Next Article in Journal
Mental Pain Correlates with Mind Wandering, Self-Reflection, and Insight in Individuals with Psychotic Disorders: A Cross-Sectional Study
Previous Article in Journal
Poor Oral Health Linked with Higher Risk of Alzheimer’s Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Nicotine, THC, and Dolutegravir Modulate E-Cigarette-Induced Changes in Addiction- and Inflammation-Associated Genes in Rat Brains and Astrocytes

by
Jacqueline Renee Kulbe
1,†,
Lauren Nguyen
1,†,
Alexandra Anh Le
1,
Anna Elizabeth Laird
1,
Michael A. Taffe
1,
Jacques D. Nguyen
2,‡ and
Jerel Adam Fields
1,*,‡
1
Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA
2
Department of Psychology and Neuroscience, Baylor University, Waco, TX 76706, USA
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
These authors contributed equally to this work.
Brain Sci. 2023, 13(11), 1556; https://doi.org/10.3390/brainsci13111556
Submission received: 11 October 2023 / Revised: 30 October 2023 / Accepted: 3 November 2023 / Published: 7 November 2023
(This article belongs to the Special Issue Addiction and Neuroinflammation)

Abstract

:
E-cigarette use has been marketed as a safer alternative to traditional cigarettes, as a means of smoking cessation, and are used at a higher rate than the general population in people with HIV (PWH). Early growth receptor 2 (EGR2) and Activity-Regulated Cytoskeleton-Associated Protein (ARC) have a role in addiction, synaptic plasticity, inflammation, and neurodegeneration. This study showed that 10 days of exposure to e-cigarette vapor altered gene expression in the brains of 6-month-old, male, Sprague Dawley rats. Specifically, the e-cigarette solvent vapor propylene glycol (PG) downregulated EGR2 and ARC mRNA expression in frontal cortex, an effect which was reversed by nicotine (NIC) and THC, suggesting that PG could have a protective role against NIC and cannabis dependence. However, in vitro, PG upregulated EGR2 and ARC mRNA expression at 18 h in cultured C6 rat astrocytes suggesting that PG may have neuroinflammatory effects. PG-induced upregulation of EGR2 and ARC mRNA was reversed by NIC but not THC. The HIV antiretroviral DTG reversed the effect NIC had on decreasing PG-induced upregulation of EGR2, which is concerning because EGR2 has been implicated in HIV latency reversal, T-cell apoptosis, and neuroinflammation, a process that underlies the development of HIV-associated neurocognitive disorders.

1. Introduction

For decades, cigarette smoking has represented a significant health crisis worldwide. Concerted public health efforts in the United States have led to a decline in tobacco use, particularly among teenagers and young adults [1]. However, rates of e-cigarette use have increased since their inception and are popular among adolescents and young adults [2]. E-cigarettes have also been marketed as a safer alternative to traditional cigarettes and as a tool for smoking cessation [3]. E-cigarettes contain a liquid composed of a psychoactive substance (ex: nicotine [NIC] and tetrahydrocannabinol [THC]) and a solvent (ex: propylene glycol) that is vaporized by a heating element and then inhaled by the user. Although the dangers of tobacco and NIC have been well studied, the effect that chronic inhalation of solvent vapors, such as propylene glycol, in combination with psychoactive substances, have on addiction, inflammation, and neurobiology is not well understood.
People with HIV (PWH) use e-cigarettes and other addictive substances at higher rates than the general population and may be more vulnerable to the development of substance use disorders [4,5]. PWH are particularly vulnerable to smoking and psychoactive substance-associated illnesses such as COPD, malignancy, cardiovascular disease, and immune dysfunction even when receiving antiretroviral therapy (ART) [6,7]. PWH on ART are also at risk of developing HIV-associated neurocognitive disorders (HAND), which is in part caused by excessive neuroinflammation [8]. Understanding the effects of e-cigarettes on addiction and neuroinflammatory pathways is important to the general population of e-cigarette users and is of special relevance to people with PWH whose brains have higher baseline levels of neuroinflammation and may be more vulnerable to insult.
Recent studies suggest that e-cigarettes may affect the brain. E-cigarette aerosols with NIC affect neuron and astrocyte function and neurotransmitter processing in mice [9,10]. Another study showed that e-cigarettes impair short- and long-term memory and cause reductions in brain-derived neurotrophic factors in rats [11]. NIC alone binds the nicotinic acetylcholine receptor and acute NIC use has been associated with improved hippocampus-dependent learning, memory, and attention. Contrary to this, chronic NIC use was associated with depressed hippocampus-dependent learning [12]. However, few studies have delineated how NIC, THC, or ART modify e-cigarette-induced changes in gene expression in the frontal cortex.
In recent years, RNA sequencing (RNAseq) and subsequent analyses of the transcriptome have empowered researchers to gain insight into how global gene expression is altered in response to a given stimulus or set of stimuli. Pairing transcriptomics unbiased search for changes in gene expression with traditional approaches such as real-time polymerase chain reaction (RT2PCR) and Western blotting is proving to be an efficient way to discover novel gene expression networks and hypotheses.
The goal of this study was to utilize RNAseq technology and transcriptome analyses to investigate the effects of propylene glycol (PG), nicotine (NIC), and THC on gene expression in the brain in an unbiased way. We next selected two important genes (EGR2 and ARC) deemed from the literature to be relevant to PWH and addiction from the transcriptomics analyses for downstream validation mRNA using RT2PCR and Western blot, respectively. Lastly, we used qRT2PCR to investigate the expression of these genes in cultured astrocytes exposed to PG, NIC, and THC. The effects of anti-retroviral dolutegravir (DTG) in combination with PG, NIC, and THC on astrocytes were also assessed due to its relevance to PWH.

2. Methods

2.1. Animal Model

Male Sprague Dawley rats (Harlan/Envigo, Livermore, CA, USA) were housed in humidity- and temperature-controlled (23  ±  2 °C) environments on reversed 12:12 h light: dark cycles. All procedures were conducted under protocols approved by the Institutional Care and Use of the University of California, San Diego (Institutional Animal Care and Use Committee protocol: S19029).

2.2. Rat Exposure to E-Cigarette Vapor Delivery Apparatus

First, 6-month old, male, Sprague Dawley rats (n = 6/group) were placed into vaporization chambers for 30 min sessions twice per day for 10 days and exposed to room air, propylene glycol (PG, 100%), PG + nicotine (NIC 30 mg/mL), or PG + THC (200 mg/mL). Two 30 min sessions were chosen in an attempt to model the multiple per day e-cigarette use seen per day in humans. Nicotine dependence has been shown to develop in as little as 7 to 14 days [13,14]. Vapor was delivered through a vapor inhalation system designed to deliver psychoactive substance vapor as previously described [15,16,17,18,19]. Nicotine and propylene glycol were obtained from Sigma-Aldrich (St. Louis, MO, USA) and THC was provided by the U.S. National Institute on Drug Abuse.

2.3. RNA Isolation, Library Prep and Sequencing

RNA (n = 6/group) was extracted from frontal cortex Qiagen RNeasy Lipid Tissue Kit per manufacturer’s instructions (Qiagen, Germantown, MD, USA; cat no. 74804). The mRNA library was generated using Illumina® Stranded mRNA Prep kit (San Diego, CA, USA, cat no. 20040532) and sequenced on a NovaSeq 6000 Sequencing System. Analysis of differential gene expression was performed by processing FASTQ files with DRAGEN RNA app and aligning to the Rattus norvegicus genome using the DRAGEN Differential Expression app on Illumina’s BaseSpace cloud software (v 4.2.4).

