in HIV-1 infection. PLoS Pathog. 2010, 6, e1000893. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Lau, B.; Sharrett, A.R.; Kingsley, L.A.; Post, W.; Palella, F.J.; Visscher, B.; Gange, S.J. C-reactive protein is a marker for human immunodeficiency virus disease progression. Arch. Intern. Med. 2006, 166, 64–70. [Google Scholar] [CrossRef] [PubMed]
  64. Noursadeghi, M.; Miller, R.F. Clinical value of C-reactive protein measurements in HIV-positive patients. Int. J. STD AIDS 2005, 16, 438–441. [Google Scholar] [CrossRef]
  65. Buch, S.; Yao, H.; Guo, M.; Mori, T.; Mathias-Costa, B.; Singh, V.; Seth, P.; Wang, J.; Su, T.P. Cocaine and HIV-1 interplay in CNS: Cellular and molecular mechanisms. Curr. HIV Res. 2012, 10, 425–428. [Google Scholar] [CrossRef]
  66. Chang, S.L.; Connaghan, K.P.; Wei, Y.; Li, M.D. NeuroHIV and use of addictive substances. Int. Rev. Neurobiol. 2014, 118, 403–440. [Google Scholar] [CrossRef]
  67. Chilunda, V.; Calderon, T.M.; Martinez-Aguado, P.; Berman, J.W. The impact of substance abuse on HIV-mediated neuropathogenesis in the current ART era. Brain Res. 2019, 1724, 146426. [Google Scholar] [CrossRef]
  68. Monnig, M.A. Immune activation and neuroinflammation in alcohol use and HIV infection: Evidence for shared mechanisms. Am. J. Drug Alcohol. Abuse 2017, 43, 7–23. [Google Scholar] [CrossRef] [Green Version]
  69. Potula, R.; Haorah, J.; Knipe, B.; Leibhart, J.; Chrastil, J.; Heilman, D.; Dou, H.; Reddy, R.; Ghorpade, A.; Persidsky, Y. Alcohol abuse enhances neuroinflammation and impairs immune responses in an animal model of human immunodeficiency virus-1 encephalitis. Am. J. Pathol. 2006, 168, 1335–1344. [Google Scholar] [CrossRef] [Green Version]
  70. Silverstein, P.S.; Shah, A.; Gupte, R.; Liu, X.; Piepho, R.W.; Kumar, S.; Kumar, A. Methamphetamine toxicity and its implications during HIV-1 infection. J. Neurovirol. 2011, 17, 401–415. [Google Scholar] [CrossRef]
  71. Guo, M.L.; Liao, K.; Periyasamy, P.; Yang, L.; Cai, Y.; Callen, S.E.; Buch, S. Cocaine-mediated microglial activation involves the ER stress-autophagy axis. Autophagy 2015, 11, 995–1009. [Google Scholar] [CrossRef] [PubMed]
  72. Liao, K.; Guo, M.; Niu, F.; Yang, L.; Callen, S.E.; Buch, S. Cocaine-mediated induction of microglial activation involves the ER stress-TLR2 axis. J. Neuroinflammation 2016, 13, 33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Sil, S.; Niu, F.; Tom, E.; Liao, K.; Periyasamy, P.; Buch, S. Cocaine Mediated Neuroinflammation: Role of Dysregulated Autophagy in Pericytes. Mol. Neurobiol. 2019, 56, 3576–3590. [Google Scholar] [CrossRef] [PubMed]
  74. Wang, X.; Sun, L.; Zhou, Y.; Su, Q.J.; Li, J.L.; Ye, L.; Liu, M.Q.; Zhou, W.; Ho, W.Z. Heroin Abuse and/or HIV Infection Dysregulate Plasma Exosomal miRNAs. J. Neuroimmune Pharmacol. 2020, 15, 400–408. [Google Scholar] [CrossRef] [PubMed]
  75. Perez, P.S.; Romaniuk, M.A.; Duette, G.A.; Zhao, Z.; Huang, Y.; Martin-Jaular, L.; Witwer, K.W.; Thery, C.; Ostrowski, M. Extracellular vesicles and chronic inflammation during HIV infection. J. Extracell Vesicles 2019, 8, 1687275. [Google Scholar] [CrossRef]
  76. Mukhamedova, N.; Hoang, A.; Dragoljevic, D.; Dubrovsky, L.; Pushkarsky, T.; Low, H.; Ditiatkovski, M.; Fu, Y.; Ohkawa, R.; Meikle, P.J.; et al. Exosomes containing HIV protein Nef reorganize lipid rafts potentiating inflammatory response in bystander cells. PLoS Pathog. 2019, 15, e1007907. [Google Scholar] [CrossRef] [Green Version]
  77. Sampey, G.C.; Saifuddin, M.; Schwab, A.; Barclay, R.; Punya, S.; Chung, M.C.; Hakami, R.M.; Zadeh, M.A.; Lepene, B.; Klase, Z.A.; et al. Exosomes from HIV-1-infected Cells Stimulate Production of Pro-inflammatory Cytokines through Trans-activating Response (TAR) RNA. J. Biol. Chem. 2016, 291, 1251–1266. [Google Scholar] [CrossRef] [Green Version]
  78. Chettimada, S.; Lorenz, D.R.; Misra, V.; Wolinsky, S.M.; Gabuzda, D. Small RNA sequencing of extracellular vesicles identifies circulating miRNAs related to inflammation and oxidative stress in HIV patients. BMC Immunol. 2020, 21, 57. [Google Scholar] [CrossRef]
  79. Ibanez, F.; Montesinos, J.; Urena-Peralta, J.R.; Guerri, C.; Pascual, M. TLR4 participates in the transmission of ethanol-induced neuroinflammation via astrocyte-derived extracellular vesicles. J. Neuroinflammation 2019, 16, 136. [Google Scholar] [CrossRef] [Green Version]
  80. Kovalevich, J.; Langford, D. Neuronal toxicity in HIV CNS disease. Future Virol. 2012, 7, 687–698. [Google Scholar] [CrossRef] [Green Version]
  81. Deren, S.; Cortes, T.; Dickson, V.V.; Guilamo-Ramos, V.; Han, B.H.; Karpiak, S.; Naegle, M.; Ompad, D.C.; Wu, B. Substance Use Among Older People Living With HIV: Challenges for Health Care Providers. Front. Public Health 2019, 7, 94. [Google Scholar] [CrossRef] [PubMed]
