Hostname: page-component-745bb68f8f-v2bm5 Total loading time: 0 Render date: 2025-02-11T13:33:12.593Z Has data issue: false hasContentIssue false

Anti-HIV drugs promote β-amyloid deposition and impair learning and memory in BALB/c mice

Published online by Cambridge University Press:  07 May 2020

S.S. Zulu*
Affiliation:
School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban, South Africa
O. Abboussi
Affiliation:
Division of Neuroscience, School of Medicine, Ninewells Hospital, University of Dundee, Dundee, UK
N. Simola
Affiliation:
Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
M.V. Mabandla
Affiliation:
School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban, South Africa
W.M.U. Daniels
Affiliation:
School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
*
Author for correspondence: S.S. Zulu, Emails: zuluss86@gmail.com or 207506590@stu.ukzn.ac.za
Rights & Permissions [Opens in a new window]

Abstract

Objectives:

Growing evidence suggested that antiretroviral (ARV) drugs may promote amyloid beta (Aβ) accumulation in HIV-1-infected brain and the persistence of HIV-associated neurocognitive disorders (HANDs). It has also been shown that lipid peroxidation upregulates β-site amyloid precursor protein (APP) cleaving enzyme 1 (BACE1) expression and subsequently promotes Aβ peptide production. In the present study, we examined whether chronic exposure to the anti-HIV drugs tenofovir disoproxil fumarate (TDF) and nevirapine induces lipid peroxidation thereby promoting BACE1 and Aβ generation and consequently impair cognitive function in mice.

Methods:

TDF or nevirapine was orally administered to female BALB/c mice once a day for 8 weeks. On the 7th week of treatment, spatial learning and memory were assessed using the Morris water maze test. The levels of lipid peroxidation, BACE1, amyloid β 1-42 (Aβ1-42) and Aβ deposits were measured in the hippocampal tissue upon completion of treatment.

Results:

Chronic administration of nevirapine induced spatial learning and memory impairment in the Morris water maze test, whereas TDF did not have an effect. TDF and nevirapine administration increased hippocampal lipid peroxidation and Aβ1-42 concentration. Nevirapine further upregulated BACE1 expression and Aβ deposits.

Conclusion:

Our results suggest that chronic exposure to TDF and nevirapine contributes to hippocampal lipid peroxidation and Aβ accumulation, respectively, as well as spatial learning and memory deficits in mice even in the absence of HIV infection. These findings further support a possible link between ARV drug toxicity, Aβ accumulation and the persistence of HANDs.

Type
Original Article
Copyright
© Scandinavian College of Neuropsychopharmacology 2020

Significant outcomes

  • Chronic exposure to the ARV drug nevirapine impairs spatial learning and memory in mice.

  • TDF and nevirapine increase level of lipid peroxidation and Aβ1-42 concentration.

  • Nevirapine upregulates BACE1 and Aβ deposition in hippocampus.

Limitations

  • ARV drugs treatments were (TDF and nevirapine) administered to naive mice; therefore, the synergistic effects of HIV and ARV treatment on cognitive function are lacking.

  • Although doses of ARV used in this study were comparable to human therapeutic doses, they do not account for differences in drug metabolism rates.

  • We only conducted a cognitive assessment on the Morris water maze. More behavioural assessments are needed in future studies to test other cognitive domains.

