Skip to main content

MINI REVIEW article

Front. Pharmacol., 28 October 2024
Sec. Neuropharmacology
This article is part of the Research Topic World No-Tobacco: Effects of Tobacco and Nicotine on the Brain View all 7 articles

The impacts of tobacco and nicotine on HIV-1 infection, inflammation, and the blood-brain barrier in the central nervous system

  • 1Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
  • 2Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States

Human immunodeficiency virus (HIV-1) remains a persistent global health crisis. Even while successfully virologically suppressed, people with HIV (PWH) experience a higher risk for inflammatory disorders such as HIV-associated neurocognitive disorder (HAND). Tobacco use puts PWH at higher risk for neurocognitive symptoms resulting from HIV-associated neuroinflammation. The NLR Family Pyrin Domain Containing 3 (NLRP3) inflammasome has been implicated as a driver of HIV-associated inflammation, including HAND. Nicotine, the psychoactive component of tobacco smoke, has also been shown to signal through the NLRP3 inflammasome and modulate inflammatory signaling in the CNS. Here, we explore the impacts of nicotine and tobacco on the complex neurobiology of HAND, including effects on cognition, inflammation, viral latency, and blood-brain barrier integrity. We outline nicotine’s role in the establishment of active and latent infection in the brain and posit the NLRP3 inflammasome as a common pathway by which HIV-1 and nicotine promote neuroinflammation in PWH.

Introduction

Human immunodeficiency virus (HIV-1) affects more than 39 million people worldwide and remains incurable despite major advances in antiretroviral therapy (ART). Although patient life expectancy and quality of life has dramatically improved over the past few decades, People with HIV (PWH) are still at greater risk for several comorbid conditions due to viral persistence in latent reservoirs and associated low-level chronic inflammation (Chun et al., 2010; Collora and Ho, 2022; Global, 2024; Lee et al., 2020; Siliciano et al., 2003; White et al., 2022). These include a variety of conditions related to aging, including cardiovascular disease, osteoporosis, cancer, and neurocognitive disorders (Aberg, 2012; Guaraldi et al., 2011; Heaton et al., 2010; Kaplan-Lewis et al., 2017; Kearns et al., 2017). PWH also tend to develop these conditions earlier than the general population (Aberg, 2012; Kaplan-Lewis et al., 2017). Chronic inflammation due to persistent infection in viral reservoirs such as the central nervous system (CNS) is thought to be the primary driver of this increased risk (Desplats et al., 2013; Sonti et al., 2021).

HIV-associated neurocognitive Disorder (HAND) is a common HIV-associated condition affecting 20%–50% of PWH (Heaton et al., 2010). HAND is characterized by increased neuroinflammation, blood-brain barrier (BBB) breakdown, metabolic dysfunction, and measurable cognitive impairment (Saylor et al., 2016). Drug use, including smoking, is a risk factor for the development of HAND in PWH (Saylor et al., 2016). Tobacco users with HIV experience poorer outcomes, including increased risk of mortality and virologic rebound and poorer response to cART (Han et al., 2018). Because of the high prevalence of tobacco use in PWH and its known detrimental effects, unraveling its role in neuroinflammation will provide much-needed insights into mechanisms and possible therapeutic strategies for PWH.

Biology of HAND: Chronic inflammation in the CNS

People with HIV are more likely to exhibit markers of inflammation even when viral levels are suppressed below detection by long-term ART treatments (Aberg, 2012; Deeks, 2011; Sieg et al., 2021). In virologically suppressed individuals, replication persists in tissues where ART has limited penetrance (Kulpa and Chomont, 2024). Despite ART, this leads to persistent immune activation, senescence, and an increased systemic inflammatory milieu (Aberg, 2012; Deeks, 2011). The brain is a viral reservoir with unique characteristics due to its immune-privileged status and thus presents a distinct set of challenges for a sterilizing cure. Although improved access to ART has greatly improved the quality of life and life expectancy for PWH, even complete viral suppression under ART treatment does not eliminate the cognitive symptoms associated with HIV infection of the brain (Heaton et al., 2010). HAND is defined clinically as a spectrum of disorders ranging from Asymptomatic Neurocognitive Impairment (ANI) to Mild Neurocognitive Disorder (MND) to HIV-associated Dementia (HAD), the most severe presentation (Heaton et al., 2011). Clinical studies have shown that biomarkers associated with CNS inflammation in blood plasma and cerebrospinal fluid (CSF) are more prevalent during acute HIV infection and decline over time and with ART treatment (Longino et al., 2022). As access to earlier ART intervention improves, rates of the more severe forms of HAND have been declining. Still, it is estimated that between 20% and 50% of PWH experience some form of neurocognitive impairment due to chronic HIV infection (Heaton et al., 2010).

Neuroinflammation in the HIV-1 infected CNS is characterized by BBB dysfunction, immune cell infiltration, and infection and inflammatory signaling of resident CNS cells, particularly microglia (Khanal et al., 2021; Saylor et al., 2016; Sreeram et al., 2022). Infected microglia compose the bulk of the viral reservoir in the CNS, and HIV is seeded into the brain in the very early stages of infection, primarily by infected monocytes, which migrate across the blood-brain barrier (Davis et al., 1992; Kahn and Walker, 1998; Longino et al., 2022; Valcour et al., 2012). Damage to the blood-brain barrier is a key aspect of many neuroinflammatory conditions, and chronic neuroinflammation from HIV infection is no exception. BBB dysfunction persists even in the presence of ART and during chronic infection (Kulpa and Chomont, 2024). Brain microvascular endothelial cells (BMVECs), pericytes, and astrocytes, are all dysregulated by the presence of virus in the CNS (Andersson et al., 2001; Eugenin et al., 2011; Leibrand et al., 2017; Osborne et al., 2020; Piekna-Przybylska et al., 2019). Endothelial cells are prone to dysfunction and death due to direct interaction with viral proteins and exposure to inflammatory cytokines released by infected cells (Andersson et al., 2001; Lee et al., 2004; Leibrand et al., 2017). Dysregulation of endothelial tight junction proteins such as claudin-5, occludin, and ZO-1 leads to increased BBB permeability and may facilitate infiltration of peripheral immune cells to the CNS (Boven et al., 2000; Chaudhuri et al., 2008; Dallasta et al., 1999; Eugenin et al., 2011). Decreased pericytes coverage of the endothelium and infected or dysregulated astrocytes have also been shown to contribute to barrier dysfunction in the HIV-infected brain (Piekna-Przybylska et al., 2019; Pla-Tenorio et al., 2023; Valdebenito et al., 2021). Although HIV can both cross and dysregulate the BBB, BBB penetrance remains a complex problem for the efficacy of antiretroviral drugs. The viral reservoir in the brain is formed by a combination of this challenge for ART delivery and the development of a population of latently infected cells, primarily microglia, which harbor HIV provirus (Osborne et al., 2020).

As the cell population most susceptible to HIV infection in the brain, microglia form the bulk of the latent reservoir (Wallet et al., 2019). An established literature shows a relationship between HIV-1 proteins, neuronal death, and microglial activation. Transgenic mouse studies show that viral proteins such as Tat and gp120 are produced by infected cells and have neurotoxic properties (Leibrand et al., 2017). Additionally, infected human microglia become reactive and release pro-inflammatory cytokines such as TNF⍺, CCL2, IL-1β, IL-6, and CCL5 (Alvarez-Carbonell et al., 2019). Infected microglia are also prone to mitochondrial dysfunction and overproduction of ROS (Alvarez-Carbonell et al., 2019; Borrajo et al., 2021).

Recent models suggest that cyclical reactivation of latent provirus in microglia may cause the ongoing cascade of inflammation in HAND (Sreeram et al., 2022). The susceptibility of microglial cells to latent infection is thought to be tied to the activation state at the time of infection, with more quiescent cells favoring a latent status than reactive or activated cells (Sreeram et al., 2022; Wallet et al., 2019). Activation of the transcription factor NF-κB or IRF3 in response to an inflammatory stimulus can reactivate viral replication in a latently infected human microglial cell line (Alvarez-Carbonell et al., 2017). Thus, inflammatory stimuli such as damaged neurons or drug exposures can potentiate latency reversal and promote the chronic inflammation characteristic of HAND (Alvarez-Carbonell et al., 2019). We will further explore the role of NLRP3 inflammasome signaling and nicotine exposures in driving this complex inflammatory cascade.