  82. Ghidei, L.; Simone, M.J.; Salow, M.J.; Zimmerman, K.M.; Paquin, A.M.; Skarf, L.M.; Kostas, T.R.; Rudolph, J.L. Aging, antiretrovirals, and adherence: A meta analysis of adherence among older HIV-infected individuals. Drugs Aging 2013, 30, 809–819. [Google Scholar] [CrossRef] [PubMed]
  83. Maciel, R.A.; Kluck, H.M.; Durand, M.; Sprinz, E. Comorbidity is more common and occurs earlier in persons living with HIV than in HIV-uninfected matched controls, aged 50 years and older: A cross-sectional study. Int. J. Infect. Dis. 2018, 70, 30–35. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Negredo, E.; Back, D.; Blanco, J.R.; Blanco, J.; Erlandson, K.M.; Garolera, M.; Guaraldi, G.; Mallon, P.; Molto, J.; Serra, J.A.; et al. Aging in HIV-Infected Subjects: A New Scenario and a New View. Biomed. Res. Int 2017, 2017, 5897298. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Kupprat, S.A.; Krause, K.D.; Ompad, D.C.; Halkitis, P.N. Substance Use and Cognitive Function as Drivers of Condomless Anal Sex Among HIV-Positive Gay, Bisexual, and Other Men Who Have Sex with Men Aged 50 and Older: The Gold Studies. LGBT Health 2017, 4, 434–441. [Google Scholar] [CrossRef] [Green Version]
  86. Sierra, A.; Abiega, O.; Shahraz, A.; Neumann, H. Janus-faced microglia: Beneficial and detrimental consequences of microglial phagocytosis. Front. Cell Neurosci. 2013, 7, 6. [Google Scholar] [CrossRef] [Green Version]
  87. Cherry, J.D.; Olschowka, J.A.; O’Banion, M.K. Neuroinflammation and M2 microglia: The good, the bad, and the inflamed. J. Neuroinflammation 2014, 11, 98. [Google Scholar] [CrossRef] [Green Version]
  88. Tremblay, M.E.; Stevens, B.; Sierra, A.; Wake, H.; Bessis, A.; Nimmerjahn, A. The role of microglia in the healthy brain. J. Neurosci. 2011, 31, 16064–16069. [Google Scholar] [CrossRef] [Green Version]
  89. Gemma, C.; Bachstetter, A.D. The role of microglia in adult hippocampal neurogenesis. Front. Cell Neurosci. 2013, 7, 229. [Google Scholar] [CrossRef] [Green Version]
  90. Su, P.; Zhang, J.; Zhao, F.; Aschner, M.; Chen, J.; Luo, W. The interaction between microglia and neural stem/precursor cells. Brain Res. Bull. 2014, 109, 32–38. [Google Scholar] [CrossRef]
  91. Kigerl, K.A.; Gensel, J.C.; Ankeny, D.P.; Alexander, J.K.; Donnelly, D.J.; Popovich, P.G. Identification of two distinct macrophage subsets with divergent effects causing either neurotoxicity or regeneration in the injured mouse spinal cord. J. Neurosci. 2009, 29, 13435–13444. [Google Scholar] [CrossRef]
  92. Jin, M.M.; Wang, F.; Qi, D.; Liu, W.W.; Gu, C.; Mao, C.J.; Yang, Y.P.; Zhao, Z.; Hu, L.F.; Liu, C.F. A Critical Role of Autophagy in Regulating Microglia Polarization in Neurodegeneration. Front. Aging Neurosci. 2018, 10, 378. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Dheen, S.T.; Kaur, C.; Ling, E.A. Microglial activation and its implications in the brain diseases. Curr. Med. Chem. 2007, 14, 1189–1197. [Google Scholar] [CrossRef] [PubMed]
  94. Lecours, C.; Bordeleau, M.; Cantin, L.; Parent, M.; Paolo, T.D.; Tremblay, M.E. Microglial Implication in Parkinson’s Disease: Loss of Beneficial Physiological Roles or Gain of Inflammatory Functions? Front. Cell Neurosci. 2018, 12, 282. [Google Scholar] [CrossRef] [Green Version]
  95. Spittau, B. Aging Microglia-Phenotypes, Functions and Implications for Age-Related Neurodegenerative Diseases. Front. Aging Neurosci. 2017, 9, 194. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Zurcher, N.R.; Loggia, M.L.; Lawson, R.; Chonde, D.B.; Izquierdo-Garcia, D.; Yasek, J.E.; Akeju, O.; Catana, C.; Rosen, B.R.; Cudkowicz, M.E.; et al. Increased in vivo glial activation in patients with amyotrophic lateral sclerosis: Assessed with [(11)C]-PBR28. Neuroimage Clin. 2015, 7, 409–414. [Google Scholar] [CrossRef] [Green Version]
  97. Germic, N.; Frangez, Z.; Yousefi, S.; Simon, H.U. Regulation of the innate immune system by autophagy: Monocytes, macrophages, dendritic cells and antigen presentation. Cell Death Differ. 2019, 26, 715–727. [Google Scholar] [CrossRef]
  98. Plaza-Zabala, A.; Sierra-Torre, V.; Sierra, A. Autophagy and Microglia: Novel Partners in Neurodegeneration and Aging. Int. J. Mol. Sci. 2017, 18, 598. [Google Scholar] [CrossRef] [Green Version]
  99. Su, P.; Zhang, J.; Wang, D.; Zhao, F.; Cao, Z.; Aschner, M.; Luo, W. The role of autophagy in modulation of neuroinflammation in microglia. Neuroscience 2016, 319, 155–167. [Google Scholar] [CrossRef]
  100. Franceschi, C.; Garagnani, P.; Vitale, G.; Capri, M.; Salvioli, S. Inflammaging and ‘Garb-aging’. Trends Endocrinol. Metab. 2017, 28, 199–212. [Google Scholar] [CrossRef] [Green Version]
  101. Costantini, E.; D’Angelo, C.; Reale, M. The Role of Immunosenescence in Neurodegenerative Diseases. Mediators Inflamm. 2018, 2018, 6039171. [Google Scholar] [CrossRef] [PubMed]
  102. Olivieri, F.; Prattichizzo, F.; Grillari, J.; Balistreri, C.R. Cellular Senescence and Inflammaging in Age-Related Diseases. Mediators Inflamm. 2018, 2018, 9076485. [Google Scholar] [CrossRef] [PubMed]