Introduction

Among HIV comorbidities, HIV-associated neurocognitive disorder (HAND) is commonly observed in persons living with HIV (McArthur et al., Reference Mcarthur, Steiner, Sacktor and Nath2010). Recent clinical observations during the antiretroviral therapy (ART) era have indicated that HAND may occur in 15–50% of HIV-infected patients (Bryant et al., Reference Bryant, Ellis, Umlauf, Gouaux, Soontornniyomkij, Letendre, Achim, Masliah, Grant and Moore2015). The introduction of combination ART (cART) has improved the well-being of HIV patients, and the prevalence of severe HIV-associated dementia has decreased significantly (Sacktor et al., Reference Sacktor, Lyles, Skolasky, Kleeberger, Selnes, Miller, Becker, Cohen and Mcarthur2001). Despite this promising outcome, the prevalence of milder forms of HAND which includes mild dysfunctions in attention, learning/memory, working memory, executive function and fine motor skills (Woods et al., Reference Woods, Moore, Weber and Grant2009) has increased significantly (Gannon, Reference Gannon2014; Shah et al., Reference Shah, Gangwani, Chaudhari, Glazyrin, Bhat and Kumar2016). However, it remains unclear whether the increased prevalence of mild forms of HAND is due to the intrinsic risk of developing dementia with age or other direct or indirect factors of the condition itself such as the prolonged exposure to antiretroviral (ARV) drugs (Ciccarelli et al., Reference Ciccarelli, Fabbiani, di Giambenedetto, Fanti, Baldonero, Bracciale, Tamburrini, Cauda, de Luca and Silveri2011; Giunta et al., Reference Giunta, Ehrhart, Obregon, Lam, Le, Jin, Fernandez, Tan and Shytle2011). Studies investigating the potential adverse effects of ARVs in the central nervous system (CNS) remains surprisingly few, and even less in females, despite the emerging evidence linking certain ARVs to adverse neuropsychiatry outcomes. For instance, Ciccarelli et al. (Reference Ciccarelli, Fabbiani, di Giambenedetto, Fanti, Baldonero, Bracciale, Tamburrini, Cauda, de Luca and Silveri2011) found that 47% patients receiving efavirenz and tenofovir disoproxil fumarate (TDF) had cognitive impairments. A recent study found that efavirenz treatment in adolescents cause fronto-striatal dysfunction indicated by blunts proactive inhibitory behavioural responses (Du Plessis et al., Reference du Plessis, Perez, Fouche, Phillips, Joska, Vink, Myer, Zar, Stein and Hoare2019). This finding suggests that ARV may contribute to the persistence of HAND. Therefore, is a need to understand the molecular mechanisms underlying these neuropsychiatric dysfunctions for the proper management of HIV-infected individuals.

There are at least six commonly prescribed classes of drugs available for the treatment of HIV infection: (1) nucleoside reverse transcriptase inhibitors (NRTIs), (2) non-nucleoside reverse transcriptase inhibitors (NNRTI), (3) protease inhibitors (PI), (4) maturation inhibitors, (5) integrase inhibitors and (6) entry inhibitors (Ene et al., Reference Ene, Duiculescu and Ruta2011; Davies & Taylor, Reference Davies and Taylor2013). NRTIs and NNRTIs are widely used and are considered the backbone of ART. TDF is a commonly used NRTI and part of the regime of first-line drugs. The most common side effect of TDF is nephrotoxicity (Kohler et al., Reference Kohler, Hosseini, Hoying-Brandt, Green, Johnson, Russ, Tran, Raper, Santoianni and Lewis2009; Lebrecht et al., Reference Lebrecht, Venhoff, Kirschner, Wiech, Venhoff and Walker2009; Cote et al., Reference Cote, Magil, Harris, Scarth, Gadawski, Wang, Yu, Yip, Zalunardo, Werb, Hogg, Harrigan and Montaner2006). Further, it was reported some adults experience adverse neuropsychiatric events after switching from a regimen of efavirenz without TDF to a regimen of EFV with TDF (Allavena et al., Reference Allavena, Le Moal, Michau, Chiffoleau and Raffi2006). Nevirapine, on the other hand, is one of the first NNRTI’s to be introduced and has been the key drug to prevent mother to child transmission of HIV. Nonetheless, nevirapine has also been associated with adverse effects such as gastric complications, hepatotoxicity, and neuropsychiatric complications including cognitive impairments and hallucinations in HIV-infected individuals (Wise et al., Reference Wise, Mistry and Reid2002). Further, an in vivo study showed that nevirapine treatment impair recognition (Romão et al., Reference Romão, Lemos, Moreira, de Chaves, Moretti, Castro, Andrade, Boeck, Quevedo and Gavioli2011). These studies suggest that ARV drug contribute to the neuropathogenesis of HAND. However, molecular mechanism remains undefined.