2.4. In-Vitro Studies of Astrocytes

The C6 rat glioma cell line (ATCC cat# CCL-107) was used here for their astroglia characteristics. Astroglia are implicated in frontal cortex function and relevant to addiction and HIV-associated neurocognitive disorders. C6 cultures were grown in DMEM with 5% FBS at 37 °F and 5% CO2. C6 rat astrocytes were split into 12 well plates at 5 × 105 cells/well. Cells were treated with various combinations of vehicle (DMEM with 5% FBS), propylene glycol (50 mM, 100 mM, or 150 mM, n = 3/group/dose response), Nicotine (10 μM), THC (10 μM), or Dolutegravir (DTG) (200 nM) for 6 h or 18 h and then processed for RNA (n = 9–15/group).

2.5. Real-Time Reverse Transcription Polymerase Chain Reaction

6 h or 18 h following treatments C6 rat astrocytes were washed with PBS and RNA was extracted and reverse transcribed as previously described [20]. Gene expression assays were performed and analyzed as previously described [21] using primers specific to EGR2 (Taqman, ThermoFisher Scientific, Carlsbad, CA, USA; cat no. Rn00571208_g1), ARC (Taqman, Rn00571208_g1), and ActB (Taqman, Rn00667869_m1).

2.6. Immunoblot of Human Brain Specimens

Frontal cortex tissues (n = 6/group) from rat brains were homogenized and Western blot and analysis per performed as previously described [20,21,22]. Antibodies were used as follows: ARC (ARC/ARG3 polyclonal; Proteintech, Chicago, IL, USA; cat no. 16290-1-AP; 1:500), EGR2 (EGR2 polyclonal; Proteintech; cat no. 13491-1-AP; 1:500), β-actin (ACTB; Sigma-Aldrich, cat. no. A5441; 1:2000), species-specific IgG conjugated to HRP (American Qualex, San Clemente, CA, USA; cat. no. A102P5; 1:5000).

2.7. Statistical Analysis

Statistical analysis was determined by one-way ANOVA with Tukey’s post hoc test using GraphPad Prism (v 10.0.2) with a p < 0.5 considered significant.

3. Results

3.1. Differential Gene Expression (Figure 1)

Following exposure to Air, PG, PG + NIC, or PG + THC (n = 6/group), RNA was extracted from whole brain lysate, sequenced, and differential gene expression was analyzed as above. Fourteen genes were significantly (p < 0.05) differentially expressed between PG and Air groups, including Egr1, ARC, Rsrp1, Junb, Emcn, Miiip, Trib1, Hspa1b, Egr2, Abca7, Slc39a&, Egr4, Fos, and Dusp6. Between the PG and NIC groups, 13 genes were significantly (p < 0.05) differentially expressed including Gemin6, Hspa1b, Zfp180, Rsrp1, Junb, Per1, Egr4, Nr4a3, Ift122, Errfil1, Egr2, Cttn, and AABR07065531.5. Between NIC and THC three genes were significantly (p < 0.05) differentially expressed, including Il1r1, Tgm2, and Ebna1bp2.
Analysis identified several Early Growth Response Factor genes that were differentially expressed between PG versus Air and PG versus NIC, including EGR2 (Early Growth Receptor 2 gene) which was downregulated in PG compared to both Air and NIC. The intermediate-early gene ARC (Activity Regulated Cytoskeleton-Associated gene) showed high levels of expression in both PG and AIR and was also downregulated in PG.
Figure 1. E-cigarette vapor, nicotine, and THC alter gene expression in rat frontal cortex. Heatmaps demonstrating significant differential gene expression (p < 0.05) for mRNA extracted and sequenced from whole brain lysate of rats exposed to e-cigarette vapor for 10 days. (a) PG (100% propylene glycol) vs. AIR (air), (b) PG vs. NIC (PG + Nicotine 30 mg/mL), (c) NIC vs. THC (PG + THC 200 mg/mL).
Figure 1. E-cigarette vapor, nicotine, and THC alter gene expression in rat frontal cortex. Heatmaps demonstrating significant differential gene expression (p < 0.05) for mRNA extracted and sequenced from whole brain lysate of rats exposed to e-cigarette vapor for 10 days. (a) PG (100% propylene glycol) vs. AIR (air), (b) PG vs. NIC (PG + Nicotine 30 mg/mL), (c) NIC vs. THC (PG + THC 200 mg/mL).
Brainsci 13 01556 g001

3.2. Frontal Cortex mRNA and Protein Expression of EGR2 and ARC (Figure 2)

Quantitative RT-PCR was used to determine the fold changes for EGR2 and ARC mRNA in frontal cortex of rats exposed to air (control), PG (vehicle), PG + THC, and PG + NIC (n = 5–6/group). For EGR2, one-way ANOVA revealed a significant decrease in PG compared to AIR (p = 0.01), THC (p = 0.0359), and NIC (p = 0.0007) (Figure 2a). However, EGR2 mRNA levels do not significantly differ between AIR, THC, and NIC, suggesting that THC and NIC negate the effect PG has on the downregulation of EGR2. These data are consistent with the differential expression results obtained from whole brain lysates which showed that EGR2 was downregulated in PG compared to AIR and NIC (Figure 1a,b) and consistent with Figure 1c which shows that EGR2 was not identified as being differentially expressed between NIC and THC. Western blot did not identify significant differences in EGR2 protein expression in the frontal cortex of rats exposed to AIR, PG, THC, or NIC (Figure 2d). However, the PG group had the lowest mean EGR2 protein expression (approximately 10% decrease compared to AIR). These data suggest that while a 10-day exposure to these substances is sufficient to induce mRNA, it may not be a long enough period of time to alter protein expression significantly.
ARC showed a similar pattern in frontal cortex as EGR2 with PG having the lowest mean fold change (approximately 10% decrease compared to AIR) and THC and NIC groups being more similar to air suggesting PG downregulates ARC mRNA and this downregulation is reversed by THC and NIC. However, these results were not statistically significant (Figure 2b). Similarly, significant changes were not detected in ARC protein expression; however, the PG group had the lowest mean level of protein expression amongst all groups (Figure 2e).
Figure 2. Propylene glycol (PG) decreases EGR2 and ARC mRNA and protein expression in frontal cortex of rats exposed to e-cigarette vapor for 10 days and these changes are reversed by THC and nicotine. (a) Fold change in EGR2 and (b) ARC mRNA normalized to ACTB. (c) Immunoblot of frontal cortex with an antibody specific for EGR2, ARC, and Actin. Quantification of immunoblot for (d) EGR2 and (e) ARC band intensity normalized to actin. AIR = Air, PG = 100% propylene glycol, THC = PG + THC 200 mg/mL. NIC = PG + nicotine 30 mg/mL. Mean ± SEM. Statistical significance was determined by one-way ANOVA, post hoc Tukey’s. ** p < 0.01 vs. Air; ^ p < 0.05, ^^^ p < 0.001 vs. PG.
Figure 2. Propylene glycol (PG) decreases EGR2 and ARC mRNA and protein expression in frontal cortex of rats exposed to e-cigarette vapor for 10 days and these changes are reversed by THC and nicotine. (a) Fold change in EGR2 and (b) ARC mRNA normalized to ACTB. (c) Immunoblot of frontal cortex with an antibody specific for EGR2, ARC, and Actin. Quantification of immunoblot for (d) EGR2 and (e) ARC band intensity normalized to actin. AIR = Air, PG = 100% propylene glycol, THC = PG + THC 200 mg/mL. NIC = PG + nicotine 30 mg/mL. Mean ± SEM. Statistical significance was determined by one-way ANOVA, post hoc Tukey’s. ** p < 0.01 vs. Air; ^ p < 0.05, ^^^ p < 0.001 vs. PG.
Brainsci 13 01556 g002