  103. von Bernhardi, R.; Eugenin-von Bernhardi, L.; Eugenin, J. Microglial cell dysregulation in brain aging and neurodegeneration. Front. Aging Neurosci. 2015, 7, 124. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Monti, D.; Ostan, R.; Borelli, V.; Castellani, G.; Franceschi, C. Inflammaging and human longevity in the omics era. Mech. Ageing Dev. 2017, 165, 129–138. [Google Scholar] [CrossRef] [PubMed]
  105. Franceschi, C.; Garagnani, P.; Parini, P.; Giuliani, C.; Santoro, A. Inflammaging: A new immune-metabolic viewpoint for age-related diseases. Nat. Rev. Endocrinol. 2018, 14, 576–590. [Google Scholar] [CrossRef]
  106. Sierra, A.; Gottfried-Blackmore, A.C.; McEwen, B.S.; Bulloch, K. Microglia derived from aging mice exhibit an altered inflammatory profile. Glia 2007, 55, 412–424. [Google Scholar] [CrossRef]
  107. VanGuilder, H.D.; Bixler, G.V.; Brucklacher, R.M.; Farley, J.A.; Yan, H.; Warrington, J.P.; Sonntag, W.E.; Freeman, W.M. Concurrent hippocampal induction of MHC II pathway components and glial activation with advanced aging is not correlated with cognitive impairment. J. Neuroinflammation 2011, 8, 138. [Google Scholar] [CrossRef] [Green Version]
  108. Godbout, J.P.; Chen, J.; Abraham, J.; Richwine, A.F.; Berg, B.M.; Kelley, K.W.; Johnson, R.W. Exaggerated neuroinflammation and sickness behavior in aged mice following activation of the peripheral innate immune system. FASEB J. 2005, 19, 1329–1331. [Google Scholar] [CrossRef]
  109. Frank, M.G.; Barrientos, R.M.; Biedenkapp, J.C.; Rudy, J.W.; Watkins, L.R.; Maier, S.F. mRNA up-regulation of MHC II and pivotal pro-inflammatory genes in normal brain aging. Neurobiol. Aging 2006, 27, 717–722. [Google Scholar] [CrossRef]
  110. Streit, W.J.; Sparks, D.L. Activation of microglia in the brains of humans with heart disease and hypercholesterolemic rabbits. J. Mol. Med. 1997, 75, 130–138. [Google Scholar] [CrossRef]
  111. Sheffield, L.G.; Berman, N.E. Microglial expression of MHC class II increases in normal aging of nonhuman primates. Neurobiol. Aging 1998, 19, 47–55. [Google Scholar] [CrossRef] [PubMed]
  112. Norden, D.M.; Godbout, J.P. Review: Microglia of the aged brain: Primed to be activated and resistant to regulation. Neuropathol. Appl. Neurobiol. 2013, 39, 19–34. [Google Scholar] [CrossRef] [PubMed]
  113. Niraula, A.; Sheridan, J.F.; Godbout, J.P. Microglia Priming with Aging and Stress. Neuropsychopharmacology 2017, 42, 318–333. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Choi, J.H.; Lee, C.H.; Hwang, I.K.; Won, M.H.; Seong, J.K.; Yoon, Y.S.; Lee, H.S.; Lee, I.S. Age-related changes in ionized calcium-binding adapter molecule 1 immunoreactivity and protein level in the gerbil hippocampal CA1 region. J. Vet. Med. Sci. 2007, 69, 1131–1136. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Hwang, I.K.; Lee, C.H.; Li, H.; Yoo, K.Y.; Choi, J.H.; Kim, D.W.; Kim, D.W.; Suh, H.W.; Won, M.H. Comparison of ionized calcium-binding adapter molecule 1 immunoreactivity of the hippocampal dentate gyrus and CA1 region in adult and aged dogs. Neurochem. Res. 2008, 33, 1309–1315. [Google Scholar] [CrossRef]
  116. Gerbino, V.; Kaunga, E.; Ye, J.; Canzio, D.; O’Keeffe, S.; Rudnick, N.D.; Guarnieri, P.; Lutz, C.M.; Maniatis, T. The Loss of TBK1 Kinase Activity in Motor Neurons or in All Cell Types Differentially Impacts ALS Disease Progression in SOD1 Mice. Neuron 2020, 106, 789–805.e785. [Google Scholar] [CrossRef]
  117. Thangaraj, A.; Periyasamy, P.; Liao, K.; Bendi, V.S.; Callen, S.; Pendyala, G.; Buch, S. HIV-1 TAT-mediated microglial activation: Role of mitochondrial dysfunction and defective mitophagy. Autophagy 2018, 14, 1596–1619. [Google Scholar] [CrossRef] [Green Version]
  118. Lull, M.E.; Block, M.L. Microglial activation and chronic neurodegeneration. Neurotherapeutics 2010, 7, 354–365. [Google Scholar] [CrossRef] [Green Version]
  119. Tripathi, A.; Thangaraj, A.; Chivero, E.T.; Periyasamy, P.; Callen, S.; Burkovetskaya, M.E.; Guo, M.L.; Buch, S. Antiretroviral-Mediated Microglial Activation Involves Dysregulated Autophagy and Lysosomal Dysfunction. Cells 2019, 8, 1168. [Google Scholar] [CrossRef] [Green Version]
  120. Ye, X.; Zhu, M.; Che, X.; Wang, H.; Liang, X.J.; Wu, C.; Xue, X.; Yang, J. Lipopolysaccharide induces neuroinflammation in microglia by activating the MTOR pathway and downregulating Vps34 to inhibit autophagosome formation. J. Neuroinflammation 2020, 17, 18. [Google Scholar] [CrossRef] [Green Version]
  121. Janda, E.; Boi, L.; Carta, A.R. Microglial Phagocytosis and Its Regulation: A Therapeutic Target in Parkinson’s Disease? Front. Mol. Neurosci. 2018, 11, 144. [Google Scholar] [CrossRef] [PubMed]
  122. Zhang, H.; Puleston, D.J.; Simon, A.K. Autophagy and Immune Senescence. Trends Mol. Med. 2016, 22, 671–686. [Google Scholar] [CrossRef] [PubMed]