There growing evidence that HIV infected individuals may present intraneuronal amyloid beta (Aβ) accumulation or perivascular diffuse Aβ depositions, whereas extracellular amyloid plaques are predominant features in Alzheimer’s disease (Andras & Toborek, Reference Andras and Toborek2013; Ortega & Ances, Reference Ortega and Ances2014). Although the molecular underlying is unknown, in vitro studies have shown that ARV expose increase reactive oxygen species (ROS) production initiating endoplasmic reticulum stress in culture cells (Akay et al., Reference Akay, Cooper, Odeleye, Jensen, White, Vassoler, Gannon, Mankowski, Dorsey, Buch, Cross, Cook, Pena, Andersen, Christofidou-Solomidou, Lindl, Zink, Clements, Pierce, Kolson and Jordan-Sciutto2014b; Nooka & Ghorpade, Reference Nooka and Ghorpade2017). The increased ROS production triggers a cascade of events including increased lipid peroxidation and β-site amyloid precursor protein (APP) cleaving enzyme 1 (BACE1) (Tamagno et al., Reference Tamagno, Guglielmotto, Aragno, Borghi, Autelli, Giliberto, Muraca, Danni, Zhu, Smith, Perry, Jo, Mattson and Tabaton2008, Reference Tamagno, Guglielmotto, Monteleone and Tabaton2012). The BACE1 is a key enzyme involved in the breakdown of the membrane bound amyloid-β precursor protein (AβPP) to generate Aβ. Also, Cerebral Aβ deposition-induced impairment of cognitive behaviour has previously been reported to be greater in female than in male mice (Wang et al., Reference Wang, Tanila, Puoliväli, Kadish and Van Groen2003; Howlett et al., Reference Howlett, Richardson, Austin, Parsons, Bate, Davies and Gonzalez2004).

Aim of this study

The present study, therefore, investigated whether chronic administration of ARV drugs, TDF and nevirapine to naïve BALB/c mice induce molecules changes linked to cognitive impairments. We hypothesised that chronic administration of these drugs would cause lipid peroxidation and upregulate BACE1 to process AβPP and increase Aβ production in the hippocampus.

Materials and methods

Experimental approach

To investigate the effects TDF and nevirapine on cognitive function, lipid peroxidation and Aβ generation, we performed an in vivo experiment using BALB/c mice. A total of 30 BALB/c female mice were assigned randomly by allocation into 3 groups (n = 10 per group) in this order: (1) Control, (2) TDF and (3) nevirapine. Animals were administered their respective treatments for a period of 8 weeks. On week 7 of treatment, they were subjected to behavioural assessment. After 8 weeks animals were sacrificed, hippocampal tissue (n = 6 per group) were used for biochemical investigations. Lipid peroxidation was assessed by 4-hydroxynonenal (4-HNE) and Aβ1-42 measurement by ELISA, BACE1 expression was assessed by Western Blotting. Immunohistochemistry (n = 4 per group) was used to measure Aβ deposition in the hippocampus.

Animals

The 30 female BALB/c mice (812 weeks old) used in this study were obtained from the Biomedical Resource Unit (BRU) at the University of KwaZulu-Natal. Female BALB/c mice were chosen firstly because preclinical studies investigating the long term effects of ART in this gender, are limited, in spite of this gender being more susceptible to drug toxicity than males (Zopf et al., Reference Zopf, Rabe, Neubert, Gaßmann, Rascher, Hahn, Brune and Dormann2008; Amacher, Reference Amacher2014) and secondly, because previous studies have shown that the formation of Aβ aggregates can be induced in this mouse strain (Boelen et al., Reference Boelen, Stassen, van der Ven, Lemmens, Steinbusch, Bruggeman, Schmitz and Steinbusch2007; Little et al., Reference Little, Joyce, Hammond, Matta, Cahn, Appelt and Balin2014). Animals were housed under standard laboratory conditions with a 12-h light-dark cycle, 23–25 °C room temperature, humidity of 55 ± 5, food and water available ad libitum. Animal’s oestrus cycle was synchronised 7 days before experiments were initiated (Schank & McClintock, Reference Schank and Mcclintock1992; Zuena et al., Reference Zuena, Giuli, Venerosi Pesciolini, Tramutola, Ajmone-Cat, Cinque, Alema, Giovine, Peluso, Minghetti, Nicolai, Calamandrei and Casolini2013). All experimental protocols and care of animals were approved by the University of KwaZulu-Natal Ethics Committee (Protocol reference number AREC/075/015D).