3.3. Time Course and Dose–Response of PG-Induced mRNA Expression in Rat Astrocytes

A dose–response (n = 3/group) of PG (5 mM, 50 mM, 150 mM) was conducted in vitro to evaluate changes in mRNA expression for EGR2 and ARC in cultured C6 rat astrocytes. At 18 h, 150 mM of PG significantly increased EGR2 mRNA (p = 0.0187) expression by 20% compared to control (Figure 3a) and significantly increased ARC mRNA expression compared to control (p = 0.0024) and 5 mM (p = 0.0080) and 50 mM doses (p = 0.0055) (Figure 3b). 150 mM PG did not result in increases in cytotoxicity. Therefore, 150 mM PG was used for further in vitro experiments. Next, ERG2 and ARC mRNA expression were evaluated in C6 rat astrocytes that had been treated with 150 mM PG for either 6 h or 18 h (n = 6–8/group). Similar to in vivo data, at 6 h EGR2 mRNA was significantly decreased compared to vehicle (Figure 3c) but then showed a significant increase at 18 h when compared to vehicle or 6 h. For ARC, mRNA was significantly increased at both 6 h and 18 h compared to vehicle with the 6 h timepoint also being significantly elevated compared to 18 h (Figure 3d).

3.4. PG, NIC, THC, and DTG-Induced EGR2 and ARC mRNA Expression in Rat Astrocytes

C6 Rat astrocytes were treated with PG (150 m), NIC (10 μM), THC (10 μM), or dolutegravir (200 ng/mL) for 18 h in the following combinations (n = 9–15/group): Vehicle (media), PG, PG + NIC, PG + THC, DTG, PG + DTG, PG + THC + DTG, PG + NIC + DTG. For EGR2 (Figure 4a), PG significantly increased (p = 0.0005) mRNA expression by 55% compared to control at 18 h, an effect that was reversed by the addition of nicotine (p = 0.2546; vehicle vs. PG + NIC). The addition of THC further elevated EGR2 mRNA by 20% compared to PG alone and was significantly increased compared to control (p = 0.016). DTG alone had minimal effect on EGR2 mRNA expression compared to vehicle (p = 0.9927). PG + DTG was significantly elevated compared to DTG (p = 0.0107) indicating that PG-induced increases in EGR2 mRNA expression occur in the presence of DTG. However, EGR2 mRNA expression was 13% but non-significantly increased (p = 0.8465) in PG + NIC + DTG compared to PG + NIC group, suggesting the addition of DTG may interfere with the ability of NIC to attenuate PG-induced increases in EGR2 mRNA expression. Although non-significant, a similar pattern was seen for ARC mRNA.