HIV and the NLRP inflamamsome

The NLR Family Pyrin Domain Containing 3 (NLRP3) inflammasome, which consists of NLRP3, ASC, and pro-caspase-1, has been implicated in HAND and other HIV-associated inflammatory pathologies. Upon activation by intracellular pathogens and binding of Damage-associated Molecular Patterns (DAMP) to PRRs (Pattern Recognition Receptors), the inflammasome activates caspase-1, leading to the release of pro-inflammatory cytokines (IL-1β and Il-18) and pyroptosis, an inflammatory form of cell death known to contribute to inflammation in PWH (Doitsh et al., 2014; Katuri et al., 2019; Mamik and Power, 2017). HIV infection in T-cells may lead to incomplete transcription and viral fragments, triggering PRRs and leading to NLRP3 activation, inflammatory cytokine release, and cell death (Doitsh et al., 2014). In myeloid cells, NLRP3 activation and inflammatory cytokine release are associated with many HIV-associated inflammatory processes, such as atherosclerosis (Mao et al., 2021; Mullis and Swartz, 2020).

Animal studies in the CNS have shown that the NLRP3 inflammasome can be activated in microglia in response to viral proteins such as Tat, Vpr, and gp120, resulting in inflammatory cytokine release and neuroinflammation (Chivero et al., 2017; He et al., 2020). HIV infection was shown to promote IL-1β release in primary human microglia (Walsh et al., 2014). Furthermore, NLRP3 inflammasome inhibitors have shown a therapeutic effect in a mouse HAND model (He et al., 2020; Mamik and Power, 2017).

Alongside microglia-induced inflammatory signaling, the NLRP3 inflammasome is also implicated in BBB dysregulation. HIV-1 dysregulates the BBB through a variety of mechanisms, from direct interaction of brain microvascular endothelial cells (BMVECs) with viral proteins to inflammatory cytokine release (Il-6, TNF-a, IL-1β) from infected microglia and infiltrating monocytes in the CNS (Atluri et al., 2015; Caligaris et al., 2021). Studies indicate that HIV readily establishes latency in microglia and that microglial activation by NLRP3-associated inflammatory cytokines such as TNF-α and IL-1β may contribute to viral emergence from proviral latency, contributing to the chronic inflammation of HAND (Alvarez-Carbonell et al., 2017; Alvarez-Carbonell et al., 2019; Ko et al., 2019; Li and Barres, 2018; Plaza-Jennings et al., 2022; Sreeram et al., 2022).

Nicotine and inflammation

Nicotine and its effects in the context of HAND are particularly of interest due to the high prevalence of smoking among PWH and the heightened health risks experienced by PWH who smoke. The established literature points to tobacco use as contributing to inflammation in a variety of contexts, both inside and outside of the CNS. However, its pro-inflammatory and anti-inflammatory effects can be disease and context dependent. Nicotine signals through the Nicotinic Acetylcholine receptor family (nAChRs), which are involved in cholinergic signaling and reward stimulus (de Kloet et al., 2015). They are also widely expressed in various cell types, including monocytes, macrophages, and microglia (Richter and Grau, 2023; Suzuki et al., 2006; Zoli et al., 2018). In non-excitable cells, signaling is primarily mediated through the influx of calcium ions directly through the ion-gated channel of the receptor (Zoli et al., 2018). In different non-neuronal cell types, signaling can be ionically or non-ionically driven and trigger downstream signaling via various pathways (Sorimachi et al., 1994; Zia et al., 2000; Zoli et al., 2018).

The general pro- and anti-inflammatory properties of nicotine are complex and context-dependent, with factors such as cell type, receptor type, and disease context playing important roles. In many studies, nicotine exhibits anti-inflammatory properties due to cholinergic signaling in neurons and non-neuronal cells (Zoli et al., 2018). However, the literature consistently links nicotine and NLRP3 inflammasome activation, especially in myeloid-derived cell types. There is a well-established relationship between nicotine and NLRP3-driven atherosclerosis progression (Duan et al., 2021; Mullis and Swartz, 2020; Wu et al., 2018; Xu et al., 2021). One study demonstrated that nicotine promotes NLRP3 inflammasome activation in peripheral myeloid cells, driving atherosclerosis progression due to inflammatory cytokine release in macrophages and monocytes (Mao et al., 2021). Nicotine was also shown to exacerbate proliferation and cell migration in lung adenocarcinoma via a5-nAChR and NLRP3 signaling (Jia et al., 2022).

In the context of the CNS, nicotine exerts direct pro-inflammatory effects on both microglia and the vasculature of the BBB. Exposure to NNKs (nicotine-derived nitrosamine ketones, a nicotine-derived metabolite) increased ROS and inflammatory cytokine release in mouse microglia (Ghosh et al., 2009). Tobacco use is generally associated with BBB disruption, CNS oxidative stress, and decreased cognitive performance in clinical studies (Ande et al., 2013; Ghosh et al., 2009; Louboutin and Strayer, 2014; Mazzone et al., 2010). BBB damage due to nicotine exposure is primarily linked to mitochondrial dysfunction, ROS production, and release of inflammatory cytokines such as TNFa and IL-6, which can trigger endothelial dysfunction by dysregulation of tight junction proteins such as claudin-5 and occludin (Hossain et al., 2011; Hutamekalin et al., 2008; Kousik et al., 2012; Manda, Mittapalli, Geldenhuys, et al., 2010; Pimentel et al., 2020). Additionally, Zhang et al. have linked the NLRP3 inflammasome directly to nicotine-induced endothelial barrier dysfunction and hyperpermeability via the release of HMGB1 (Zhang et al., 2019).

HIV and nicotine in HAND

While nicotine and HIV alone both contribute to neuroinflammation and BBB dysfunction, in PWH who smoke, additive effects have been observed on neuroinflammation and decreased cognitive performance (Bryant et al., 2013; Chang et al., 2020). These clinical findings are supported by studies in HIV-1 transgenic rat models, where nicotine promotes overexpression of immune-related genes and inflammatory cytokine expression (Royal et al., 2018; Yang et al., 2016). Additionally, Delgado-Velez et al. showed that gp120 exposure can directly upregulate the expression of a7-nAChRs in peripheral immune cells (Delgado-Vélez et al., 2015). In addition to promoting their upregulation, the viral protein gp120 can bind to the α7 receptors. α7-nAChR signaling promoted amyloid-beta accumulation in an HIV-gp120 mouse model (Liu et al., 2017). While certain animal studies show that nicotine alone increases cognitive performance in behavioral tests, this improvement is ameliorated in HIV-1 Tg rats (Nizri et al., 2009; Revathikumar et al., 2016; Royal et al., 2018; Yang et al., 2016). Along a similar vein, α7-nAChRs were found to be upregulated in the monocytes of women with HIV, but greater receptor abundance did not result in a protective anti-inflammatory effect when MDMs were treated with LPS (Delgado-Vélez et al., 2015). These findings, when taken together, suggest that the presence of HIV may alter the cholinergic anti-inflammatory response via the α7-nAChRs, dampening or even reversing the anti-inflammatory effects of nicotine alone.