Drugs and treatment

ARV drugs were purchased from Pharmed Pharmaceutical (Pty) Ltd, South Africa. Mice were administered dose of 5 mg/kg/day TDF or 3.3 mg/kg/day Nevirapine for 8 weeks. These drugs (TDF and nevirapine) have been approved by the FDA as therapy for HIV infected humans and are administered orally by patients. Therefore, to remain clinically relevant, animal dosing was performed daily (morning) via oral gavage. There is limited information in the literature regarding ARV drug metabolism and clearance in animal models, hence a dose equivalent to human therapeutic dose was used (dose calculation- human TDF daily dose: 300 mg/60 kg human = 5 mg/kg; 30 g mouse = 0.150 mg/day). Treatments included TDF (0.150 mg/day), Nevirapine (0.1 mg/day), or vehicle (distilled water). This treatment regime also allowed comparison to previous studies which evaluated ARV toxicity mouse models (Kohler et al., Reference Kohler, Hosseini, Hoying-Brandt, Green, Johnson, Russ, Tran, Raper, Santoianni and Lewis2009; Romão et al., Reference Romão, Lemos, Moreira, de Chaves, Moretti, Castro, Andrade, Boeck, Quevedo and Gavioli2011; de Oliveira et al., Reference de Oliveira, Damiani, Dias Rde, Romao and Andrade2014). Drugs treatments were freshly prepared daily by dissolving tablets (TDF and nevirapine) in distilled water and then administered to the animals via oral gavage. The control group received distilled water also via oral gavage. The weight of the animals was recorded daily, and drug volumes were adjusted accordingly (10 ml/kg).

Morris water maze

On week 7 of treatment, all mice were subjected to the Morris water maze which is an established test used to assess spatial learning and memory in rodents (Vorhees & Williams, Reference Vorhees and Williams2006). Our Morris water maze consisted of a circular pool with a 120 cm diameter and a wall height of 85 cm. It had a hidden platform of 10 × 10 cm located in one of the quadrants. The pool was filled with water to a depth of 30 cm (22 ± 1 °C) that was 1 cm above the level of the platform. Visual cues were attached to the walls of the pool and to the walls of the testing room to orientate the animals to the location of the platform. Each mouse was positioned with its face towards the wall and released from each of the four different quadrants of the maze to find the hidden platform. A time interval of 60 s was allowed for the animal to find the platform. If the animal failed to locate the platform, it was guided to the platform and allowed to remain on the platform for 15 s to orientate itself. This training session was repeated for 5 consecutive days. Time to reach the hidden platform was recorded and is reported as escape latency. On the sixth day, the platform was removed, and memory retention of the platform location was assessed. The time spent in the target quadrant was therefore recorded. Behavioural assessments were manually scored during the test by an experience investigator.

Tissue collection for respective assays

On the final day of drug administration 6 hours after the final drug dosage, animals to be used for ELISA analysis were euthanised and immediately decapitated, dorsal hippocampal tissue was harvested, snap frozen in liquid nitrogen and stored in a bio-freezer at −80 °C for subsequent biochemical analyses. For the immunohistochemistry procedure, whole brains were collected following transcardial perfusion.

Mouse 4-HNE ELISA

4-HNE levels were quantified using an ELISA kit (Elabscience Biotechnology Co., China). Hippocampal tissue was homogenised in phosphate-buffered saline (0.01 M, pH = 7.4). The tissue homogenate was centrifuged for 5 min at 5000 g. The supernatant (100 µl) of each sample or HNE standards were added to each well of a 96-well plate and incubated for 10 min at 37 °C. After incubation, wells were washed and 100 µl of biotinylated antibody was added to each well and incubated at 37 °C for a further 1 h on an orbital shaker. After incubation, wells were washed after which a 100 µl of horseradish peroxidase (HRP) conjugated working solution was added to each well. The plate was then incubated for 30 min at 37 °C. The wells were once again washed, and the substrate solution was added to each well. The optical density (OD) of each well was measured using a micro-plate reader set to 450 nm as per the manufacturer’s protocol.