  123. Fu, R.; Shen, Q.; Xu, P.; Luo, J.J.; Tang, Y. Phagocytosis of microglia in the central nervous system diseases. Mol. Neurobiol. 2014, 49, 1422–1434. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Martinez, J.; Almendinger, J.; Oberst, A.; Ness, R.; Dillon, C.P.; Fitzgerald, P.; Hengartner, M.O.; Green, D.R. Microtubule-associated protein 1 light chain 3 alpha (LC3)-associated phagocytosis is required for the efficient clearance of dead cells. Proc. Natl. Acad. Sci. USA 2011, 108, 17396–17401. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Koellhoffer, E.C.; McCullough, L.D.; Ritzel, R.M. Old Maids: Aging and Its Impact on Microglia Function. Int J. Mol. Sci 2017, 18, 769. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Choi, I.; Zhang, Y.; Seegobin, S.P.; Pruvost, M.; Wang, Q.; Purtell, K.; Zhang, B.; Yue, Z. Microglia clear neuron-released alpha-synuclein via selective autophagy and prevent neurodegeneration. Nat. Commun. 2020, 11, 1386. [Google Scholar] [CrossRef] [Green Version]
  127. Pomilio, C.; Gorojod, R.M.; Riudavets, M.; Vinuesa, A.; Presa, J.; Gregosa, A.; Bentivegna, M.; Alaimo, A.; Alcon, S.P.; Sevlever, G.; et al. Microglial autophagy is impaired by prolonged exposure to beta-amyloid peptides: Evidence from experimental models and Alzheimer’s disease patients. Geroscience 2020, 42, 613–632. [Google Scholar] [CrossRef]
  128. Fang, E.F.; Hou, Y.; Palikaras, K.; Adriaanse, B.A.; Kerr, J.S.; Yang, B.; Lautrup, S.; Hasan-Olive, M.M.; Caponio, D.; Dan, X.; et al. Mitophagy inhibits amyloid-beta and tau pathology and reverses cognitive deficits in models of Alzheimer’s disease. Nat. Neurosci. 2019, 22, 401–412. [Google Scholar] [CrossRef]
  129. Thangaraj, A.; Sil, S.; Tripathi, A.; Chivero, E.T.; Periyasamy, P.; Buch, S. Targeting endoplasmic reticulum stress and autophagy as therapeutic approaches for neurological diseases. Int. Rev. Cell Mol. Biol. 2020, 350, 285–325. [Google Scholar] [CrossRef]
  130. Cho, K.S.; Lee, J.H.; Cho, J.; Cha, G.H.; Song, G.J. Autophagy Modulators and Neuroinflammation. Curr. Med. Chem. 2020, 27, 955–982. [Google Scholar] [CrossRef]
  131. Navia, B.A.; Cho, E.S.; Petito, C.K.; Price, R.W. The AIDS dementia complex: II. Neuropathology. Ann. Neurol. 1986, 19, 525–535. [Google Scholar] [CrossRef] [PubMed]
  132. Bansal, A.K.; Mactutus, C.F.; Nath, A.; Maragos, W.; Hauser, K.F.; Booze, R.M. Neurotoxicity of HIV-1 proteins gp120 and Tat in the rat striatum. Brain Res. 2000, 879, 42–49. [Google Scholar] [CrossRef] [PubMed]
  133. Cheney, L.; Guzik, H.; Macaluso, F.P.; Macian, F.; Cuervo, A.M.; Berman, J.W. HIV Nef and Antiretroviral Therapy Have an Inhibitory Effect on Autophagy in Human Astrocytes that May Contribute to HIV-Associated Neurocognitive Disorders. Cells 2020, 9, 1426. [Google Scholar] [CrossRef] [PubMed]
  134. Mehla, R.; Chauhan, A. HIV-1 differentially modulates autophagy in neurons and astrocytes. J. Neuroimmunol. 2015, 285, 106–118. [Google Scholar] [CrossRef]
  135. Fields, J.; Dumaop, W.; Eleuteri, S.; Campos, S.; Serger, E.; Trejo, M.; Kosberg, K.; Adame, A.; Spencer, B.; Rockenstein, E.; et al. HIV-1 Tat alters neuronal autophagy by modulating autophagosome fusion to the lysosome: Implications for HIV-associated neurocognitive disorders. J. Neurosci. 2015, 35, 1921–1938. [Google Scholar] [CrossRef] [Green Version]
  136. Cao, L.; Fu, M.; Kumar, S.; Kumar, A. Methamphetamine potentiates HIV-1 gp120-mediated autophagy via Beclin-1 and Atg5/7 as a pro-survival response in astrocytes. Cell Death Dis. 2016, 7, e2425. [Google Scholar] [CrossRef] [Green Version]
  137. Wu, X.; Dong, H.; Ye, X.; Zhong, L.; Cao, T.; Xu, Q.; Wang, J.; Zhang, Y.; Xu, J.; Wang, W.; et al. HIV-1 Tat increases BAG3 via NF-kappaB signaling to induce autophagy during HIV-associated neurocognitive disorder. Cell Cycle 2018, 17, 1614–1623. [Google Scholar] [CrossRef] [Green Version]
  138. Saribas, A.S.; Khalili, K.; Sariyer, I.K. Dysregulation of autophagy by HIV-1 Nef in human astrocytes. Cell Cycle 2015, 14, 2899–2904. [Google Scholar] [CrossRef] [Green Version]
  139. Chompre, G.; Martinez-Orengo, N.; Cruz, M.; Porter, J.T.; Noel, R.J., Jr. TGFbetaRI antagonist inhibits HIV-1 Nef-induced CC chemokine family ligand 2 (CCL2) in the brain and prevents spatial learning impairment. J. Neuroinflammation 2019, 16, 262. [Google Scholar] [CrossRef] [Green Version]
  140. Mordelet, E.; Kissa, K.; Cressant, A.; Gray, F.; Ozden, S.; Vidal, C.; Charneau, P.; Granon, S. Histopathological and cognitive defects induced by Nef in the brain. FASEB J. 2004, 18, 1851–1861. [Google Scholar] [CrossRef]
  141. Sami Saribas, A.; Cicalese, S.; Ahooyi, T.M.; Khalili, K.; Amini, S.; Sariyer, I.K. HIV-1 Nef is released in extracellular vesicles derived from astrocytes: Evidence for Nef-mediated neurotoxicity. Cell Death Dis. 2017, 8, e2542. [Google Scholar] [CrossRef] [PubMed]