4. Discussion

Our results indicate that ten days (2 × 30 min per day) of exposure to PG ± NIC or THC is sufficient to alter gene expression in the brains of 6-month-old, male, Sprague Dawley rats. RNA sequencing revealed several differentially expressed genes between treatment groups including several early growth response genes (EGR1, EGR2, EGR4), with EGR2 being downregulated in PG-exposed animals compared to AIR or PG + Nicotine (Figure 1a,b) and ARC being downregulated in PG compared to AIR (Figure 1a). Similarly, EGR2 mRNA was significantly decreased, and ARC mRNA was non-significantly decreased in PG-exposed frontal cortex (Figure 2a,b). These effects were reversed by the addition of NIC or THC (Figure 2a,b). Typically, it is unwise to draw conclusions from non-significant data. However, because the ARC mRNA levels show a similar overall trend to that of ERG2 we believe that these trends deserve discussion. Therefore, the hypothesis and implication regarding both EGR2 and ARC changes are discussed below.
PG is a food additive generally considered safe by the FDA. PG vapor has been found to contain dangerous chemicals such as formaldehyde and acrolein but animal studies evaluating the effects of PG vapor generally indicate it to be non-toxic [23]. However, the majority of these studies focus on the respiratory tract and other peripheral organ systems. Our results indicated that PG vapor is sufficient to induce changes in gene expression within the brains of rats after only 10 days of exposure. Alterations in gene expression for our validated genes, EGR2 and ARC, did not translate to changes in protein expression (Figure 2c,d) as assessed by Western blot. Although it is possible that the 10-day exposure was not long enough for alternations in mRNA expression to translate to alterations in protein expression, given the previously rapid induction of ARC and EGR2 seen in models of psychoactive substance administration [24,25], it is also possible that alterations in post-translational modification are occurring. Post-translational modifications are known to occur and regulate both EGR2 and ARC function [26]. Future studies focusing on evaluation of post-translational modifications as well as upstream modulators are needed.
EGR2 and ARC were chosen for further evaluation because they both have a role in addiction, inflammation, and neurodegeneration. EGR2 is a transcription factor characterized as an immediate-early gene and its transcription is rapidly induced by a variety of extracellular stimuli [27]. ARC is also an immediate-early gene and encodes for activity-regulated cytoskeletal-associated protein. Both are important in synaptic plasticity [28,29,30,31] which is an underlying mechanism for both addiction [32] and the memory and cognitive impairment seen in neurodegenerative disorders such as Alzheimer’s Disease and HAND [33].
Alterations in EGR2 and ARC are found in addiction models [25,31,34,35,36]. Changes in ARC are induced rapidly, in as little as 30 min [24], following psychoactive substance administration, with increases sustained chronically [31,37], and decreases induced by psychoactive substance withdrawal [31,37]. EGR-2 is also induced within hours of psychoactive substance administration, including nicotine administration [25], with increases sustained chronically depending on brain region and treatment paradigm [38,39]. EGR2 and ARC are induced or upregulated by psychoactive substances including cocaine, methamphetamine, alcohol, heroin, and nicotine [25,31,35]. CBD also induces EGR2 expression [40]. Therefore, it is unsurprising that in the brains of rats exposed to PG, nicotine was able to reverse PG-induced decreases in EGR2 and ARC mRNA expression (Figure 2a,b). Nor is it surprising that THC also reversed PG-induced decreases in EGR2 expression. However, the ability of THC to reverse PG-induced decreases in ARC mRNA expression is in contrast to a previous study in which THC decreased ARC protein expression [41]. However, that study was conducted in adolescent rats, THC was administered systemically without PG exposure, and significant differences were only found in the prefrontal cortex of female rats [41].
It is unclear how inhalation of PG vapor causes decreases in EGR2 and ARC mRNA expression and why the effect is more robust in EGR2. It is possible that the modulation of EGR2 and ARC are not induced directly by PG, NIC, or THC themselves, but that these substances are acting through upstream transcription factors or kinases. EGR2 itself is a transcription factor capable of binding ARC [42], which may explain the more robust changes seen in EGR2 compared to ARC. ARC is regulated by a variety of upstream factors including cAMP response element-binding protein (CREB), Myocyte Enhancer Factor 2 (MEF2), and Serum response factor (SRF) [43], as well as the kinase Extracellular Signal-regulated Kinases (ERK) [44]. Both ARC and EGR2 have also been associated with Nrf2 (Nuclear factor erythroid-2 related factor 2), a transcription factor involved in the regulation of antioxidant and inflammatory responses. Specifically, knockdown of ARC has been demonstrated to inhibit Nrf2 expression [45]. And is increased in parallel to Nrf2 in transgenic animals less prone to oxidative stress [46], whereas parallel increases in EGR2 and Nrf2 are associated with improved retention memory [47]. Interestingly, CBD has been shown to modulate Nrf2 and its upstream regulator (Nuclear factor kappa-light-chain-enhancer of activated B cells) NFkB [48], which also regulates EGR2. Further studies are needed to evaluate the role of upstream transcription factors in the modulation of EGR2 and ARC by PG, NIC, or THC. It is also unclear what the neurobiological implications of these findings might be. It is possible that PG-induced decreases in EGR2 could have a protective effect against nicotine- or THC-induced increases in EGR2 expression, lowering the overall risk of nicotine or cannabis dependence. However, that conclusion cannot be made based on the current work because behavioral studies were not conducted. However, in support of this hypothesis, a previous study demonstrated that the addition of PG decreased the intracranial self-administration threshold of high-dose nicotine in rats [49]. On the other hand, as discussed further below, because decreases in ARC correlate with both neuroinflammation and cognitive impairment [33], it is possible that PG-induced decreases in ARC could enhance neuroinflammation and cognitive impairment in e-cigarette users who are more vulnerable to those pathologies such as those with HIV.
The importance of EGR2 and ARC is not limited to addiction. Immunologically, EGR2 is involved in B- and T-cell development [50], macrophage polarization [51], and T-cell suppression [52]. ARC is involved in peripheral immune cell migration, T-cell activation [53], and upregulated following prenatal inflammation [54]. In specific regard to HIV, upregulation of early growth response proteins, including EGR2, can lead to HIV reactivation and latency reversal [55] and binding of the HIV tat protein to EGR2 can enhance T-cell apoptosis [56]. Neurologically, in addition to their roles in synaptic plasticity, EGR2 is involved in hindbrain development, peripheral nerve myelination, heritable peripheral neuropathies, and Huntington’s Disease [57,58,59]. Alterations in ARC expression and post-translational modifications of ARC occur both in genetic neurodevelopmental disorders and Alzheimer’s Disease [30]. In fact, ARC enhances the cleavage of amyloid precursor protein (APP) to Aβ peptide [30] which is known to accumulate in both Alzheimer’s and HAND. Less is known about the role EGR2 and ARC have in neuroinflammation. Although EGR2 is upregulated in Aβ plaque-associated microglia [60], neuroinflammatory stimuli lead to upregulation of EGR2 in neurons [61], microglia, and astrocytes [62], and neuroinflammation alters activity-dependent ARC synthesis in hippocampal neurons [33].
Similar to the in vivo findings (Figure 2), PG downregulated EGR2 mRNA expression in cultured C6 rat astrocytes at 6 h but upregulated EGR2 at 18 h (Figure 3c). ARC was significantly upregulated at both 6 h and 18 h (Figure 3d). The majority of ARC studies focus on neuronal expression of ARC, making the finding that PG is able to upregulate ARC mRNA in C6 rat astrocytes quite novel.
It is unclear why ERG2 was initially downregulated in C6 rat astrocytes, however, the discrepancy between the in vivo PG-induced downregulation of EGR2 and ARC and the in vitro upregulation of EGR2 and ARC is possibly due to a cell-specific effect and the in vivo environment contains many cell-types interacting together to maintain homeostasis. Alternatively, although the concentration of PG was chosen to ensure a robust in vitro response in order to study the effects of the addition of NIC, THC, and DTG, we concede that a 50–150 mM concentration of PG is unlikely comparable to physiologic levels following e-cigarette exposure which are likely not to be more than in the micromolar range [63]. Therefore, modulation of EGR2 and ARC in cultured rat astrocytes at 6 h and 18 h may be the result of a stress response to a non-physiologic chemical or osmotic insult rather than a PG-specific effect. Reassuringly, these concentrations of PG did not induce cytotoxicity. Both EGR2 and ARC are known to be upregulated under stressful conditions or alterations in metabolic activity [64]. Although both EGR2 and ARC are induced rapidly following stimulus [24,25,62], we suspect that the different pattern seen between EGR2 and ARC at 6 h and 18 h is likely due to temporal differences in expression induction between EGR2 and ARC, with decreases in ARC at 18 h possibly being the result of other homeostatic mechanisms. Future studies are needed to evaluate more physiologically relevant concentrations of PG, and earlier and later time points. However, we find the ability of THC to further enhance this effect and NIC to reduce EGR2 to levels similar to control particularly intriguing. Additionally, future in vivo studies evaluating the effects of e-cigarettes on individual cell types, including neuroimmune cells such as astrocytes and microglia are needed.
Similar to the in vivo studies (Figure 2), NIC reversed PG-induced alterations in EGR2 gene expression. In this case, reversing PG-induced upregulation of EGR2 (Figure 4a). ARC expression changes were subtle and non-significant but followed a similar overall pattern to that of EGR2 (Figure 4). PG vapor has been previously shown to increase pro-inflammatory cytokine expression [65]. It is possible that in this model, PG is acting as a pro-inflammatory stimulus, leading to upregulation of EGR2, and that nicotine is having an anti-inflammatory effect [66]. To test this hypothesis, future studies evaluating cytokine production and astrocyte morphology in vivo and in vitro will be needed.
The ability of PG to upregulate EGR2 in cultured astrocytes, possibly due to an underlying neuroinflammatory mechanism, is particularly concerning to THC e-cigarette users because THC did not reverse PG-induced upregulation of EGR2 mRNA in cultured C6 rat astrocytes, but instead furthered increased EGR2 mRNA levels compared to PG alone (Figure 4a). However, it should be noted that variability was high in this group. It is unclear why nicotine but not THC was able to reduce PG-induced upregulation of EGR2 mRNA given the current interest in the anti-inflammatory properties of cannabis [67]. However, the anti-inflammatory properties of cannabis are most likely due to non-THC cannabinoids [68]. In regard to PWH, the ability of PG to upregulate EGR2 in cultured astrocytes is alarming due to the possibility that EGR2 is involved in HIV latency reversal and CD4 apoptosis [55,56] and that chronic neuroinflammation contributes to the development of HAND [8]. However, the differences in the effect of PG on EGR2 expression in rat brains (decreased expression) versus in C6 rat astrocytes (decreased at six hours and increased at 18 h) may indicate a differential of PG on astrocytes in the in vivo environment. Bystander brain cell types (neurons, microglia, oligodendrocytes) may harbor the observed changes in EGR2 expression or may modulate the effects of PG on astrocytes in vivo. Moreover, the differences in the in vivo versus in vitro studies may be driven by acute versus chronic exposure to PG, NIC, and THC. Further, Dolutegravir, an HIV integrase inhibitor found in the common HIV antiretroviral medication Tivicay, reversed the effect NIC had on decreasing PG-induced upregulation of EGR2 (Figure 4a), suggesting that for PWH, DTG has the ability to further modulate e-cigarette-induced alterations in gene expression in ways that we do not yet understand. Future studies focusing on the interaction between e-cigarettes and antiretrovirals in models of HIV are needed.
Limitations of the study: This study did not investigate the combinatorial effect of DTG with Nic or THC in the absence of PG, which is relevant to PWH smoking cannabis in traditional combustion of marijuana cigarettes or pipes. While this is important for understanding the effects of NIC and THC on the brain in PWH and PWoH, the studies here focused on e-cigs which generally use a vehicle for NIC or THC, in this case PG. This study does not fully address the differences in gene expression in response to PG, NIC, and THC in rat brains versus C6 rat astrocytes. Additionally, although nicotine dependence in rats had been shown to develop in as little as 7 days during nicotine vaporization, and one rat day is equivalent to approximately 30 human days, now that this study has demonstrated acute gene changes following 10 days of e-cigarette exposure, more in-depth studies are necessary to determine the effects of acute versus chronic exposure to e-cigarettes and also to understand how PG affects EGR2, ARC and other gene expression in the brain.