It is important to consider that in people who smoke cigarettes, nicotine is always accompanied by the thousands of other compounds present in tobacco smoke. Tobacco smoke and nicotine alone have been shown to have differing effects on inflammation, and their respective contributions to HIV associated neuroinflammation are not yet clearly delineated. Tobacco smoke is almost universally pro-inflammatory in both pre-clinical and clinical studies. Cigarette smoke condensate increases apoptosis, viral replication, and oxidative stress in human monocytes (Rao et al., 2016). Tobacco smoke extract similarly increases viral replication in bronchial epithelial cells and alveolar macrophages (Abbud et al., 1995; Chinnapaiyan et al., 2018). In clinical studies, PWH who smoke exhibit higher levels of inflammatory markers, increased viral load, and increased risk of HAND compared to non-smokers (Han et al., 2018; Valiathan et al., 2014; Wojna et al., 2007). As mentioned above, nicotine can induce anti-inflammatory cholinergic signaling through the ⍺7 nAChR and can be neuroprotective in certain contexts, including in some HIV-1 Tg rat studies (Cao et al., 2013; Cao et al., 2016). However, chronic nicotine exposure is known to cause BBB disruption, a key component of HAND pathology (Feldman and Anderson, 2013). Furthermore, there is evidence that nicotine treatment promotes HIV infection in human microglia (Rock et al., 2008). Further studies with both nicotine and tobacco smoke are needed to unravel their roles in the context of HAND.

In addition to the many compounds present in tobacco smoke, antiretroviral drug treatments and other substance use in PWH add another layer of complexity to the neuroinflammatory context of the HIV-infected CNS. Many studies have shown that earlier intervention with ART can significantly reduce the risk of more severe forms of HAND in PWH (Brew, 2004; Sacktor et al., 2002). However, as outlined earlier, infection and inflammation persist in the brain despite the presence of ART. As reviewed elsewhere, certain antiretroviral drugs can have neurotoxic effects through mechanisms such as oxidative stress and mitochondrial dysfunction, and balancing this neurotoxicity against viral suppression and penetrance of the CNS reservoir remains an important clinical challenge (Shah et al., 2016; Yuan and Kaul, 2021). In PWH who smoke tobacco, there is the additional factor of drug-drug interactions between nicotine and ARTs (reviewed by Ghura et al., 2020). Nicotine has been shown to affect ART metabolism directly and can impact drug delivery by compromising BBB integrity (Kumar et al., 2015; Manda, Mittapalli, Bohn, et al., 2010; Manda, Mittapalli, Geldenhuys, et al., 2010; Pal et al., 2011). Animal model studies with the protease inhibitor saquinavir demonstrate that nicotine-induced BBB compromise may facilitate entry of ART into the CNS, but nicotine and saquinavir cause additive oxidative stress in the brain endothelium leading to dysregulation of Notch-4 and ZO-1 (Manda, Mittapalli, Bohn, et al., 2010; Manda, Mittapalli, Geldenhuys, et al., 2010). Balancing ART toxicity with BBB penetrance remains an important clinical problem, as long-term exposure to certain ARTs has been associated with neurovascular toxicity (Bertrand et al., 2021). Thus, although nicotine’s ability to disrupt the BBB may lead to greater drug penetrance, it is likely that the additive impacts on BBB dysfunction and subsequent inflammation and immune infiltration of the CNS represent an overall detrimental outcome (Ahmed et al., 2018; Bertrand et al., 2021). ART has also been observed to increase nicotine metabolism in PWH (Ashare et al., 2019; Earla et al., 2014). ARTs may also impact nicotine signaling by acting on nicotinic receptors. For example, the protease inhibitor indinavir was demonstrated to impact cholinergic signaling by inhibiting the α7-nAChR activity (Ekins et al., 2017). More investigation is necessary to understand the specific interactions between various ARTs and nicotine to inform best practices for the treatment of PWH.

As outlined here, the NLRP3 inflammasome pathway is a common mechanism by which HIV and nicotine promote neuroinflammation. More studies are needed to fully understand how HIV and nicotine interact to promote inflammation, BBB dysregulation, and viral reactivation in the CNS.

Discussion

Chronic inflammation leads to increased risks for comorbid disorders in PWH, even in the presence of ART (Chun et al., 2010; Collora and Ho, 2022; Lee et al., 2020; Siliciano et al., 2003; White et al., 2022). In the brain, this manifests as an increased risk for neuroinflammation and cognitive impairment classified under the family of neurocognitive disorders known as HAND (Heaton et al., 2011; Saylor et al., 2016). The persistence of HIV in viral reservoirs such as the CNS is a primary driver of this chronic inflammation (Desplats et al., 2013; Sonti et al., 2021). The use of substances such as tobacco increases the risk of comorbidities for PWH and further contributes to HIV-associated inflammatory pathologies (Han et al., 2018).

There is a well-established literature linking HIV-1 with NLRP3 inflammasome signaling both in the periphery and in the CNS. Studies have shown that HIV can stimulate the inflammasome through the binding of viral fragments and proteins to PRRs and through the activation of purinergic receptors (Doitsh et al., 2014; Freeman and Swartz, 2020; Swartz et al., 2015). In the brain, HIV drives NLRP3-associated inflammation primarily through infected and activated microglia, which contribute to the inflammatory environment by releasing neurotoxic factors and pro-inflammatory cytokines (Chivero et al., 2017; He et al., 2020; Walsh et al., 2014). HIV also drives BBB damage through several mechanisms, including NLRP3-driven dysregulation of endothelial cells (Atluri et al., 2015; Caligaris et al., 2021).

NLRP3 inflammasome activation, a key feature of neuroinflammation and BBB dysregulation, is influenced by both HIV and nicotine. Like HIV infection, nicotine is known to drive endothelial dysfunction by promoting ROS, mitochondrial dysfunction, and NLRP3 inflammasome activation (Ghosh et al., 2009; Hossain et al., 2011; Hutamekalin et al., 2008; Kousik et al., 2012; Manda, Mittapalli, Bohn, et al., 2010; Pimentel et al., 2020; Zhang et al., 2019). There is evidence that nicotine in tobacco enhances viral replication in microglia and macrophages and has been shown to cause activation of these resident immune cells and compromise the BBB (Ghosh et al., 2009; Manda, Mittapalli, Geldenhuys, et al., 2010; Pimentel et al., 2020; Rock et al., 2008). Taken together, the literature suggests that the combined effect of HIV and nicotine-driven NLRP3 activation plays a key role in the heightened neuroinflammation and cognitive symptoms observed in PWH who smoke.

Further investigation is necessary to understand the mechanism of HIV and nicotine’s interactions in the CNS and the precise role of NLRP3 in driving their combined effects. Furthermore, there are many critical unanswered questions surrounding the impact of substance use on the establishment and reactivation of viral latency in the CNS. Atluri et al. suggest that nicotine increases the risk of viral latency establishment due to the upregulation of HDAC2 in a neuronal cell line (Atluri et al., 2014). In this model, the transcriptional repressor HDAC2 is synergistically upregulated by HIV and nicotine, leading to more compact chromatin organization, reduced gene transcription, and increased latent infection. Treatment with the HDAC inhibitor vorinostat reversed this effect and reactivated latent virus. However, is important to note that the biological relevance of this study is limited, as neurons are not typically infected with HIV-1 and do not form a significant portion of the CNS viral reservoir. In a murine macrophage model of atherosclerosis, another histone deacetylase, HDAC6, was shown to promote nicotine-mediated inflammation and pyroptosis via deacetylation of p65, and activation of NF-kB and NLRP3 transcription (Xu et al., 2021). HIV-1 is known to integrate itself into transcriptionally active regions of the genome located in regions of open chromatin (Schröder et al., 2002). It relies on host cell machinery and activation states to regulate its latency, favoring activated over quiescent states in both microglia and CD4+ T-cells (Mbonye and Karn, 2017; Sreeram et al., 2022; Wallet et al., 2019). All of this considered, it seems likely that nicotine may play a role in regulating viral latency in microglia by promoting activation of NF-kB and NLRP3 associated genes, leading to transcription of latent provirus. However, nicotine’s specific role in viral latency formation and maintenance in the CNS remains largely unexplored in the literature, and thorough investigation of its impacts on latent infection in microglia is especially necessary.