  142. Ojeda, D.S.; Grasso, D.; Urquiza, J.; Till, A.; Vaccaro, M.I.; Quarleri, J. Cell Death Is Counteracted by Mitophagy in HIV-Productively Infected Astrocytes but Is Promoted by Inflammasome Activation Among Non-productively Infected Cells. Front. Immunol. 2018, 9, 2633. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. El-Hage, N.; Wu, G.; Wang, J.; Ambati, J.; Knapp, P.E.; Reed, J.L.; Bruce-Keller, A.J.; Hauser, K.F. HIV-1 Tat and opiate-induced changes in astrocytes promote chemotaxis of microglia through the expression of MCP-1 and alternative chemokines. Glia 2006, 53, 132–146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Suzuki, M.; El-Hage, N.; Zou, S.; Hahn, Y.K.; Sorrell, M.E.; Sturgill, J.L.; Conrad, D.H.; Knapp, P.E.; Hauser, K.F. Fractalkine/CX3CL1 protects striatal neurons from synergistic morphine and HIV-1 Tat-induced dendritic losses and death. Mol. Neurodegener. 2011, 6, 78. [Google Scholar] [CrossRef] [Green Version]
  145. El-Hage, N.; Rodriguez, M.; Dever, S.M.; Masvekar, R.R.; Gewirtz, D.A.; Shacka, J.J. HIV-1 and morphine regulation of autophagy in microglia: Limited interactions in the context of HIV-1 infection and opioid abuse. J. Virol. 2015, 89, 1024–1035. [Google Scholar] [CrossRef] [Green Version]
  146. Reynolds, J.L.; Mahajan, S.D.; Bindukumar, B.; Sykes, D.; Schwartz, S.A.; Nair, M.P. Proteomic analysis of the effects of cocaine on the enhancement of HIV-1 replication in normal human astrocytes (NHA). Brain Res. 2006, 1123, 226–236. [Google Scholar] [CrossRef] [Green Version]
  147. Natarajaseenivasan, K.; Cotto, B.; Shanmughapriya, S.; Lombardi, A.A.; Datta, P.K.; Madesh, M.; Elrod, J.W.; Khalili, K.; Langford, D. Astrocytic metabolic switch is a novel etiology for Cocaine and HIV-1 Tat-mediated neurotoxicity. Cell Death Dis. 2018, 9, 415. [Google Scholar] [CrossRef] [Green Version]
  148. Borgmann, K.; Ghorpade, A. Methamphetamine Augments Concurrent Astrocyte Mitochondrial Stress, Oxidative Burden, and Antioxidant Capacity: Tipping the Balance in HIV-Associated Neurodegeneration. Neurotox Res. 2018, 33, 433–447. [Google Scholar] [CrossRef]
  149. Cao, L.; Walker, M.P.; Vaidya, N.K.; Fu, M.; Kumar, S.; Kumar, A. Cocaine-Mediated Autophagy in Astrocytes Involves Sigma 1 Receptor, PI3K, mTOR, Atg5/7, Beclin-1 and Induces Type II Programed Cell Death. Mol. Neurobiol. 2016, 53, 4417–4430. [Google Scholar] [CrossRef] [Green Version]
  150. Rodriguez, M.; Lapierre, J.; Ojha, C.R.; Pawitwar, S.; Karuppan, M.K.M.; Kashanchi, F.; El-Hage, N. Morphine counteracts the antiviral effect of antiretroviral drugs and causes upregulation of p62/SQSTM1 and histone-modifying enzymes in HIV-infected astrocytes. J. Neurovirol. 2019, 25, 263–274. [Google Scholar] [CrossRef]
  151. Sil, S.; Periyasamy, P.; Guo, M.L.; Callen, S.; Buch, S. Morphine-Mediated Brain Region-Specific Astrocytosis Involves the ER Stress-Autophagy Axis. Mol. Neurobiol. 2018, 55, 6713–6733. [Google Scholar] [CrossRef] [PubMed]
  152. Chang, L.; Andres, M.; Sadino, J.; Jiang, C.S.; Nakama, H.; Miller, E.; Ernst, T. Impact of apolipoprotein E epsilon4 and HIV on cognition and brain atrophy: Antagonistic pleiotropy and premature brain aging. Neuroimage 2011, 58, 1017–1027. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Jackson, B.; Cubela, R.; Johnston, C.I. Tissue angiotensin converting enzyme (ACE) during changes in the renin-angiotensin system. J. Cardiovasc. Pharmacol. 1987, 10 (Suppl. S7), S137–S140. [Google Scholar] [CrossRef] [PubMed]
  154. Crawford, F.; Wood, M.; Ferguson, S.; Mathura, V.; Gupta, P.; Humphrey, J.; Mouzon, B.; Laporte, V.; Margenthaler, E.; O’Steen, B.; et al. Apolipoprotein E-genotype dependent hippocampal and cortical responses to traumatic brain injury. Neuroscience 2009, 159, 1349–1362. [Google Scholar] [CrossRef]
  155. Theendakara, V.; Peters-Libeu, C.A.; Spilman, P.; Poksay, K.S.; Bredesen, D.E.; Rao, R.V. Direct Transcriptional Effects of Apolipoprotein E. J. Neurosci. 2016, 36, 685–700. [Google Scholar] [CrossRef] [Green Version]
  156. Babu, H.; Ambikan, A.T.; Gabriel, E.E.; Svensson Akusjarvi, S.; Palaniappan, A.N.; Sundaraj, V.; Mupanni, N.R.; Sperk, M.; Cheedarla, N.; Sridhar, R.; et al. Systemic Inflammation and the Increased Risk of Inflamm-Aging and Age-Associated Diseases in People Living With HIV on Long Term Suppressive Antiretroviral Therapy. Front. Immunol. 2019, 10, 1965. [Google Scholar] [CrossRef] [Green Version]
  157. Cohen, J.; D’Agostino, L.; Wilson, J.; Tuzer, F.; Torres, C. Astrocyte Senescence and Metabolic Changes in Response to HIV Antiretroviral Therapy Drugs. Front. Aging Neurosci. 2017, 9, 281. [Google Scholar] [CrossRef] [Green Version]