5. Conclusions

E-cigarette use has been marketed as a safer alternative to traditional cigarettes, as a means of smoking cessation, and is used at a higher rate than the general population in PWH. EGR2 and ARC have a role in addiction, synaptic plasticity, inflammation, and neurodegeneration. Overall, this study showed that 10 days of exposure to e-cigarette vapor was sufficient to alter gene expression in the brains of 6-month-old, male, Sprague Dawley rats. Specifically, PG significantly downregulated EGR2 and non-significantly downregulated ARC mRNA expression in frontal cortex, an effect which was reversed by NIC and THC. However, this did not translate to alterations in protein expression. PG-induced decreases in EGR2 mRNA in rat frontal cortex may suggest that PG could have a protective role against NIC and cannabis dependence. However, in vitro, PG upregulated EGR2 and ARC mRNA expression at 18 h in cultured C6 rat astrocytes, suggesting that PG may have neuroinflammatory effects. The finding that PG can induce ARC expression in astrocytes is novel because previous ARC studies have focused on neuronal expression. PG-induced significant upregulation of EGR2 and non-significant upregulation of ARC mRNA in cultured C6 rat astrocytes was reversed by NIC but not THC. In fact, THC enhanced the PG-induced increases in EGR2, which may have concerning implications for THC e-cigarette users. The HIV antiretroviral DTG reversed the effect NIC had on decreasing PG-induced upregulation of EGR2. This is concerning because EGR2 has been implicated in HIV latency reversal, T-cell apoptosis, and neuroinflammation, a process that underlies the development of HAND. Future in vitro and in vivo studies investigating the effects of e-cigarettes on addiction, neuroinflammation, and neurodegeneration are needed and should include HIV models, evaluation of the interaction between e-cigarettes and antiretrovirals, and focus on neuroimmune cells such as astrocytes and microglia.

Author Contributions

J.R.K. and L.N. drafted and edited the manuscript. A.A.L. and A.E.L. performed biochemical and molecular analyses of rat brain tissues and cell cultures. M.A.T., J.D.N. and J.A.F. conceptualized the project, analyzed and interpreted data, drafted and edited the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research was financially supported by NIH/NIMH grants 5R01MH128108 to JAF, 5R25MH101072 to JRK, and NIH/NIDA grant DA047413 to JDN. RNA sequencing was conducted at the IGM Genomics Center, University of California, San Diego, La Jolla, CA (P30DK063491; P30CA023100; P30DK120515). University of California Center for Medicinal Cannabis Research award P64-04-002 to JDN/MAT.

Institutional Review Board Statement

The animal study protocol was approved by the Institutional Review Board of University of California San Diego (protocol code S19029) for studies involving animals.

Informed Consent Statement

Not applicable.