There are additionally many open questions regarding poly-substance use in PWH and the impacts of combined drug use on cognitive impairment in these patients. As previously reviewed by our group, cannabis use is largely associated with protective effects against inflammation in PWH, and CB2R signaling has been linked to reduced HIV-1 infection and NLRP3 inflammasome activation (Min et al., 2023). Further study is needed to better understand the interactions between the cannabinoid system, nicotine, and NLRP3 and the impacts of multiple drug exposures on neuroinflammation in PWH. Investigating these questions will provide valuable insights into the mechanism of nicotine’s impact on the pathogenesis of HIV-1-associated neurodegeneration, informing possibilities for future therapeutic development.

Author contributions

AK: Conceptualization, Data curation, Formal Analysis, Investigation, Visualization, Writing–original draft, Writing–review and editing. TS: Conceptualization, Data curation, Formal Analysis, Funding acquisition, Investigation, Methodology, Project administration, Resources, Software, Supervision, Validation, Visualization, Writing–original draft, Writing–review and editing.

Funding

The author(s) declare that financial support was received for the research, authorship, and/or publication of this article. The authors receive funding from the following sources: R01DA052255 (TS), R01DA059876 (TS), R01MH134319 (TS), R01DA054526 (TS).

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

Abbreviations

PWH, People with HIV; BBB, Blood brain barrier; nAChR, nicotinic acetylcholine receptor; HAND, HIV-associated neurocognitive disorder; ART, antiretroviral therapy.

References

Abbud, R. A., Finegan, C. K., Guay, L. A., and Rich, E. A. (1995). Enhanced production of human immunodeficiency virus type 1 by in vitro-infected alveolar macrophages from otherwise healthy cigarette smokers. J. Infect. Dis. 172 (3), 859–863. doi:10.1093/infdis/172.3.859

PubMed Abstract | CrossRef Full Text | Google Scholar

Aberg, J. A. (2012). Aging, inflammation, and HIV infection. Top. Antivir. Med. 20 (3), 101–105.

PubMed Abstract | Google Scholar

Ahmed, D., Roy, D., and Cassol, E. (2018). Examining relationships between metabolism and persistent inflammation in HIV patients on antiretroviral therapy. Mediat. Inflamm. 2018, 6238978. doi:10.1155/2018/6238978

PubMed Abstract | CrossRef Full Text | Google Scholar

Alvarez-Carbonell, D., Garcia-Mesa, Y., Milne, S., Das, B., Dobrowolski, C., Rojas, R., et al. (2017). Toll-like receptor 3 activation selectively reverses HIV latency in microglial cells. Retrovirology 14 (1), 9. doi:10.1186/s12977-017-0335-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Alvarez-Carbonell, D., Ye, F., Ramanath, N., Garcia-Mesa, Y., Knapp, P. E., Hauser, K. F., et al. (2019). Cross-talk between microglia and neurons regulates HIV latency. PLoS Pathog. 15 (12), e1008249. doi:10.1371/journal.ppat.1008249

PubMed Abstract | CrossRef Full Text | Google Scholar

Ande, A., McArthur, C., Kumar, A., and Kumar, S. (2013). Tobacco smoking effect on HIV-1 pathogenesis: role of cytochrome P450 isozymes. Expert Opin. Drug Metabolism and Toxicol. 9 (11), 1453–1464. doi:10.1517/17425255.2013.816285

PubMed Abstract | CrossRef Full Text | Google Scholar

Andersson, L. M., Hagberg, L., Fuchs, D., Svennerholm, B., and Gisslén, M. (2001). Increased blood-brain barrier permeability in neuro-asymptomatic HIV-1-infected individuals—correlation with cerebrospinal fluid HIV-1 RNA and neopterin levels. J. Neurovirology 7 (6), 542–547. doi:10.1080/135502801753248123

PubMed Abstract | CrossRef Full Text | Google Scholar

Ashare, R. L., Thompson, M., Leone, F., Metzger, D., Gross, R., Mounzer, K., et al. (2019). Differences in the rate of nicotine metabolism among smokers with and without HIV. AIDS Lond. Engl. 33 (6), 1083–1088. doi:10.1097/QAD.0000000000002127

PubMed Abstract | CrossRef Full Text | Google Scholar

Atluri, V. S. R., Hidalgo, M., Samikkannu, T., Kurapati, K. R. V., Jayant, R. D., Sagar, V., et al. (2015). Effect of human immunodeficiency virus on blood-brain barrier integrity and function: an update. Front. Cell. Neurosci. 9, 212. doi:10.3389/fncel.2015.00212

PubMed Abstract | CrossRef Full Text | Google Scholar

Atluri, V. S. R., Pilakka-Kanthikeel, S., Samikkannu, T., Sagar, V., Kurapati, K. R. V., Saxena, S. K., et al. (2014). Vorinostat positively regulates synaptic plasticity genes expression and spine density in HIV infected neurons: role of nicotine in progression of HIV-associated neurocognitive disorder. Mol. Brain 7 (1), 37. doi:10.1186/1756-6606-7-37

PubMed Abstract | CrossRef Full Text | Google Scholar

Bertrand, L., Velichkovska, M., and Toborek, M. (2021). Cerebral vascular toxicity of antiretroviral therapy. J. Neuroimmune Pharmacol. 16 (1), 74–89. doi:10.1007/s11481-019-09858-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Borrajo, A., Spuch, C., Penedo, M. A., Olivares, J. M., and Agís-Balboa, R. C. (2021). Important role of microglia in HIV-1 associated neurocognitive disorders and the molecular pathways implicated in its pathogenesis. Ann. Med. 53 (1), 43–69. doi:10.1080/07853890.2020.1814962

PubMed Abstract | CrossRef Full Text | Google Scholar

Boven, L. A., Middel, J., Verhoef, J., De Groot, C. J. A., and Nottet, H. S. L. M. (2000). Monocyte infiltration is highly associated with loss of the tight junction protein zonula occludens in HIV-1-associated dementia. Neuropathology Appl. Neurobiol. 26 (4), 356–360. doi:10.1046/j.1365-2990.2000.00255.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Brew, B. J. (2004). Evidence for a change in AIDS dementia complex in the era of highly active antiretroviral therapy and the possibility of new forms of AIDS dementia complex. AIDS Lond. Engl. 18 (Suppl. 1), S75–S78. doi:10.1097/00002030-200401001-00011

PubMed Abstract | CrossRef Full Text | Google Scholar

Bryant, V. E., Kahler, C. W., Devlin, K. N., Monti, P. M., and Cohen, R. A. (2013). The effects of cigarette smoking on learning and memory performance among people living with HIV/AIDS. AIDS Care 25 (10), 1308–1316. doi:10.1080/09540121.2013.764965

PubMed Abstract | CrossRef Full Text | Google Scholar

Caligaris, G., Trunfio, M., Ghisetti, V., Cusato, J., Nigra, M., Atzori, C., et al. (2021). Blood–brain barrier impairment in patients living with HIV: predictors and associated biomarkers. Diagnostics 11 (5), 867. Article 5. doi:10.3390/diagnostics11050867

PubMed Abstract | CrossRef Full Text | Google Scholar

Cao, J., Nesil, T., Wang, S., Chang, S. L., and Li, M. D. (2016). Expression profile of nicotinic acetylcholine receptor subunits in the brain of HIV-1 transgenic rats given chronic nicotine treatment. J. Neurovirology 22 (5), 626–633. doi:10.1007/s13365-016-0438-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Cao, J., Wang, S., Wang, J., Cui, W., Nesil, T., Vigorito, M., et al. (2013). RNA deep sequencing Analysis reveals that nicotine restores impaired gene expression by viral proteins in the brains of HIV-1 transgenic rats. PLOS ONE 8 (7), e68517. doi:10.1371/journal.pone.0068517

PubMed Abstract | CrossRef Full Text | Google Scholar

Chang, L., Liang, H., Kandel, S. R., and He, J. J. (2020). Independent and combined effects of nicotine or chronic tobacco smoking and HIV on the brain: a review of preclinical and clinical studies. J. Neuroimmune Pharmacol. 15 (4), 658–693. doi:10.1007/s11481-020-09963-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Chaudhuri, A., Duan, F., Morsey, B., Persidsky, Y., and Kanmogne, G. D. (2008). HIV-1 activates proinflammatory and interferon-inducible genes in human brain microvascular endothelial cells: putative mechanisms of blood—brain barrier dysfunction. J. Cereb. Blood Flow and Metabolism 28 (4), 697–711. doi:10.1038/sj.jcbfm.9600567