  158. Jarvis, R.; Tamashiro-Orrego, A.; Promes, V.; Tu, L.; Shi, J.; Yang, Y. Cocaine Self-administration and Extinction Inversely Alter Neuron to Glia Exosomal Dynamics in the Nucleus Accumbens. Front. Cell Neurosci. 2019, 13, 581. [Google Scholar] [CrossRef] [Green Version]
  159. Yu, C.; Narasipura, S.D.; Richards, M.H.; Hu, X.T.; Yamamoto, B.; Al-Harthi, L. HIV and drug abuse mediate astrocyte senescence in a beta-catenin-dependent manner leading to neuronal toxicity. Aging Cell 2017, 16, 956–965. [Google Scholar] [CrossRef] [Green Version]
  160. de Guglielmo, G.; Fu, Y.; Chen, J.; Larrosa, E.; Hoang, I.; Kawamura, T.; Lorrai, I.; Zorman, B.; Bryant, J.; George, O.; et al. Increases in compulsivity, inflammation, and neural injury in HIV transgenic rats with escalated methamphetamine self-administration under extended-access conditions. Brain Res. 2020, 1726, 146502. [Google Scholar] [CrossRef]
  161. Weed, M.; Adams, R.J.; Hienz, R.D.; Meulendyke, K.A.; Linde, M.E.; Clements, J.E.; Mankowski, J.L.; Zink, M.C. SIV/macaque model of HIV infection in cocaine users: Minimal effects of cocaine on behavior, virus replication, and CNS inflammation. J. Neuroimmune Pharmacol. 2012, 7, 401–411. [Google Scholar] [CrossRef] [PubMed]
  162. Nookala, A.R.; Schwartz, D.C.; Chaudhari, N.S.; Glazyrin, A.; Stephens, E.B.; Berman, N.E.J.; Kumar, A. Methamphetamine augment HIV-1 Tat mediated memory deficits by altering the expression of synaptic proteins and neurotrophic factors. Brain Behav. Immun. 2018, 71, 37–51. [Google Scholar] [CrossRef] [PubMed]
  163. Volpe, G.E.; Ward, H.; Mwamburi, M.; Dinh, D.; Bhalchandra, S.; Wanke, C.; Kane, A.V. Associations of cocaine use and HIV infection with the intestinal microbiota, microbial translocation, and inflammation. J. Stud. Alcohol. Drugs 2014, 75, 347–357. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Ishii, T.; Takanashi, Y.; Sugita, K.; Miyazawa, M.; Yanagihara, R.; Yasuda, K.; Onouchi, H.; Kawabe, N.; Nakata, M.; Yamamoto, Y.; et al. Endogenous reactive oxygen species cause astrocyte defects and neuronal dysfunctions in the hippocampus: A new model for aging brain. Aging Cell 2017, 16, 39–51. [Google Scholar] [CrossRef]
  165. Escobar, K.A.; Cole, N.H.; Mermier, C.M.; VanDusseldorp, T.A. Autophagy and aging: Maintaining the proteome through exercise and caloric restriction. Aging Cell 2019, 18, e12876. [Google Scholar] [CrossRef] [Green Version]
  166. Chang, J.T.; Kumsta, C.; Hellman, A.B.; Adams, L.M.; Hansen, M. Spatiotemporal regulation of autophagy during Caenorhabditis elegans aging. Elife 2017, 6, e18459. [Google Scholar] [CrossRef]
  167. Alirezaei, M.; Kiosses, W.B.; Flynn, C.T.; Brady, N.R.; Fox, H.S. Disruption of neuronal autophagy by infected microglia results in neurodegeneration. PLoS ONE 2008, 3, e2906. [Google Scholar] [CrossRef]
  168. Alirezaei, M.; Kiosses, W.B.; Fox, H.S. Decreased neuronal autophagy in HIV dementia: A mechanism of indirect neurotoxicity. Autophagy 2008, 4, 963–966. [Google Scholar] [CrossRef] [Green Version]
  169. Lapierre, J.; Karuppan, M.K.M.; Perry, M.; Rodriguez, M.; El-Hage, N. Different Roles of Beclin1 in the Interaction Between Glia and Neurons after Exposure to Morphine and the HIV- Trans-Activator of Transcription (Tat) Protein. J. Neuroimmune Pharmacol. 2021. [Google Scholar] [CrossRef]
  170. Komatsu, M.; Waguri, S.; Ueno, T.; Iwata, J.; Murata, S.; Tanida, I.; Ezaki, J.; Mizushima, N.; Ohsumi, Y.; Uchiyama, Y.; et al. Impairment of starvation-induced and constitutive autophagy in Atg7-deficient mice. J. Cell Biol. 2005, 169, 425–434. [Google Scholar] [CrossRef]
  171. Rudnick, N.D.; Griffey, C.J.; Guarnieri, P.; Gerbino, V.; Wang, X.; Piersaint, J.A.; Tapia, J.C.; Rich, M.M.; Maniatis, T. Distinct roles for motor neuron autophagy early and late in the SOD1(G93A) mouse model of ALS. Proc. Natl. Acad. Sci. USA 2017, 114, E8294–E8303. [Google Scholar] [CrossRef] [PubMed]
  172. Petersen, A.; Larsen, K.E.; Behr, G.G.; Romero, N.; Przedborski, S.; Brundin, P.; Sulzer, D. Expanded CAG repeats in exon 1 of the Huntington’s disease gene stimulate dopamine-mediated striatal neuron autophagy and degeneration. Hum. Mol. Genet. 2001, 10, 1243–1254. [Google Scholar] [CrossRef] [PubMed]
  173. Florez-McClure, M.L.; Linseman, D.A.; Chu, C.T.; Barker, P.A.; Bouchard, R.J.; Le, S.S.; Laessig, T.A.; Heidenreich, K.A. The p75 neurotrophin receptor can induce autophagy and death of cerebellar Purkinje neurons. J. Neurosci. 2004, 24, 4498–4509. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Schultz, J.L.; Nopoulos, P.C.; Gonzalez-Alegre, P. Human Immunodeficiency Virus Infection in Huntington’s Disease is Associated with an Earlier Age of Symptom Onset. J. Huntingtons Dis. 2018, 7, 163–166. [Google Scholar] [CrossRef] [PubMed]