Data Availability Statement

All data will be available upon reasonable request.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Meza, R.; Jimenez-Mendoza, E.; Levy, D.T. Trends in Tobacco Use among Adolescents by Grade, Sex, and Race, 1991–2019. JAMA Netw. Open 2020, 3, e2027465. [Google Scholar] [CrossRef] [PubMed]
  2. U.S. Department of Health and Human Services. E-Cigarette Use among Youth and Young Adults: A Report of the Surgeon General; Publications and Reports of the Surgeon General; U.S. Department of Health and Human Services: Washington, DC, USA, 2016.
  3. Hartmann-Boyce, J.; Lindson, N.; Butler, A.R.; McRobbie, H.; Bullen, C.; Begh, R.; Theodoulou, A.; Notley, C.; Rigotti, N.A.; Turner, T.; et al. Electronic cigarettes for smoking cessation. Cochrane Database Syst. Rev. 2022, 11, CD010216. [Google Scholar] [CrossRef] [PubMed]
  4. 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]
  5. McLaurin, K.A.; Li, H.; Mactutus, C.F.; Booze, R.M. Constitutive expression of HIV-1 viral proteins induces progressive synaptodendritic alterations in medium spiny neurons: Implications for substance use disorders. NeuroImmune Pharmacol. Ther. 2023, 2, 153–167. [Google Scholar] [CrossRef]
  6. Rahmanian, S.; Wewers, M.E.; Koletar, S.; Reynolds, N.; Ferketich, A.; Diaz, P. Cigarette smoking in the HIV-infected population. Proc. Am. Thorac. Soc. 2011, 8, 313–319. [Google Scholar] [CrossRef]
  7. Cheng, D.; Luo, Z.; Fitting, S.; Stoops, W.; Heath, S.L.; Ndhlovu, L.C.; Jiang, W. The link between chronic cocaine use, B cell perturbations, and blunted immune recovery in HIV-infected individuals on suppressive ART. NeuroImmune Pharmacol. Ther. 2023, 2, 71–79. [Google Scholar] [CrossRef]
  8. Harezlak, J.; Buchthal, S.; Taylor, M.; Schifitto, G.; Zhong, J.; Daar, E.; Alger, J.; Singer, E.; Campbell, T.; Yiannoutsos, C.; et al. Persistence of HIV-associated cognitive impairment, inflammation, and neuronal injury in era of highly active antiretroviral treatment. Aids 2011, 25, 625–633. [Google Scholar] [CrossRef]
  9. Alasmari, F.; Crotty Alexander, L.E.; Hammad, A.M.; Horton, A.; Alhaddad, H.; Schiefer, I.T.; Shin, J.; Moshensky, A.; Sari, Y. E-cigarette aerosols containing nicotine modulate nicotinic acetylcholine receptors and astroglial glutamate transporters in mesocorticolimbic brain regions of chronically exposed mice. Chem. Biol. Interact. 2021, 333, 109308. [Google Scholar] [CrossRef]
  10. Alasmari, F.; Crotty Alexander, L.E.; Nelson, J.A.; Schiefer, I.T.; Breen, E.; Drummond, C.A.; Sari, Y. Effects of chronic inhalation of electronic cigarettes containing nicotine on glial glutamate transporters and alpha-7 nicotinic acetylcholine receptor in female CD-1 mice. Prog. Neuropsychopharmacol. Biol. Psychiatry 2017, 77, 1–8. [Google Scholar] [CrossRef]
  11. Alzoubi, K.H.; Batran, R.M.; Al-Sawalha, N.A.; Khabour, O.F.; Karaoghlanian, N.; Shihadeh, A.; Eissenberg, T. The effect of electronic cigarettes exposure on learning and memory functions: Behavioral and molecular analysis. Inhal. Toxicol. 2021, 33, 234–243. [Google Scholar] [CrossRef]
  12. Hajdusianek, W.; Zorawik, A.; Waliszewska-Prosol, M.; Poreba, R.; Gac, P. Tobacco and Nervous System Development and Function-New Findings 2015–2020. Brain Sci. 2021, 11, 797. [Google Scholar] [CrossRef]
  13. George, O.; Grieder, T.E.; Cole, M.; Koob, G.F. Exposure to chronic intermittent nicotine vapor induces nicotine dependence. Pharmacol. Biochem. Behav. 2010, 96, 104–107. [Google Scholar] [CrossRef] [PubMed]
  14. Martinez, M.; Espinoza, V.E.; Garcia, V.; Uribe, K.P.; Negishi, K.; Estevao, I.L.; Carcoba, L.M.; O’Dell, L.E.; Khan, A.M.; Mendez, I.A. Withdrawal from repeated nicotine vapor exposure increases somatic signs of physical dependence, anxiety-like behavior, and brain reward thresholds in adult male rats. Neuropharmacology 2023, 240, 109681. [Google Scholar] [CrossRef]
  15. Gutierrez, A.; Creehan, K.M.; Taffe, M.A. A vapor exposure method for delivering heroin alters nociception, body temperature and spontaneous activity in female and male rats. J. Neurosci. Methods 2021, 348, 108993. [Google Scholar] [CrossRef] [PubMed]
  16. Javadi-Paydar, M.; Creehan, K.M.; Kerr, T.M.; Taffe, M.A. Vapor inhalation of cannabidiol (CBD) in rats. Pharmacol. Biochem. Behav. 2019, 184, 172741. [Google Scholar] [CrossRef] [PubMed]
  17. Javadi-Paydar, M.; Kerr, T.M.; Harvey, E.L.; Cole, M.; Taffe, M.A. Effects of nicotine and THC vapor inhalation administered by an electronic nicotine delivery system (ENDS) in male rats. Drug Alcohol. Depend. 2019, 198, 54–62. [Google Scholar] [CrossRef]
  18. Nguyen, J.D.; Aarde, S.M.; Vandewater, S.A.; Grant, Y.; Stouffer, D.G.; Parsons, L.H.; Cole, M.; Taffe, M.A. Inhaled delivery of Δ9-tetrahydrocannabinol (THC) to rats by e-cigarette vapor technology. Neuropharmacology 2016, 109, 112–120. [Google Scholar] [CrossRef]
  19. Nguyen, J.D.; Grant, Y.; Creehan, K.M.; Hwang, C.S.; Vandewater, S.A.; Janda, K.D.; Cole, M.; Taffe, M.A. Δ9-tetrahydrocannabinol attenuates oxycodone self-administration under extended access conditions. Neuropharmacology 2019, 151, 127–135. [Google Scholar] [CrossRef]
  20. Swinton, M.K.; Carson, A.; Telese, F.; Sanchez, A.B.; Soontornniyomkij, B.; Rad, L.; Batki, I.; Quintanilla, B.; Perez-Santiago, J.; Achim, C.L.; et al. Mitochondrial biogenesis is altered in HIV+ brains exposed to ART: Implications for therapeutic targeting of astroglia. Neurobiol. Dis. 2019, 130, 104502. [Google Scholar] [CrossRef]
  21. Canchi, S.; Swinton, M.K.; Rissman, R.A.; Fields, J.A. Transcriptomic analysis of brain tissues identifies a role for CCAAT enhancer binding protein beta in HIV-associated neurocognitive disorder. J. Neuroinflamm. 2020, 17, 112. [Google Scholar] [CrossRef]
  22. Fields, J.; Dumaop, W.; Rockenstein, E.; Mante, M.; Spencer, B.; Grant, I.; Ellis, R.; Letendre, S.; Patrick, C.; Adame, A.; et al. Age-dependent molecular alterations in the autophagy pathway in HIVE patients and in a gp120 tg mouse model: Reversal with beclin-1 gene transfer. J. NeuroVirology 2013, 19, 89–101. [Google Scholar] [CrossRef]
  23. Merecz-Sadowska, A.; Sitarek, P.; Zielinska-Blizniewska, H.; Malinowska, K.; Zajdel, K.; Zakonnik, L.; Zajdel, R. A Summary of In Vitro and In Vivo Studies Evaluating the Impact of E-Cigarette Exposure on Living Organisms and the Environment. Int. J. Mol. Sci. 2020, 21, 652. [Google Scholar] [CrossRef] [PubMed]
  24. Salery, M.; Dos Santos, M.; Saint-Jour, E.; Moumne, L.; Pages, C.; Kappes, V.; Parnaudeau, S.; Caboche, J.; Vanhoutte, P. Activity-Regulated Cytoskeleton-Associated Protein Accumulates in the Nucleus in Response to Cocaine and Acts as a Brake on Chromatin Remodeling and Long-Term Behavioral Alterations. Biol. Psychiatry 2017, 81, 573–584. [Google Scholar] [CrossRef]
  25. Belluardo, N.; Olsson, P.A.; Mudo, G.; Sommer, W.H.; Amato, G.; Fuxe, K. Transcription factor gene expression profiling after acute intermittent nicotine treatment in the rat cerebral cortex. Neuroscience 2005, 133, 787–796. [Google Scholar] [CrossRef] [PubMed]
  26. EGR2 Gene-Early Growth Response 2. Available online: https://www.genecards.org/cgi-bin/carddisp.pl?gene=EGR2#proteins-ptm (accessed on 27 October 2023).
  27. Beckmann, A.M.; Wilce, P.A. Egr transcription factors in the nervous system. Neurochem. Int. 1997, 31, 477–510, discussion 517–476. [Google Scholar] [CrossRef]
  28. Chen, Y.; Wang, X.; Xiao, B.; Luo, Z.; Long, H. Mechanisms and Functions of Activity-Regulated Cytoskeleton-Associated Protein in Synaptic Plasticity. Mol. Neurobiol. 2023, 60, 5738–5754. [Google Scholar] [CrossRef] [PubMed]
  29. Alberini, C.M. Transcription factors in long-term memory and synaptic plasticity. Physiol. Rev. 2009, 89, 121–145. [Google Scholar] [CrossRef]
  30. Nikolaienko, O.; Patil, S.; Eriksen, M.S.; Bramham, C.R. Arc protein: A flexible hub for synaptic plasticity and cognition. Semin. Cell Dev. Biol. 2018, 77, 33–42. [Google Scholar] [CrossRef]
  31. Yakout, D.W.; Shree, N.; Mabb, A.M. Effect of pharmacological manipulations on Arc function. Curr. Res. Pharmacol. Drug Discov. 2021, 2, 100013. [Google Scholar] [CrossRef]
  32. Appelbaum, L.G.; Shenasa, M.A.; Stolz, L.; Daskalakis, Z. Synaptic plasticity and mental health: Methods, challenges and opportunities. Neuropsychopharmacology 2023, 48, 113–120. [Google Scholar] [CrossRef]
  33. Rosi, S. Neuroinflammation and the plasticity-related immediate-early gene Arc. Brain Behav. Immun. 2011, 25 (Suppl. S1), S39–S49. [Google Scholar] [CrossRef] [PubMed]
  34. Pagano, R.; Salamian, A.; Zielinski, J.; Beroun, A.; Nalberczak-Skora, M.; Skonieczna, E.; Caly, A.; Tay, N.; Banaschewski, T.; Desrivieres, S.; et al. Arc controls alcohol cue relapse by a central amygdala mechanism. Mol. Psychiatry 2023, 28, 733–745. [Google Scholar] [CrossRef] [PubMed]
  35. Mukherjee, D.; Gonzales, B.J.; Ashwal-Fluss, R.; Turm, H.; Groysman, M.; Citri, A. Egr2 induction in spiny projection neurons of the ventrolateral striatum contributes to cocaine place preference in mice. Elife 2021, 10, e65228. [Google Scholar] [CrossRef] [PubMed]
  36. Imperio, C.G.; McFalls, A.J.; Hadad, N.; Blanco-Berdugo, L.; Masser, D.R.; Colechio, E.M.; Coffey, A.A.; Bixler, G.V.; Stanford, D.R.; Vrana, K.E.; et al. Exposure to environmental enrichment attenuates addiction-like behavior and alters molecular effects of heroin self-administration in rats. Neuropharmacology 2018, 139, 26–40. [Google Scholar] [CrossRef] [PubMed]
  37. Ammon, S.; Mayer, P.; Riechert, U.; Tischmeyer, H.; Hollt, V. Microarray analysis of genes expressed in the frontal cortex of rats chronically treated with morphine and after naloxone precipitated withdrawal. Brain Res. Mol. Brain Res. 2003, 112, 113–125. [Google Scholar] [CrossRef]
  38. Kim, M.; Jeon, S.J.; Custodio, R.J.; Lee, H.J.; Sayson, L.V.; Ortiz, D.M.D.; Cheong, J.H.; Kim, H.J. Gene Expression Profiling in the Striatum of Per2 KO Mice Exhibiting More Vulnerable Responses against Methamphetamine. Biomol. Ther. 2021, 29, 135–143. [Google Scholar] [CrossRef]
  39. Gao, P.; Limpens, J.H.; Spijker, S.; Vanderschuren, L.J.; Voorn, P. Stable immediate early gene expression patterns in medial prefrontal cortex and striatum after long-term cocaine self-administration. Addict. Biol. 2017, 22, 354–368. [Google Scholar] [CrossRef]