Western blotting

Dorsal hippocampal tissue was lysed in RIPA lysis buffer (w/v) (Sigma, USA). The homogenates were centrifuged at 3578g for 10 min at 4 °C. Thereafter, supernatants were extracted and their protein concentration determined using the Bradford method (Kruger, Reference Kruger2009). Samples were then denatured for 5 min at 95 °C. Equal amounts of protein (20 µg) were resolved by electrophoresis on a 10% sodium dodecyl-sulfate polyacrylamide gel in running buffer (Bio-Rad, South Africa), at 200 V for 1 h. Proteins were transferred onto nitrocellulose membrane (Bio-Rad, South Africa) using a transfer buffer (Bio-Rad, South Africa) at 100 V for 1 h. The membrane was blocked with blocking buffer (Li-COR, Germany) for 2 h and thereafter incubated with primary monoclonal BACE rabbit antibody (Cell Signaling, USA) and monoclonal β-actin mouse antibody (Cell Signaling, USA) (1:1000 dilution in blocking buffer) overnight. After completion of incubation with primary antibodies, the membrane was washed three times with 0.1 M PBS-Tween for 10 min. The secondary antibodies IRDye 800CW goat anti-rabbit and IRDye 680RD goat anti-mouse (LI-COR, Germany) (1:10,000 dilution in blocking buffer) were then added to the membrane and incubated for 2 h at room temperature. Afterwards, the membrane was washed three times with 0.1 M PBS-Tween and washed once with PBS. Finally, the membrane was viewed on an Odyssey CLx LI-COR infrared fluorescence imaging system (Biosciences, Germany). The intensities of signals on the blots were normalised to β-actin in a single channel using the LI-COR Odyssey Image Studio software (LI-COR Biosciences).

Mouse Aβ1-42 ELISA

The concentration levels of Aβ1-42 were quantified using an ELISA kit (Elabscience Biotechnology Co., China). Dorsal hippocampal tissue was homogenised in phosphate-buffered saline (0.01 M, pH = 7.4). The tissue homogenate was centrifuged for 5 min at 5000 g. A volume of 100 µl of supernatant or Aβ1-42 standards were added to each well and then incubated for 90 min at 37 °C. After incubation, the liquid was removed and 100 µl of biotinylated antibody was added to each well and incubated at 37 °C for 1 h on an orbital shaker. After 1-h incubation, wells were washed three times with wash buffer and 100 µl of HRP-conjugated working solution was added to each well. The plate was then incubated for 30 min at 37 °C. The liquid was aspirated and wells were washed. Substrate solution was added to each well and then finally stop solution was added. Optical density of each well was measured using a micro-plate reader set to 450 nm as per the manufacturer’s protocol.

Immunohistochemistry

Mice were anaesthetised with an intraperitoneal injection of pentobarbital (100 mg/kg) and then transcardially perfused with physiological saline (0.9%) containing protease inhibitors followed by 4% paraformaldehyde before removal of brains. Whole brains were post-fixed in 4% paraformaldehyde for 24 h. All brains were then cryopreserved in 15% (w/v) and 30% (w/v) sucrose in PBS for 24 h at 4 °C. Brains were embedded in optimal cutting temperature compound and then frozen in liquid nitrogen. Coronal sections (30 μm) were cut using a cryostat (Leica, Germany). Frozen sections were air-dried at room temperature for 15 min and covered with 4% paraformaldehyde for 10 min. The sections were then rinsed with PBST (0.25% Triton-X100 in PBS) and blocked for 30 min at room temperature in 5% goat serum. After incubation with monoclonal mouse APP/β-amyloid antibody (NAB228) (Cell Signaling, USA) diluted in PBS, for 24 h at 4 °C, sections were washed with PBS and incubated with anti-mouse HRP-linked secondary antibody (Cell Signaling, USA) for 1 h at room temperature. The DAB substrate kit (Cell Signaling, USA) was used for detection. Sections were washed again and mounted. The images were captured using a microscope (Leica, Germany) and analysed using ImageJ software (National Institutes of Health, USA). A measure of amyloid load was determined as follows: Aβ deposits in each of five representative sections of the dorsal hippocampus of each of the four animals per group were counted, and an average Aβ deposits number per animal was recorded. This number of Aβ deposits occupying in the investigated dorsal hippocampal area was taken as a measure of hippocampal amyloid deposit load per mouse.

Statistical analysis

Data were analysed using GraphPad Prism version 5 software. The Shapiro–Wilk test was used to test the data for normality. The results are presented as mean ± standard error of the mean. The escape latencies in the Morris water maze were analysed with two-way repeated measures ANOVA with drug treatment as main factor and days as repeated measure, followed by Bonferroni post hoc comparisons to determine differences between groups. All other data were analysed by one-way ANOVA followed by Bonferroni post hoc test to determine differences between groups. Differences were considered statistically significant when p < 0.05.