PubMed Abstract | CrossRef Full Text | Google Scholar

Chinnapaiyan, S., Dutta, R., Bala, J., Parira, T., Agudelo, M., Nair, M., et al. (2018). Cigarette smoke promotes HIV infection of primary bronchial epithelium and additively suppresses CFTR function. Sci. Rep. 8 (1), 7984. doi:10.1038/s41598-018-26095-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Chivero, E. T., Guo, M. L., Periyasamy, P., Liao, K., Callen, S. E., and Buch, S. (2017). HIV-1 tat primes and activates microglial NLRP3 inflammasome-mediated neuroinflammation. J. Neurosci. 37 (13), 3599–3609. doi:10.1523/JNEUROSCI.3045-16.2017

PubMed Abstract | CrossRef Full Text | Google Scholar

Chun, T.-W., Justement, J. S., Murray, D., Hallahan, C. W., Maenza, J., Collier, A. C., et al. (2010). Rebound of plasma viremia following cessation of antiretroviral therapy despite profoundly low levels of HIV reservoir: implications for eradication. AIDS Lond. Engl. 24 (18), 2803–2808. doi:10.1097/QAD.0b013e328340a239

PubMed Abstract | CrossRef Full Text | Google Scholar

Collora, J. A., and Ho, Y.-C. (2022). The loud minority: transcriptionally active HIV-1-infected cells survive, proliferate, and persist. Cell 185 (2), 227–229. doi:10.1016/j.cell.2021.12.038

PubMed Abstract | CrossRef Full Text | Google Scholar

Dallasta, L. M., Pisarov, L. A., Esplen, J. E., Werley, J. V., Moses, A. V., Nelson, J. A., et al. (1999). Blood-brain barrier tight junction disruption in human immunodeficiency virus-1 encephalitis. Am. J. Pathology 155 (6), 1915–1927. doi:10.1016/S0002-9440(10)65511-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Davis, L., Hjelle, B., Miller, V., Palmer, D., Llewellyn, A., Tl, M., et al. (1992). Early viral brain invasion in iatrogenic human immunodeficiency virus infection. Neurology 42 (9), 1736–1739. doi:10.1212/wnl.42.9.1736

PubMed Abstract | CrossRef Full Text | Google Scholar

Deeks, S. G. (2011). HIV infection, inflammation, immunosenescence, and aging. Annu. Rev. Med. 62, 141–155. doi:10.1146/annurev-med-042909-093756

PubMed Abstract | CrossRef Full Text | Google Scholar

de Kloet, S. F., Mansvelder, H. D., and De Vries, T. J. (2015). Cholinergic modulation of dopamine pathways through nicotinic acetylcholine receptors. Biochem. Pharmacol. 97 (4), 425–438. doi:10.1016/j.bcp.2015.07.014

PubMed Abstract | CrossRef Full Text | Google Scholar

Delgado-Vélez, M., Báez-Pagán, C. A., Gerena, Y., Quesada, O., Santiago-Pérez, L. I., Capó-Vélez, C. M., et al. (2015). The α7-nicotinic receptor is upregulated in immune cells from HIV-seropositive women: consequences to the cholinergic anti-inflammatory response. Clin. and Transl. Immunol. 4 (12), e53. doi:10.1038/cti.2015.31

CrossRef Full Text | Google Scholar

Desplats, P., Dumaop, W., Smith, D., Adame, A., Everall, I., Letendre, S., et al. (2013). Molecular and pathologic insights from latent HIV-1 infection in the human brain. Neurology 80 (15), 1415–1423. doi:10.1212/WNL.0b013e31828c2e9e

PubMed Abstract | CrossRef Full Text | Google Scholar

Doitsh, G., Galloway, N. L. K., Geng, X., Yang, Z., Monroe, K. M., Zepeda, O., et al. (2014). Cell death by pyroptosis drives CD4 T-cell depletion in HIV-1 infection. Nature 505 (7484), 509–514. doi:10.1038/nature12940

PubMed Abstract | CrossRef Full Text | Google Scholar

Duan, F., Zeng, C., Liu, S., Gong, J., Hu, J., Li, H., et al. (2021). α1-nAchR-Mediated signaling through lipid raft is required for nicotine-induced NLRP3 inflammasome activation and nicotine-accelerated atherosclerosis. Front. Cell Dev. Biol. 9, 724699. doi:10.3389/fcell.2021.724699

PubMed Abstract | CrossRef Full Text | Google Scholar

Earla, R., Ande, A., McArthur, C., Kumar, A., and Kumar, S. (2014). Enhanced nicotine metabolism in HIV-1-positive smokers compared with HIV-negative smokers: simultaneous determination of nicotine and its four metabolites in their plasma using a simple and sensitive electrospray ionization liquid chromatography-tandem mass spectrometry technique. Drug Metabolism Dispos. Biol. Fate Chem. 42 (2), 282–293. doi:10.1124/dmd.113.055186

PubMed Abstract | CrossRef Full Text | Google Scholar

Ekins, S., Mathews, P., Saito, E. K., Diaz, N., Naylor, D., Chung, J., et al. (2017). α7-Nicotinic acetylcholine receptor inhibition by indinavir: implications for cognitive dysfunction in treated HIV disease. AIDS Lond. Engl. 31 (8), 1083–1089. doi:10.1097/QAD.0000000000001488

PubMed Abstract | CrossRef Full Text | Google Scholar

Eugenin, E. A., Clements, J. E., Zink, M. C., and Berman, J. W. (2011). Human immunodeficiency virus infection of human astrocytes disrupts blood-brain barrier integrity by a gap junction-dependent mechanism. J. Neurosci. Official J. Soc. Neurosci. 31 (26), 9456–9465. doi:10.1523/JNEUROSCI.1460-11.2011

PubMed Abstract | CrossRef Full Text | Google Scholar

Feldman, C., and Anderson, R. (2013). Cigarette smoking and mechanisms of susceptibility to infections of the respiratory tract and other organ systems. J. Infect. 67 (3), 169–184. doi:10.1016/j.jinf.2013.05.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Freeman, T. L., and Swartz, T. H. (2020). Purinergic receptors: elucidating the role of these immune mediators in HIV-1 fusion. Viruses 12 (3), 290. Article 3. doi:10.3390/v12030290

PubMed Abstract | CrossRef Full Text | Google Scholar

Ghosh, D., Mishra, M. K., Das, S., Kaushik, D. K., and Basu, A. (2009). Tobacco carcinogen induces microglial activation and subsequent neuronal damage. J. Neurochem. 110 (3), 1070–1081. doi:10.1111/j.1471-4159.2009.06203.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Ghura, S., Gross, R., Jordan-Sciutto, K., Dubroff, J., Schnoll, R., Collman, R. G., et al. (2020). Bidirectional associations among nicotine and tobacco smoke, NeuroHIV, and antiretroviral therapy. J. Neuroimmune Pharmacol. 15 (4), 694–714. doi:10.1007/s11481-019-09897-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Global, H. I. V. (2024). AIDS statistics—fact sheet. Available at: https://www.unaids.org/en/resources/fact-sheet.