  175. Anderson, D.G.; Ferreira-Correia, A.; Rodrigues, F.B.; Aziz, N.A.; Carr, J.; Wild, E.J.; Margolis, R.L.; Krause, A. Comparison of the Huntington’s Disease like 2 and Huntington’s Disease Clinical Phenotypes. Mov. Disord. Clin. Pract. 2019, 6, 302–311. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Achenbach, J.; Faissner, S.; Saft, C. Differential Diagnosis of Chorea-HIV Infection Delays Diagnosis of Huntington’s Disease by Years. Brain Sci. 2021, 11, 710. [Google Scholar] [CrossRef]
  177. Ma, R.; Kutchy, N.A.; Hu, G. Astrocyte-Derived Extracellular Vesicle-Mediated Activation of Primary Ciliary Signaling Contributes to the Development of Morphine Tolerance. Biol. Psychiatry 2021, 90, 575–585. [Google Scholar] [CrossRef]
  178. Leibrand, C.R.; Paris, J.J.; Jones, A.M.; Masuda, Q.N.; Halquist, M.S.; Kim, W.K.; Knapp, P.E.; Kashuba, A.D.M.; Hauser, K.F.; McRae, M. HIV-1 Tat and opioids act independently to limit antiretroviral brain concentrations and reduce blood-brain barrier integrity. J. Neurovirol. 2019, 25, 560–577. [Google Scholar] [CrossRef]
  179. Dever, S.M.; Rodriguez, M.; Lapierre, J.; Costin, B.N.; El-Hage, N. Differing roles of autophagy in HIV-associated neurocognitive impairment and encephalitis with implications for morphine co-exposure. Front. Microbiol. 2015, 6, 653. [Google Scholar] [CrossRef]
  180. Bianchi, M.E.; Agresti, A. HMG proteins: Dynamic players in gene regulation and differentiation. Curr. Opin. Genet. Dev. 2005, 15, 496–506. [Google Scholar] [CrossRef]
  181. Wang, H.; Bloom, O.; Zhang, M.; Vishnubhakat, J.M.; Ombrellino, M.; Che, J.; Frazier, A.; Yang, H.; Ivanova, S.; Borovikova, L.; et al. HMG-1 as a late mediator of endotoxin lethality in mice. Science 1999, 285, 248–251. [Google Scholar] [CrossRef] [PubMed]
  182. Zhu, L.; Huang, G.; Sheng, J.; Fu, Q.; Chen, A. High-mobility group box 1 induces neuron autophagy in a rat spinal root avulsion model. Neuroscience 2016, 315, 286–295. [Google Scholar] [CrossRef] [PubMed]
  183. Yin, W.Y.; Ye, Q.; Huang, H.J.; Xia, N.G.; Chen, Y.Y.; Zhang, Y.; Qu, Q.M. Salidroside protects cortical neurons against glutamate-induced cytotoxicity by inhibiting autophagy. Mol. Cell Biochem. 2016, 419, 53–64. [Google Scholar] [CrossRef] [PubMed]
  184. Tripathi, P.; Rodriguez-Muela, N.; Klim, J.R.; de Boer, A.S.; Agrawal, S.; Sandoe, J.; Lopes, C.S.; Ogliari, K.S.; Williams, L.A.; Shear, M.; et al. Reactive Astrocytes Promote ALS-like Degeneration and Intracellular Protein Aggregation in Human Motor Neurons by Disrupting Autophagy through TGF-beta1. Stem. Cell Reports 2017, 9, 667–680. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Lang, Y.; Li, Y.; Yu, H.; Lin, L.; Chen, X.; Wang, S.; Zhang, H. HOTAIR drives autophagy in midbrain dopaminergic neurons in the substantia nigra compacta in a mouse model of Parkinson’s disease by elevating NPTX2 via miR-221-3p binding. Aging 2020, 12, 7660–7678. [Google Scholar] [CrossRef]
  186. Tian, F.; Morimoto, N.; Liu, W.; Ohta, Y.; Deguchi, K.; Miyazaki, K.; Abe, K. In vivo optical imaging of motor neuron autophagy in a mouse model of amyotrophic lateral sclerosis. Autophagy 2011, 7, 985–992. [Google Scholar] [CrossRef] [Green Version]
  187. Cuervo, A.M. Autophagy and aging: Keeping that old broom working. Trends Genet. 2008, 24, 604–612. [Google Scholar] [CrossRef] [Green Version]
  188. Kaushik, S.; Arias, E.; Kwon, H.; Lopez, N.M.; Athonvarangkul, D.; Sahu, S.; Schwartz, G.J.; Pessin, J.E.; Singh, R. Loss of autophagy in hypothalamic POMC neurons impairs lipolysis. EMBO Rep. 2012, 13, 258–265. [Google Scholar] [CrossRef] [Green Version]
  189. Quan, W.; Kim, H.K.; Moon, E.Y.; Kim, S.S.; Choi, C.S.; Komatsu, M.; Jeong, Y.T.; Lee, M.K.; Kim, K.W.; Kim, M.S.; et al. Role of hypothalamic proopiomelanocortin neuron autophagy in the control of appetite and leptin response. Endocrinology 2012, 153, 1817–1826. [Google Scholar] [CrossRef]
  190. Montagne, A.; Barnes, S.R.; Sweeney, M.D.; Halliday, M.R.; Sagare, A.P.; Zhao, Z.; Toga, A.W.; Jacobs, R.E.; Liu, C.Y.; Amezcua, L.; et al. Blood-brain barrier breakdown in the aging human hippocampus. Neuron 2015, 85, 296–302. [Google Scholar] [CrossRef] [Green Version]
  191. Bates, C.A.; Zheng, W. Brain disposition of alpha-Synuclein: Roles of brain barrier systems and implications for Parkinson’s disease. Fluids Barriers CNS 2014, 11, 17. [Google Scholar] [CrossRef] [PubMed]
  192. Yamazaki, Y.; Kanekiyo, T. Blood-Brain Barrier Dysfunction and the Pathogenesis of Alzheimer’s Disease. Int J. Mol. Sci 2017, 18, 1965. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Chaganti, J.; Marripudi, K.; Staub, L.P.; Rae, C.D.; Gates, T.M.; Moffat, K.J.; Brew, B.J. Imaging correlates of the blood-brain barrier disruption in HIV-associated neurocognitive disorder and therapeutic implications. AIDS 2019, 33, 1843–1852. [Google Scholar] [CrossRef] [PubMed]
  194. Nation, D.A.; Sweeney, M.D.; Montagne, A.; Sagare, A.P.; D’Orazio, L.M.; Pachicano, M.; Sepehrband, F.; Nelson, A.R.; Buennagel, D.P.; Harrington, M.G.; et al. Blood-brain barrier breakdown is an early biomarker of human cognitive dysfunction. Nat. Med. 2019, 25, 270–276. [Google Scholar] [CrossRef]