  40. Kozela, E.; Juknat, A.; Kaushansky, N.; Ben-Nun, A.; Coppola, G.; Vogel, Z. Cannabidiol, a non-psychoactive cannabinoid, leads to EGR2-dependent anergy in activated encephalitogenic T cells. J. Neuroinflamm. 2015, 12, 52. [Google Scholar] [CrossRef]
  41. Llorente-Berzal, A.; Puighermanal, E.; Burokas, A.; Ozaita, A.; Maldonado, R.; Marco, E.M.; Viveros, M.P. Sex-dependent psychoneuroendocrine effects of THC and MDMA in an animal model of adolescent drug consumption. PLoS ONE 2013, 8, e78386. [Google Scholar] [CrossRef]
  42. Adams, K.W.; Kletsov, S.; Lamm, R.J.; Elman, J.S.; Mullenbrock, S.; Cooper, G.M. Role for Egr1 in the Transcriptional Program Associated with Neuronal Differentiation of PC12 Cells. PLoS ONE 2017, 12, e0170076. [Google Scholar] [CrossRef]
  43. Kawashima, T.; Okuno, H.; Nonaka, M.; Adachi-Morishima, A.; Kyo, N.; Okamura, M.; Takemoto-Kimura, S.; Worley, P.F.; Bito, H. Synaptic activity-responsive element in the Arc/Arg3.1 promoter essential for synapse-to-nucleus signaling in activated neurons. Proc. Natl. Acad. Sci. USA 2009, 106, 316–321. [Google Scholar] [CrossRef] [PubMed]
  44. Epstein, I.; Finkbeiner, S. The Arc of cognition: Signaling cascades regulating Arc and implications for cognitive function and disease. Semin. Cell Dev. Biol. 2018, 77, 63–72. [Google Scholar] [CrossRef] [PubMed]
  45. Chen, T.; Xu, Y.P.; Chen, Y.; Sun, S.; Yan, Z.Z.; Wang, Y.H. Arc regulates brain damage and neuroinflammation via Sirt1 signaling following subarachnoid hemorrhage. Brain Res. Bull. 2023, 203, 110780. [Google Scholar] [CrossRef] [PubMed]
  46. Yang, Z.; Li, H.; Tang, Y.; Liu, X.; Liao, Q.; Fan, C.; Wang, S. CYP1B1 deiciency ameliorates learning and memory deficits caused by high fat diet in mice. Am. J. Transl. Res. 2019, 11, 2194–2206. [Google Scholar] [PubMed]
  47. Snow, W.M.; Pahlavan, P.S.; Djordjevic, J.; McAllister, D.; Platt, E.E.; Alashmali, S.; Bernstein, M.J.; Suh, M.; Albensi, B.C. Morris Water Maze Training in Mice Elevates Hippocampal Levels of Transcription Factors Nuclear Factor (Erythroid-derived 2)-like 2 and Nuclear Factor Kappa B p65. Front. Mol. Neurosci. 2015, 8, 70. [Google Scholar] [CrossRef]
  48. Kozela, E.; Juknat, A.; Gao, F.; Kaushansky, N.; Coppola, G.; Vogel, Z. Pathways and gene networks mediating the regulatory effects of cannabidiol, a nonpsychoactive cannabinoid, in autoimmune T cells. J. Neuroinflamm. 2016, 13, 136. [Google Scholar] [CrossRef]
  49. Harris, A.C.; Muelken, P.; Haave, Z.; Swain, Y.; Smethells, J.R.; LeSage, M.G. Propylene glycol, a major electronic cigarette constituent, attenuates the adverse effects of high-dose nicotine as measured by intracranial self-stimulation in rats. Drug Alcohol. Depend. 2018, 193, 162–168. [Google Scholar] [CrossRef]
  50. Bains, M.; Hall, E.D. Antioxidant therapies in traumatic brain and spinal cord injury. Biochim. Biophys. Acta 2012, 1822, 675–684. [Google Scholar] [CrossRef]
  51. Czimmerer, Z.; Nagy, L. Epigenomic regulation of macrophage polarization: Where do the nuclear receptors belong? Immunol. Rev. 2023, 317, 152–165. [Google Scholar] [CrossRef]
  52. Okamura, T.; Fujio, K.; Sumitomo, S.; Yamamoto, K. Roles of LAG3 and EGR2 in regulatory T cells. Ann. Rheum. Dis. 2012, 71 (Suppl. S2), i96–i100. [Google Scholar] [CrossRef]
  53. Ufer, F.; Vargas, P.; Engler, J.B.; Tintelnot, J.; Schattling, B.; Winkler, H.; Bauer, S.; Kursawe, N.; Willing, A.; Keminer, O.; et al. Arc/Arg3.1 governs inflammatory dendritic cell migration from the skin and thereby controls T cell activation. Sci. Immunol. 2016, 1, eaaf8665. [Google Scholar] [CrossRef] [PubMed]
  54. Zhang, Z.Z.; Zeng, L.P.; Chen, J.; Wu, Y.F.; Wang, Y.T.; Xia, L.; Yang, Q.G.; Wang, F.; Chen, G.H. Long-Term Environmental Enrichment Relieves Dysfunctional Cognition and Synaptic Protein Levels Induced by Prenatal Inflammation in Older CD-1 Mice. Neural Plast. 2022, 2022, 1483101. [Google Scholar] [CrossRef] [PubMed]
  55. Vemula, S.V.; Maxwell, J.W.; Nefedov, A.; Wan, B.L.; Steve, J.; Newhard, W.; Sanchez, R.I.; Tellers, D.; Barnard, R.J.; Blair, W.; et al. Identification of proximal biomarkers of PKC agonism and evaluation of their role in HIV reactivation. Antivir. Res. 2017, 139, 161–170. [Google Scholar] [CrossRef] [PubMed]
  56. Yang, Y.; Dong, B.; Mittelstadt, P.R.; Xiao, H.; Ashwell, J.D. HIV Tat binds Egr proteins and enhances Egr-dependent transactivation of the Fas ligand promoter. J. Biol. Chem. 2002, 277, 19482–19487. [Google Scholar] [CrossRef] [PubMed]
  57. Topilko, P.; Schneider-Maunoury, S.; Levi, G.; Baron-Van Evercooren, A.; Chennoufi, A.B.; Seitanidou, T.; Babinet, C.; Charnay, P. Krox-20 controls myelination in the peripheral nervous system. Nature 1994, 371, 796–799. [Google Scholar] [CrossRef] [PubMed]
  58. Shy, M.E.; Garbern, J.Y.; Kamholz, J. Hereditary motor and sensory neuropathies: A biological perspective. Lancet Neurol. 2002, 1, 110–118. [Google Scholar] [CrossRef]
  59. Wang, Z.; Dong, X.; Cong, S. Bioinformatics analysis of the molecular mechanism underlying Huntington’s disease. J. Integr. Neurosci. 2019, 18, 369–376. [Google Scholar] [CrossRef]
  60. Yin, Z.; Raj, D.; Saiepour, N.; Van Dam, D.; Brouwer, N.; Holtman, I.R.; Eggen, B.J.L.; Moller, T.; Tamm, J.A.; Abdourahman, A.; et al. Immune hyperreactivity of Abeta plaque-associated microglia in Alzheimer’s disease. Neurobiol. Aging 2017, 55, 115–122. [Google Scholar] [CrossRef]
  61. Karrer, M.; Lopez, M.A.; Meier, D.; Mikhail, C.; Ogunshola, O.O.; Muller, A.F.; Strauss, L.; Tafti, M.; Fontana, A. Cytokine-induced sleep: Neurons respond to TNF with production of chemokines and increased expression of Homer1a in vitro. Brain Behav. Immun. 2015, 47, 186–192. [Google Scholar] [CrossRef]
  62. Yan, Y.; Tan, X.; Wu, X.; Shao, B.; Wu, X.; Cao, J.; Xu, J.; Jin, W.; Li, L.; Xu, W.; et al. Involvement of early growth response-2 (Egr-2) in lipopolysaccharide-induced neuroinflammation. J. Mol. Histol. 2013, 44, 249–257. [Google Scholar] [CrossRef]
  63. Werley, M.S.; McDonald, P.; Lilly, P.; Kirkpatrick, D.; Wallery, J.; Byron, P.; Venitz, J. Non-clinical safety and pharmacokinetic evaluations of propylene glycol aerosol in Sprague-Dawley rats and Beagle dogs. Toxicology 2011, 287, 76–90. [Google Scholar] [CrossRef] [PubMed]
  64. Li, Y.; Pehrson, A.L.; Waller, J.A.; Dale, E.; Sanchez, C.; Gulinello, M. A critical evaluation of the activity-regulated cytoskeleton-associated protein (Arc/Arg3.1)’s putative role in regulating dendritic plasticity, cognitive processes, and mood in animal models of depression. Front. Neurosci. 2015, 9, 279. [Google Scholar] [CrossRef] [PubMed]
  65. Escobar, Y.H.; Nipp, G.; Cui, T.; Petters, S.S.; Surratt, J.D.; Jaspers, I. In Vitro Toxicity and Chemical Characterization of Aerosol Derived from Electronic Cigarette Humectants Using a Newly Developed Exposure System. Chem. Res. Toxicol. 2020, 33, 1677–1688. [Google Scholar] [CrossRef] [PubMed]
  66. Mahmoudzadeh, L.; Abtahi Froushani, S.M.; Ajami, M.; Mahmoudzadeh, M. Effect of Nicotine on Immune System Function. Adv. Pharm. Bull. 2023, 13, 69–78. [Google Scholar] [CrossRef] [PubMed]
  67. Min, A.K.; Keane, A.M.; Weinstein, M.P.; Swartz, T.H. The impact of cannabinoids on inflammasome signaling in HIV-1 infection. NeuroImmune Pharmacol. Ther. 2023, 2, 79–88. [Google Scholar] [CrossRef] [PubMed]
  68. Henshaw, F.R.; Dewsbury, L.S.; Lim, C.K.; Steiner, G.Z. The Effects of Cannabinoids on Pro- and Anti-Inflammatory Cytokines: A Systematic Review of In Vivo Studies. Cannabis Cannabinoid Res. 2021, 6, 177–195. [Google Scholar] [CrossRef]
Figure 3. PG (propylene glycol) alters EGR2 and ARC mRNA expression at 6 h and increases EGR2 and ARC mRNA expression in a dose-dependent manner at 18 h in cultured C6 rat astrocytes. Fold change in (a) EGR2 and (b) ARC mRNA levels normalized to ACTB following 18 h treatment with 5 mM, 50 mM, or 150 mM PG. Fold in of (c) EGR2 and (d) ARC mRNA levels normalized to ACTB following 6 h or 18 h treatment with 150 mM PG. Mean ± SEM. Statistical significance was determined by one-way ANOVA, post hoc Tukey’s. (a,b) * p < 0.05, ** p < 0.01 vs. vehicle; ^^ p <0.01 vs. 150 mM PG (c,d) **** p < 0.0001 vs. 0 h; ^^ p < 0.005 vs. 6 h, ^^^^ p < 0.00001 vs. 6 h.
Figure 3. PG (propylene glycol) alters EGR2 and ARC mRNA expression at 6 h and increases EGR2 and ARC mRNA expression in a dose-dependent manner at 18 h in cultured C6 rat astrocytes. Fold change in (a) EGR2 and (b) ARC mRNA levels normalized to ACTB following 18 h treatment with 5 mM, 50 mM, or 150 mM PG. Fold in of (c) EGR2 and (d) ARC mRNA levels normalized to ACTB following 6 h or 18 h treatment with 150 mM PG. Mean ± SEM. Statistical significance was determined by one-way ANOVA, post hoc Tukey’s. (a,b) * p < 0.05, ** p < 0.01 vs. vehicle; ^^ p <0.01 vs. 150 mM PG (c,d) **** p < 0.0001 vs. 0 h; ^^ p < 0.005 vs. 6 h, ^^^^ p < 0.00001 vs. 6 h.
Brainsci 13 01556 g003
Figure 4. PG (propylene glycol) increases EGR2 and ARC mRNA expression in C6 rat astrocytes at 18 h and is reversed by Nicotine (NIC), an effect that is lost with the addition of dolutegravir (DTG). Fold change in (a) EGR2 and (b) ARC mRNA levels normalized to ATCB following 18 h treatment with Vehicle, PG (150 mM), PG + THC (10µM), PG + NIC (10µM), DTG (200 nM), DTG, PG + DTG, PG + THC + DTG, or PG + NIC + DTG. Mean ± SEM. Statistical significance was determined by one-way ANOVA, post hoc Tukey’s. ** p < 0.01, *** p <0.001, **** p < 0.0001 vs. vehicle, ^ p <0.05, ^^^ p < 0.001, ^^^^ p < 0.0001 vs. DTG.
Figure 4. PG (propylene glycol) increases EGR2 and ARC mRNA expression in C6 rat astrocytes at 18 h and is reversed by Nicotine (NIC), an effect that is lost with the addition of dolutegravir (DTG). Fold change in (a) EGR2 and (b) ARC mRNA levels normalized to ATCB following 18 h treatment with Vehicle, PG (150 mM), PG + THC (10µM), PG + NIC (10µM), DTG (200 nM), DTG, PG + DTG, PG + THC + DTG, or PG + NIC + DTG. Mean ± SEM. Statistical significance was determined by one-way ANOVA, post hoc Tukey’s. ** p < 0.01, *** p <0.001, **** p < 0.0001 vs. vehicle, ^ p <0.05, ^^^ p < 0.001, ^^^^ p < 0.0001 vs. DTG.
Brainsci 13 01556 g004
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