Results

Morris water maze

We assessed spatial learning by analysing the time taken to find the hidden platform (escape latency) and subsequently looked at memory retention as indicated by the time spent in the target quadrant during the probe test. Repeated measures ANOVA showed a main effect of treatment on spatial learning [F (2, 108) = 8.834, p = 0.01]. Bonferroni post hoc comparisons showed no significant differences in escape latency between groups on day 1; however, on day 2, the escape latency of the nevirapine-treated group (n = 10) was significantly increased compared to control group (n = 10) (p < 0.05) or TDF (n = 10) (p < 0.01, Fig. 1A). On day 3 and 4, the latency was not significantly different between the groups; however, there is a trend towards an increased latency in mice treated with nevirapine. On day 5, the latency to escape of the nevirapine-treated group was again significantly increased compared to the control group (p < 0.01).

Fig. 1. The effect of chronic administration of TDF and nevirapine on spatial learning and memory in mice. (A) Learning in the Morris water maze, that is, time to locate the hidden platform. Nevirapine-treated mice showed significantly greater escape latencies in the hidden platform test compared to control mice (*p < 0.05; **p < 0.01) or TDF (##p < 0.01). (B) Probe trials to assess spatial memory retrieval, that is, time spent in the target quadrant in the Morris water maze. Nevirapine-treated mice spent significantly less time in the target quadrant compared to control group (**p < 0.01). Data are presented as means ± SEM (n = 10 per group).

In the probe test, one-way ANOVA data analysis showed a significant impairment of memory retention [F (2, 27) = 7.123, p = 0.0033]. Bonferroni post hoc test showed no significant differences were observed for the TDF-treated group compared to the control group (p > 0.05); however, a significant decrease in time spent by the nevirapine-treated group in the target quadrant compared to the control group (p < 0.01, Fig. 1B).

Lipid peroxidation (4-HNE) concentration

4-HNE is a cytotoxic lipid-derived aldehyde often used as an indicator of lipid peroxidation (Ayala et al., Reference Ayala, Muñoz and Argüelles2014). 4-HNE increase in the brain is also associated with increased Aβ production and is used as a key indicator of neurodegenerative pathologies (Di Domenico et al., Reference di Domenico, Tramutola and Butterfield2017). HNE concentration in hippocampal tissue was measured in all groups after 8 weeks of drug administration. One-way ANOVA showed a main effect of treatment on HNE concentrations [F (2, 15) = 11.83, p = 0.0008]. Bonferroni post hoc test showed a significantly higher concentration of HNE in the TDF-treated group (p < 0.05) and nevirapine-treated group (p < 0.001) compared to the control group (Fig. 2).

Fig. 2. Long-term administration of antiretroviral drugs, TDF and nevirapine, increased the concentration of 4-hydroxynonenal in the hippocampus. The TDF group (*p < 0.05) and the nevirapine group (***p < 0.001) had significantly higher levels than the control group. Data are presented as means ± SEM (n = 6 per group).

BACE1 expression

Hippocampus BACE1 expression was measured using Western blotting in all groups. One-way ANOVA revealed a significant difference in BACE1 expression in the hippocampus [F (2, 15) = 10.66, p = 0.0013]. Bonferroni post hoc test showed a significant increase in the expression of BACE1 in the nevirapine-treated group compared to control (p < 0.01) and TDF groups (p < 0.05, Fig. 3).

Fig. 3. Effect of TDF and nevirapine treatment on BACE1 expression in the hippocampus of mice. BACE1 expression was measured by Western blot and band intensity was normalized against β-actin. Results revealed a significant increase in BACE1 expression in the nevirapine-treated group when compared to the control group (**p < 0.01) and TDF (#p < 0.05) groups. Data are presented as means ± SEM (n = 6 per group).

Aβ1-42 concentration

The Aβ1-42 peptide is considered to be more neurotoxic than Aβ1-40 (Tamagno et al., Reference Tamagno, Guglielmotto, Monteleone and Tabaton2012); therefore, we thought that measuring the longer version of the peptide would be more relevant to our current study. One-way ANOVA showed a significant difference [F (2, 15) = 10.56, p = 0.0014] in the Aβ1-42 concentrations of the various groups of animals. Bonferroni post hoc test showed a significantly high Aβ1-42 concentration in the TDF (p < 0.05) and nevirapine-treated (p < 0.01) group compared to controls, Fig. 4.