Google Scholar

Guaraldi, G., Orlando, G., Zona, S., Menozzi, M., Carli, F., Garlassi, E., et al. (2011). Premature age-related comorbidities among HIV-infected persons compared with the general population. Clin. Infect. Dis. An Official Publ. Infect. Dis. Soc. Am. 53 (11), 1120–1126. doi:10.1093/cid/cir627

PubMed Abstract | CrossRef Full Text | Google Scholar

Han, H., Yang, Z., Chang, S. L., and Li, M. D. (2018). Modulatory effects of nicotine on neuroHIV/neuroAIDS. J. Neuroimmune Pharmacol. 13 (4), 467–478. doi:10.1007/s11481-018-9806-5

PubMed Abstract | CrossRef Full Text | Google Scholar

He, X., Yang, W., Zeng, Z., Wei, Y., Gao, J., Zhang, B., et al. (2020). NLRP3-dependent pyroptosis is required for HIV-1 gp120-induced neuropathology. Cell. Mol. Immunol. 17 (3), 283–299. doi:10.1038/s41423-019-0260-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Heaton, R. K., Clifford, D. B., Franklin, D. R., Woods, S. P., Ake, C., Vaida, F., et al. (2010). HIV-associated neurocognitive disorders persist in the era of potent antiretroviral therapy: CHARTER Study. Neurology 75 (23), 2087–2096. doi:10.1212/WNL.0b013e318200d727

PubMed Abstract | CrossRef Full Text | Google Scholar

Heaton, R. K., Franklin, D. R., Ellis, R. J., McCutchan, J. A., Letendre, S. L., LeBlanc, S., et al. (2011). HIV-associated neurocognitive disorders before and during the era of combination antiretroviral therapy: differences in rates, nature, and predictors. J. NeuroVirology 17 (1), 3–16. doi:10.1007/s13365-010-0006-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Hossain, M., Mazzone, P., Tierney, W., and Cucullo, L. (2011). In vitro assessment of tobacco smoke toxicity at the BBB: do antioxidant supplements have a protective role? BMC Neurosci. 12, 92. doi:10.1186/1471-2202-12-92

PubMed Abstract | CrossRef Full Text | Google Scholar

Hutamekalin, P., Farkas, A. E., Orbók, A., Wilhelm, I., Nagyőszi, P., Veszelka, S., et al. (2008). Effect of nicotine and polyaromtic hydrocarbons on cerebral endothelial cells. Cell Biol. Int. 32 (2), 198–209. doi:10.1016/j.cellbi.2007.08.026

PubMed Abstract | CrossRef Full Text | Google Scholar

Jia, Y., Zhang, Q., Liu, Z., Pan, P., Jia, Y., Zhu, P., et al. (2022). The role of α5-nicotinic acetylcholine receptor/NLRP3 signaling pathway in lung adenocarcinoma cell proliferation and migration. Toxicology 469, 153120. doi:10.1016/j.tox.2022.153120

PubMed Abstract | CrossRef Full Text | Google Scholar

Kahn, J. O., and Walker, B. D. (1998). Acute human immunodeficiency virus type 1 infection. N. Engl. J. Med. 339 (1), 33–39. doi:10.1056/NEJM199807023390107

PubMed Abstract | CrossRef Full Text | Google Scholar

Kaplan-Lewis, E., Aberg, J. A., and Lee, M. (2017). Aging with HIV in the ART era. Seminars Diagnostic Pathology 34 (4), 384–397. doi:10.1053/j.semdp.2017.04.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Katuri, A., Bryant, J., Heredia, A., and Makar, T. K. (2019). Role of the inflammasomes in HIV-associated neuroinflammation and neurocognitive disorders. Exp. Mol. Pathology 108, 64–72. doi:10.1016/j.yexmp.2019.03.008

PubMed Abstract | CrossRef Full Text | Google Scholar

Kearns, A., Gordon, J., Burdo, T. H., and Qin, X. (2017). HIV-1-Associated atherosclerosis: unraveling the missing link. J. Am. Coll. Cardiol. 69 (25), 3084–3098. doi:10.1016/j.jacc.2017.05.012

PubMed Abstract | CrossRef Full Text | Google Scholar

Khanal, S., Schank, M., El Gazzar, M., Moorman, J. P., and Yao, Z. Q. (2021). HIV-1 latency and viral reservoirs: existing reversal approaches and potential technologies, targets, and pathways involved in HIV latency studies. Cells 10 (2), 475. Article 2. doi:10.3390/cells10020475

PubMed Abstract | CrossRef Full Text | Google Scholar

Ko, A., Kang, G., Hattler, J. B., Galadima, H. I., Zhang, J., Li, Q., et al. (2019). Macrophages but not astrocytes harbor HIV DNA in the brains of HIV-1-Infected aviremic individuals on suppressive antiretroviral therapy. J. Neuroimmune Pharmacol. Official J. Soc. NeuroImmune Pharmacol. 14 (1), 110–119. doi:10.1007/s11481-018-9809-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Kousik, S., Napier, T. C., and Carvey, P. (2012). The effects of psychostimulant drugs on blood brain barrier function and neuroinflammation. Front. Pharmacol. 3, 121. doi:10.3389/fphar.2012.00121

PubMed Abstract | CrossRef Full Text | Google Scholar

Kulpa, D. A., and Chomont, N. (2024). HIV persistence in the setting of antiretroviral therapy: when, where and how does HIV hide? J. Virus Erad. 1 (2), 59–66. doi:10.1016/s2055-6640(20)30490-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Kumar, S., Rao, P. S. S., Earla, R., and Kumar, A. (2015). Drug-drug interactions between anti-retroviral therapies and drugs of abuse in HIV systems. Expert Opin. Drug Metabolism and Toxicol. 11 (3), 343–355. doi:10.1517/17425255.2015.996546

PubMed Abstract | CrossRef Full Text | Google Scholar

Lee, E., von Stockenstrom, S., Morcilla, V., Odevall, L., Hiener, B., Shao, W., et al. (2020). Impact of antiretroviral therapy duration on HIV-1 infection of T cells within anatomic sites. J. Virology 94 (3), e01270–19. doi:10.1128/JVI.01270-19

PubMed Abstract | CrossRef Full Text | Google Scholar

Lee, Y. W., Eum, S. Y., Nath, A., and Toborek, M. (2004). Estrogen-mediated protection against HIV Tat protein-induced inflammatory pathways in human vascular endothelial cells. Cardiovasc. Res. 63 (1), 139–148. doi:10.1016/j.cardiores.2004.03.006

PubMed Abstract | CrossRef Full Text | Google Scholar

Leibrand, C. R., Paris, J. J., Ghandour, M. S., Knapp, P. E., Kim, W.-K., Hauser, K. F., et al. (2017). HIV-1 Tat disrupts blood-brain barrier integrity and increases phagocytic perivascular macrophages and microglia in the dorsal striatum of transgenic mice. Neurosci. Lett. 640, 136–143. doi:10.1016/j.neulet.2016.12.073

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, Q., and Barres, B. A. (2018). Microglia and macrophages in brain homeostasis and disease. Nat. Rev. Immunol. 18 (4), 225–242. Article 4. doi:10.1038/nri.2017.125

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, L., Yu, J., Li, L., Zhang, B., Liu, L., Wu, C.-H., et al. (2017). Alpha7 nicotinic acetylcholine receptor is required for amyloid pathology in brain endothelial cells induced by Glycoprotein 120, methamphetamine and nicotine. Sci. Rep. 7 (1), 40467. doi:10.1038/srep40467

PubMed Abstract | CrossRef Full Text | Google Scholar

Longino, A. A., Paul, R., Wang, Y., Lama, J. R., Brandes, P., Ruiz, E., et al. (2022). HIV disease dynamics and markers of inflammation and CNS injury during primary HIV infection and their relationship to cognitive performance. JAIDS J. Acquir. Immune Defic. Syndromes 89 (2), 183–190. doi:10.1097/QAI.0000000000002832

CrossRef Full Text | Google Scholar

Louboutin, J.-P., and Strayer, D. (2014). Role of oxidative stress in HIV-1-Associated neurocognitive disorder and protection by gene delivery of antioxidant enzymes. Antioxidants 3 (4), 770–797. Article 4. doi:10.3390/antiox3040770

PubMed Abstract | CrossRef Full Text | Google Scholar

Mamik, M. K., and Power, C. (2017). Inflammasomes in neurological diseases: emerging pathogenic and therapeutic concepts. Brain A J. Neurology 140 (9), 2273–2285. doi:10.1093/brain/awx133