Kulbe, J.R.; Nguyen, L.; Le, A.A.; Laird, A.E.; Taffe, M.A.; Nguyen, J.D.; Fields, J.A. Nicotine, THC, and Dolutegravir Modulate E-Cigarette-Induced Changes in Addiction- and Inflammation-Associated Genes in Rat Brains and Astrocytes. Brain Sci. 2023, 13, 1556. https://doi.org/10.3390/brainsci13111556

AMA Style

Kulbe JR, Nguyen L, Le AA, Laird AE, Taffe MA, Nguyen JD, Fields JA. Nicotine, THC, and Dolutegravir Modulate E-Cigarette-Induced Changes in Addiction- and Inflammation-Associated Genes in Rat Brains and Astrocytes. Brain Sciences. 2023; 13(11):1556. https://doi.org/10.3390/brainsci13111556

Chicago/Turabian Style

Kulbe, Jacqueline Renee, Lauren Nguyen, Alexandra Anh Le, Anna Elizabeth Laird, Michael A. Taffe, Jacques D. Nguyen, and Jerel Adam Fields. 2023. "Nicotine, THC, and Dolutegravir Modulate E-Cigarette-Induced Changes in Addiction- and Inflammation-Associated Genes in Rat Brains and Astrocytes" Brain Sciences 13, no. 11: 1556. https://doi.org/10.3390/brainsci13111556

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