  195. Honda, Y.; Tanaka, M.; Honda, S. Trehalose extends longevity in the nematode Caenorhabditis elegans. Aging Cell 2010, 9, 558–569. [Google Scholar] [CrossRef]
  196. Cuervo, A.M.; Dice, J.F. Age-related decline in chaperone-mediated autophagy. J. Biol. Chem. 2000, 275, 31505–31513. [Google Scholar] [CrossRef] [Green Version]
  197. Malkiewicz, M.A.; Szarmach, A.; Sabisz, A.; Cubala, W.J.; Szurowska, E.; Winklewski, P.J. Blood-brain barrier permeability and physical exercise. J. Neuroinflammation 2019, 16, 15. [Google Scholar] [CrossRef] [Green Version]
  198. Greene, C.; Campbell, M. Tight junction modulation of the blood brain barrier: CNS delivery of small molecules. Tissue Barriers 2016, 4, e1138017. [Google Scholar] [CrossRef] [Green Version]
  199. Elahy, M.; Jackaman, C.; Mamo, J.C.; Lam, V.; Dhaliwal, S.S.; Giles, C.; Nelson, D.; Takechi, R. Blood-brain barrier dysfunction developed during normal aging is associated with inflammation and loss of tight junctions but not with leukocyte recruitment. Immun. Ageing 2015, 12, 2. [Google Scholar] [CrossRef] [Green Version]
  200. Bors, L.; Toth, K.; Toth, E.Z.; Bajza, A.; Csorba, A.; Szigeti, K.; Mathe, D.; Perlaki, G.; Orsi, G.; Toth, G.K.; et al. Age-dependent changes at the blood-brain barrier. A Comparative structural and functional study in young adult and middle aged rats. Brain Res. Bull. 2018, 139, 269–277. [Google Scholar] [CrossRef]
  201. Shen, L.; Weber, C.R.; Raleigh, D.R.; Yu, D.; Turner, J.R. Tight junction pore and leak pathways: A dynamic duo. Annu. Rev. Physiol. 2011, 73, 283–309. [Google Scholar] [CrossRef] [PubMed]
  202. Liao, K.; Niu, F.; Hu, G.; Guo, M.L.; Sil, S.; Buch, S. HIV Tat-mediated induction of autophagy regulates the disruption of ZO-1 in brain endothelial cells. Tissue Barriers 2020, 8, 1748983. [Google Scholar] [CrossRef] [PubMed]
  203. Sweeney, M.D.; Ayyadurai, S.; Zlokovic, B.V. Pericytes of the neurovascular unit: Key functions and signaling pathways. Nat. Neurosci. 2016, 19, 771–783. [Google Scholar] [CrossRef] [PubMed]
  204. Daneman, R.; Zhou, L.; Kebede, A.A.; Barres, B.A. Pericytes are required for blood-brain barrier integrity during embryogenesis. Nature 2010, 468, 562–566. [Google Scholar] [CrossRef] [Green Version]
  205. Bell, R.D.; Winkler, E.A.; Sagare, A.P.; Singh, I.; LaRue, B.; Deane, R.; Zlokovic, B.V. Pericytes control key neurovascular functions and neuronal phenotype in the adult brain and during brain aging. Neuron 2010, 68, 409–427. [Google Scholar] [CrossRef] [Green Version]
  206. Proebstl, D.; Voisin, M.B.; Woodfin, A.; Whiteford, J.; D’Acquisto, F.; Jones, G.E.; Rowe, D.; Nourshargh, S. Pericytes support neutrophil subendothelial cell crawling and breaching of venular walls in vivo. J. Exp. Med. 2012, 209, 1219–1234. [Google Scholar] [CrossRef] [Green Version]
  207. Nelson, A.R.; Sagare, M.A.; Wang, Y.; Kisler, K.; Zhao, Z.; Zlokovic, B.V. Channelrhodopsin Excitation Contracts Brain Pericytes and Reduces Blood Flow in the Aging Mouse Brain in vivo. Front. Aging Neurosci. 2020, 12, 108. [Google Scholar] [CrossRef]
  208. Zhang, Y.; Zhang, X.; Wei, Q.; Leng, S.; Li, C.; Han, B.; Bai, Y.; Zhang, H.; Yao, H. Activation of Sigma-1 Receptor Enhanced Pericyte Survival via the Interplay Between Apoptosis and Autophagy: Implications for Blood-Brain Barrier Integrity in Stroke. Transl. Stroke Res. 2020, 11, 267–287. [Google Scholar] [CrossRef]
  209. Bertrand, L.; Cho, H.J.; Toborek, M. Blood-brain barrier pericytes as a target for HIV-1 infection. Brain 2019, 142, 502–511. [Google Scholar] [CrossRef]
Figure 1. Schematic depicting the interplay of drugs of abuse, HIV-1/HIV-1 proteins, and cART in the context of autophagy and neuroinflammation.
Figure 1. Schematic depicting the interplay of drugs of abuse, HIV-1/HIV-1 proteins, and cART in the context of autophagy and neuroinflammation.
Viruses 15 00044 g001
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Sil, S.; Thangaraj, A.; Oladapo, A.; Hu, G.; Kutchy, N.A.; Liao, K.; Buch, S.; Periyasamy, P. Role of Autophagy in HIV-1 and Drug Abuse-Mediated Neuroinflammaging. Viruses 2023, 15, 44. https://doi.org/10.3390/v15010044

AMA Style

Sil S, Thangaraj A, Oladapo A, Hu G, Kutchy NA, Liao K, Buch S, Periyasamy P. Role of Autophagy in HIV-1 and Drug Abuse-Mediated Neuroinflammaging. Viruses. 2023; 15(1):44. https://doi.org/10.3390/v15010044

Chicago/Turabian Style

Sil, Susmita, Annadurai Thangaraj, Abiola Oladapo, Guoku Hu, Naseer A Kutchy, Ke Liao, Shilpa Buch, and Palsamy Periyasamy. 2023. "Role of Autophagy in HIV-1 and Drug Abuse-Mediated Neuroinflammaging" Viruses 15, no. 1: 44. https://doi.org/10.3390/v15010044

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

Article Metrics

Back to TopTop