PubMed Abstract | CrossRef Full Text | Google Scholar

Manda, V. K., Mittapalli, R. K., Bohn, K. A., Adkins, C. E., and Lockman, P. R. (2010). Nicotine and cotinine increases the brain penetration of saquinavir in rat. J. Neurochem. 115 (6), 1495–1507. doi:10.1111/j.1471-4159.2010.07054.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Mao, C., Li, D., Zhou, E., Zhang, J., Wang, C., and Xue, C. (2021). Nicotine exacerbates atherosclerosis through a macrophage-mediated endothelial injury pathway. Aging (Albany NY) 13 (5), 7627–7643. doi:10.18632/aging.202660

PubMed Abstract | CrossRef Full Text | Google Scholar

Mazzone, P., Tierney, W., Hossain, M., Puvenna, V., Janigro, D., and Cucullo, L. (2010). Pathophysiological impact of cigarette smoke exposure on the cerebrovascular system with a focus on the blood-brain barrier: expanding the awareness of smoking toxicity in an underappreciated area. Int. J. Environ. Res. Public Health 7 (12), 4111–4126. doi:10.3390/ijerph7124111

PubMed Abstract | CrossRef Full Text | Google Scholar

Mbonye, U., and Karn, J. (2017). The molecular basis for human immunodeficiency virus latency. Annu. Rev. Virology 4 (1), 261–285. doi:10.1146/annurev-virology-101416-041646

PubMed Abstract | CrossRef Full Text | Google Scholar

Min, A. K., Javidfar, B., Missall, R., Doanman, D., Durens, M., Vil, S. S., et al. (2023). HIV-1 infection of genetically engineered iPSC-derived central nervous system-engrafted microglia in a humanized mouse model. Neuroscience, 538461. doi:10.1101/2023.04.26.538461

CrossRef Full Text | Google Scholar

Mullis, C., and Swartz, T. H. (2020). NLRP3 inflammasome signaling as a link between HIV-1 infection and atherosclerotic cardiovascular disease. Front. Cardiovasc. Med. 7, 95. doi:10.3389/fcvm.2020.00095

PubMed Abstract | CrossRef Full Text | Google Scholar

Nizri, E., Irony-Tur-Sinai, M., Lory, O., Orr-Urtreger, A., Lavi, E., and Brenner, T. (2009). Activation of the cholinergic anti-inflammatory system by nicotine attenuates neuroinflammation via suppression of Th1 and Th17 responses. J. Immunol. Baltim. Md 183 (10), 6681–6688. doi:10.4049/jimmunol.0902212

PubMed Abstract | CrossRef Full Text | Google Scholar

Osborne, O., Peyravian, N., Nair, M., Daunert, S., and Toborek, M. (2020). The paradox of HIV blood–brain barrier penetrance and antiretroviral drug delivery deficiencies. Trends Neurosci. 43 (9), 695–708. doi:10.1016/j.tins.2020.06.007

PubMed Abstract | CrossRef Full Text | Google Scholar

Pal, D., Kwatra, D., Minocha, M., Paturi, D. K., Budda, B., and Mitra, A. K. (2011). Efflux transporters- and cytochrome P-450-mediated interactions between drugs of abuse and antiretrovirals. Life Sci. 88 (21–22), 959–971. doi:10.1016/j.lfs.2010.09.012

PubMed Abstract | CrossRef Full Text | Google Scholar

Piekna-Przybylska, D., Nagumotu, K., Reid, D. M., and Maggirwar, S. B. (2019). HIV-1 infection renders brain vascular pericytes susceptible to the extracellular glutamate. J. Neurovirology 25 (1), 114–126. doi:10.1007/s13365-018-0693-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Pimentel, E., Sivalingam, K., Doke, M., and Samikkannu, T. (2020). Effects of drugs of abuse on the blood-brain barrier: a brief overview. Front. Neurosci. 14, 513. doi:10.3389/fnins.2020.00513

PubMed Abstract | CrossRef Full Text | Google Scholar

Pla-Tenorio, J., Roig, A. M., García-Cesaní, P. A., Santiago, L. A., Sepulveda-Orengo, M. T., and Noel, R. J. (2023). Astrocytes: role in pathogenesis and effect of commonly misused drugs in the HIV infected brain. Curr. Res. Neurobiol. 5, 100108. doi:10.1016/j.crneur.2023.100108

PubMed Abstract | CrossRef Full Text | Google Scholar

Plaza-Jennings, A. L., Valada, A., O’Shea, C., Iskhakova, M., Hu, B., Javidfar, B., et al. (2022). HIV integration in the human brain is linked to microglial activation and 3D genome remodeling. Mol. Cell 82 (24), 4647–4663.e8. doi:10.1016/j.molcel.2022.11.016

PubMed Abstract | CrossRef Full Text | Google Scholar

Rao, P. S. S., Ande, A., Sinha, N., Kumar, A., and Kumar, S. (2016). Effects of cigarette smoke condensate on oxidative stress, apoptotic cell death, and HIV replication in human monocytic cells. PLOS ONE 11 (5), e0155791. doi:10.1371/journal.pone.0155791

PubMed Abstract | CrossRef Full Text | Google Scholar

Revathikumar, P., Bergqvist, F., Gopalakrishnan, S., Korotkova, M., Jakobsson, P.-J., Lampa, J., et al. (2016). Immunomodulatory effects of nicotine on interleukin 1β activated human astrocytes and the role of cyclooxygenase 2 in the underlying mechanism. J. Neuroinflammation 13, 256. doi:10.1186/s12974-016-0725-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Richter, K., and Grau, V. (2023). Signaling of nicotinic acetylcholine receptors in mononuclear phagocytes. Pharmacol. Res. 191, 106727. doi:10.1016/j.phrs.2023.106727

PubMed Abstract | CrossRef Full Text | Google Scholar

Rock, R. B., Gekker, G., Aravalli, R. N., Hu, S., Sheng, W. S., and Peterson, P. K. (2008). Potentiation of HIV-1 expression in microglial cells by nicotine: involvement of transforming growth factor-beta 1. J. Neuroimmune Pharmacol. Official J. Soc. NeuroImmune Pharmacol. 3 (3), 143–149. doi:10.1007/s11481-007-9098-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Royal, W., Can, A., Gould, T. D., Guo, M., Huse, J., Jackson, M., et al. (2018). Cigarette smoke and nicotine effects on brain proinflammatory responses and behavioral and motor function in HIV-1 transgenic rats. J. NeuroVirology 24 (2), 246–253. doi:10.1007/s13365-018-0623-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Sacktor, N., McDermott, M. P., Marder, K., Schifitto, G., Selnes, O. A., McArthur, J. C., et al. (2002). HIV-associated cognitive impairment before and after the advent of combination therapy. J. Neurovirology 8 (2), 136–142. doi:10.1080/13550280290049615

PubMed Abstract | CrossRef Full Text | Google Scholar

Saylor, D., Dickens, A. M., Sacktor, N., Haughey, N., Slusher, B., Pletnikov, M., et al. (2016). HIV-associated neurocognitive disorder—pathogenesis and prospects for treatment. Nat. Rev. Neurol. 12 (4), 309–248. doi:10.1038/nrneurol.2016.53

PubMed Abstract | CrossRef Full Text | Google Scholar

Schröder, A. R. W., Shinn, P., Chen, H., Berry, C., Ecker, J. R., and Bushman, F. (2002). HIV-1 integration in the human genome favors active genes and local hotspots. Cell 110 (4), 521–529. doi:10.1016/s0092-8674(02)00864-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Shah, A., Gangwani, M. R., Chaudhari, N. S., Glazyrin, A., Bhat, H. K., and Kumar, A. (2016). Neurotoxicity in the post-HAART era: caution for the antiretroviral therapeutics. Neurotox. Res. 30 (4), 677–697. doi:10.1007/s12640-016-9646-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Sieg, S. F., Shive, C. L., Panigrahi, S., and Freeman, M. L. (2021). Probing the interface of HIV and inflammaging. Curr. HIV/AIDS Rep. 18 (3), 198–210. doi:10.1007/s11904-021-00547-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Siliciano, J. D., Kajdas, J., Finzi, D., Quinn, T. C., Chadwick, K., Margolick, J. B., et al. (2003). Long-term follow-up studies confirm the stability of the latent reservoir for HIV-1 in resting CD4+ T cells. Nat. Med. 9 (6), 727–728. doi:10.1038/nm880