Fig. 4. The effects of TDF and nevirapine treatment on amyloid-β 1-42 peptide concentration in the hippocampus. Data showed significantly higher levels of Aβ1-42 in the hippocampus of TDF-treated group compared to the control (*p < 0.05) and nevirapine group (**p < 0.01). Data are presented as means ± SEM (n = 6 per group).

Aβ deposition

The last objective of the present study was to determine, through immunohistochemical analysis, whether ARV drugs affect Aβ deposition in the hippocampus. One-way ANOVA showed a main effect of treatment on the number of Aβ deposits in the hippocampus [F (2, 09) = 5.957, p = 0.0255]. Bonferroni post hoc test showed a significant increase in the number of Aβ deposits in the nevirapine group compared to control mice (p < 0.05, Fig. 5).

Fig. 5. Effect of chronic administration of antiretroviral drugs, TDF and nevirapine, on Aβ deposition in the dorsal hippocampus (scale bar = 200  µm). Immunohistochemical staining for Aβ showed no significant increase in Aβ deposit number in the TDF-treated group compared to control animals. A significant increase in the number of Aβ deposition was observed in the nevirapine-treated group when compared to the control group (*p  <  0.05). Data are presented as means ± SEM (n  =  4 per group).

Discussion

Understanding the aetiology of HAND is critical for better management of HIV. Whether ARV drugs contribute to the development of HAND has not yet been clarified. This study subsequently set out to investigate whether chronic exposure to anti-HIV drugs (TDF and nevirapine) per se can negatively impact cognitive function. Since Aβ accumulation is among the neuropathologies observed in post-mortem brains of patients that were infected with HIV (Turner et al., Reference Turner, Chadwick, Horton, Simon, Jiang and Esposito2016; Solomon et al., Reference Solomon, de Girolami, Chettimada, Misra, Singer and Gabuzda2017), the present study tested the hypothesis that ARV drug treatment promote of Aβ and cognitive impairment. We further proposed that the accumulation of the Aβ is associated with drug-induced lipid peroxidation and the upregulation of the enzyme BACE1.

In the Morris water maze test, our findings showed that chronic exposure to nevirapine impaired spatial learning and memory. This finding is in line with a previous study by Romão et al. (Reference Romão, Lemos, Moreira, de Chaves, Moretti, Castro, Andrade, Boeck, Quevedo and Gavioli2011) which showed nevirapine treatment impaired recognition memory formation evaluated in the object recognition test in male CF-1 mice aged 60 days. These impairments were associated with lipid peroxidation (4-HNE), upregulation of BACE1 and increased Aβ in the hippocampus. Similarly, TDF treatment impaired learning and induced lipid peroxidation, but the effects were not as profound as nevirapine. Further, TDF did not affect BACE1expression and in Aβ deposition, despite an increase in Aβ. In overall, animals that were treated with TDF showed milder neurochemical changes than nevirapine-treated mice. These findings are consistent with previous in vitro studies showing that neuronal cell exposed to ARV had an increase Aβ production (Giunta et al., Reference Giunta, Ehrhart, Obregon, Lam, Le, Jin, Fernandez, Tan and Shytle2011; Brown et al., Reference Brown, Jin, Ferrell, Sadic, Obregon, Smith, Tan and Giunta2014b; Hui et al., Reference Hui, Ye, Soliman, Lakpa, Miller, Afghah, Geiger and Chen2019). Although the mechanisms by which ART promote Aβ are still unclear, a recent study by Hui et al. (Reference Hui, Ye, Soliman, Lakpa, Miller, Afghah, Geiger and Chen2019) found that nevirapine and TDF cause de-acidification endolysosomes and inhibited clearance of Aβ. Our findings are corroborate previous studies which showed that ARV treatment induce oxidative damage in mice (Zuena et al., Reference Zuena, Giuli, Venerosi Pesciolini, Tramutola, Ajmone-Cat, Cinque, Alema, Giovine, Peluso, Minghetti, Nicolai, Calamandrei and Casolini2013; de Oliveira et al., Reference de Oliveira, Damiani, Dias Rde, Romao and Andrade2014). We speculate that differences in effect between the two drugs can be attributed to (1) metabolisation of these drugs and (2) weak inhibition of DNA polymerase by TDF, as well as TDF previously reported limited penetration into the central nervous system (Birkus et al., Reference Birkus, Hajek, Kramata, Votruba, Holy and Otova2002; Ferrer & Rakhmanina, Reference Ferrer and Rakhmanina2013). On the other hand, the considerable deleterious effect of nevirapine on spatial learning and memory may be related to its ability to produce greater lipid peroxidation in the hippocampus. Lipid peroxidation indicated by 4-HNE in the present study signify a greater intensity of oxidative stress in the hippocampus leading to pathological conditions that may include cognitive dysfunction. This reasoning is plausible as several studies have associated with oxidative stress with brain ageing and neurodegenerative processes (Mariani et al., Reference Mariani, Polidori, Cherubini and Mecocci2005; Reddy et al., Reference Reddy, Reddy, Manczak, Calkins, Shirendeb and Mao2011; Montgomery et al., Reference Montgomery, Buttemer and Hulbert2012).