PubMed Abstract | CrossRef Full Text | Google Scholar

Sonti, S., Sharma, A. L., and Tyagi, M. (2021). HIV-1 persistence in the CNS: mechanisms of latency, pathogenesis and an update on eradication strategies. Virus Res. 303, 198523. doi:10.1016/j.virusres.2021.198523

PubMed Abstract | CrossRef Full Text | Google Scholar

Sorimachi, M., Nishimura, S., and Yamagami, K. (1994). Inability of Ca2+ influx through nicotinic ACh receptor channels to stimulate catecholamine secretion in bovine adrenal chromaffin cells: studies with fura-2 and SBFI microfluorometry. Jpn. J. Physiology 44 (4), 343–356. doi:10.2170/jjphysiol.44.343

PubMed Abstract | CrossRef Full Text | Google Scholar

Sreeram, S., Ye, F., Garcia-Mesa, Y., Nguyen, K., Sayed, A. E., Leskov, K., et al. (2022). The potential role of HIV-1 latency in promoting neuroinflammation and HIV-1-associated neurocognitive disorder. Trends Immunol. 43 (8), 630–639. doi:10.1016/j.it.2022.06.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Suzuki, T., Hide, I., Matsubara, A., Hama, C., Harada, K., Miyano, K., et al. (2006). Microglial alpha7 nicotinic acetylcholine receptors drive a phospholipase C/IP3 pathway and modulate the cell activation toward a neuroprotective role. J. Neurosci. Res. 83 (8), 1461–1470. doi:10.1002/jnr.20850

PubMed Abstract | CrossRef Full Text | Google Scholar

Swartz, T. H., Dubyak, G. R., and Chen, B. K. (2015). Purinergic receptors: key mediators of HIV-1 infection and inflammation. Front. Immunol. 6, 585. doi:10.3389/fimmu.2015.00585

PubMed Abstract | CrossRef Full Text | Google Scholar

Valcour, V., Chalermchai, T., Sailasuta, N., Marovich, M., Lerdlum, S., and Suttichom, D.on behalf of the RV254/SEARCH 010 Study Group (2012). Central nervous system viral invasion and inflammation during acute HIV infection. J. Infect. Dis. 206 (2), 275–282. doi:10.1093/infdis/jis326

PubMed Abstract | CrossRef Full Text | Google Scholar

Valdebenito, S., Castellano, P., Ajasin, D., and Eugenin, E. A. (2021). Astrocytes are HIV reservoirs in the brain: a cell type with poor HIV infectivity and replication but efficient cell-to-cell viral transfer. J. Neurochem. 158 (2), 429–443. doi:10.1111/jnc.15336

PubMed Abstract | CrossRef Full Text | Google Scholar

Valiathan, R., Miguez, M. J., Patel, B., Arheart, K. L., and Asthana, D. (2014). Tobacco smoking increases immune activation and impairs T-cell function in HIV infected patients on antiretrovirals: a cross-sectional pilot study. PLOS ONE 9 (5), e97698. doi:10.1371/journal.pone.0097698

PubMed Abstract | CrossRef Full Text | Google Scholar

Wallet, C., De Rovere, M., Van Assche, J., Daouad, F., De Wit, S., Gautier, V., et al. (2019). Microglial cells: the main HIV-1 reservoir in the brain. Front. Cell. Infect. Microbiol. 9, 362. doi:10.3389/fcimb.2019.00362

PubMed Abstract | CrossRef Full Text | Google Scholar

Walsh, J. G., Reinke, S. N., Mamik, M. K., McKenzie, B. A., Maingat, F., Branton, W. G., et al. (2014). Rapid inflammasome activation in microglia contributes to brain disease in HIV/AIDS. Retrovirology 11 (1), 35. doi:10.1186/1742-4690-11-35

PubMed Abstract | CrossRef Full Text | Google Scholar

White, J. A., Kufera, J. T., Bachmann, N., Dai, W., Simonetti, F. R., Armstrong, C., et al. (2022). Measuring the latent reservoir for HIV-1: quantification bias in near full-length genome sequencing methods. PLoS Pathog. 18 (9), e1010845. doi:10.1371/journal.ppat.1010845

PubMed Abstract | CrossRef Full Text | Google Scholar

Wojna, V., Robles, L., Skolasky, R. L., Mayo, R., Selnes, O., de la Torre, T., et al. (2007). Associations of cigarette smoking with viral immune and cognitive function in human immunodeficiency virus-seropositive women. J. Neurovirology 13 (6), 561–568. doi:10.1080/13550280701620747

PubMed Abstract | CrossRef Full Text | Google Scholar

Wu, X., Zhang, H., Qi, W., Zhang, Y., Li, J., Li, Z., et al. (2018). Nicotine promotes atherosclerosis via ROS-NLRP3-mediated endothelial cell pyroptosis. Cell Death and Dis. 9 (2), 171. Article 2. doi:10.1038/s41419-017-0257-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Xu, S., Chen, H., Ni, H., and Dai, Q. (2021). Targeting HDAC6 attenuates nicotine-induced macrophage pyroptosis via NF-κB/NLRP3 pathway. Atherosclerosis 317, 1–9. doi:10.1016/j.atherosclerosis.2020.11.021

PubMed Abstract | CrossRef Full Text | Google Scholar

Yang, Z., Nesil, T., Connaghan, K. P., Li, M. D., and Chang, S. L. (2016). Modulation effect of HIV-1 viral proteins and nicotine on expression of the immune-related genes in brain of the HIV-1 transgenic rats. J. Neuroimmune Pharmacol. Official J. Soc. NeuroImmune Pharmacol. 11 (3), 562–571. doi:10.1007/s11481-016-9679-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Yuan, N. Y., and Kaul, M. (2021). Beneficial and adverse effects of cART affect neurocognitive function in HIV-1 infection: balancing viral suppression against neuronal stress and injury. J. Neuroimmune Pharmacol. 16 (1), 90–112. doi:10.1007/s11481-019-09868-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, Y., Chen, Y., Zhang, Y., Li, P.-L., and Li, X. (2019). Contribution of cathepsin B-dependent Nlrp3 inflammasome activation to nicotine-induced endothelial barrier dysfunction. Eur. J. Pharmacol. 865, 172795. doi:10.1016/j.ejphar.2019.172795

PubMed Abstract | CrossRef Full Text | Google Scholar

Zia, S., Ndoye, A., Lee, T. X., Webber, R. J., and Grando, S. A. (2000). Receptor-mediated inhibition of keratinocyte migration by nicotine involves modulations of calcium influx and intracellular concentration. J. Pharmacol. Exp. Ther. 293 (3), 973–981. Available at: https://jpet.aspetjournals.org/content/293/3/973.

PubMed Abstract | Google Scholar

Zoli, M., Pucci, S., Vilella, A., and Gotti, C. (2018). Neuronal and extraneuronal nicotinic acetylcholine receptors. Curr. Neuropharmacol. 16 (4), 338–349. doi:10.2174/1570159X15666170912110450

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: HIV-1, NLRP3 inflammasome, neuroinflammation, nicotine, blood-brain barrier (BBB)

Citation: Keane AM and Swartz TH (2024) The impacts of tobacco and nicotine on HIV-1 infection, inflammation, and the blood-brain barrier in the central nervous system. Front. Pharmacol. 15:1477845. doi: 10.3389/fphar.2024.1477845

Received: 08 August 2024; Accepted: 17 October 2024;
Published: 28 October 2024.

Edited by:

Massimo Grilli, University of Genoa, Italy

Reviewed by:

David Tattersall, GlaxoSmithKline, United States
Debashis Dutta, University of Nebraska Medical Center, United States
Joseph Bryant, University of Maryland, United States

Copyright © 2024 Keane and Swartz. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Talia H. Swartz, dGFsaWEuc3dhcnR6QG1zc20uZWR1

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.