Studies suggest that NRTIs inhibit mitochondrial DNA polymerase-γ causing the depletion of mitochondrial DNA. This aberration in DNA quantity leads to excessive production of mitochondrial ROS which in turn may be responsible for oxidative damage to lipid, amino acid and DNA components of the cell (Bienstock & Copeland, Reference Bienstock and Copeland2004; Apostolova et al., Reference Apostolova, Blas-Garcia and Esplugues2011). Similarly, NNRTIs have shown to mediate cellular toxicity by inhibition of creatine kinase and cytochrome C complex IV activity in the brain (Apostolova et al., Reference Apostolova, Gomez-Sucerquia, Moran, Alvarez, Blas-Garcia and Esplugues2010; Streck et al., Reference Streck, Ferreira, Scaini, Rezin, Goncalves, Jeremias, Zugno, Ferreira, Moreira, Fochesato and Romao2011). Preclinical and clinical studies have linked NRTIs and NNRTIs exposure with cognitive dysfunction in HIV-1-infected individuals through mechanism involving oxidative stress (Akay et al., Reference Akay, Cooper, Odeleye, Jensen, White, Vassoler, Gannon, Mankowski, Dorsey and Buch2014a; Brown et al., Reference Brown, Jin, Ferrell, Sadic, Obregon, Smith, Tan and Giunta2014a; Sharma, Reference Sharma2014). It seems however that alternative downstream molecular mechanisms may be recruited by the different classes of ARVs resulting in the one drug (NNRTIs) showing greater iatrogenic damage than the other (NRTIs).

The aetiology of Alzheimer’s disease has been closely linked to oxidative stress and Aβ neurotoxicity (Hardy & Selkoe, Reference Hardy and Selkoe2002). BACE1 is regarded as a key enzyme in Aβ peptide synthesis (Vassar et al., Reference Vassar, Bennett, Babu-Khan, Kahn, Mendiaz, Denis, Teplow, Ross, Amarante, Loeloff, Luo, Fisher, Fuller, Edenson, Lile, Jarosinski, Biere, Curran, Burgess, Louis, Collins, Treanor, Rogers and Citron1999), and cleavage of APP by BACE1 is the initial step towards Aβ production. Different studies have shown that 4-HNE upregulates BACE1 in neuroblastoma cell lines and primary cortical cultures (Tamagno et al., Reference Tamagno, Parola, Bardini, Piccini, Borghi, Guglielmotto, Santoro, Davit, Danni, Smith, Perry and Tabaton2005, Reference Tamagno, Guglielmotto, Aragno, Borghi, Autelli, Giliberto, Muraca, Danni, Zhu, Smith, Perry, Jo, Mattson and Tabaton2008; de O’Leary et al., Reference de O’Leary, Obregon, Fernandez, Tan and Giunta2012), a finding supported by another study that showed elevated Aβ in wild-type mice following intracerebral infusion of 4-HNE (Arimon et al., Reference Arimon, Takeda, Post, Svirsky, Hyman and Berezovska2015). The link between lipid peroxidation, BACE1 expression and Aβ production has previously been established (Mouton-Liger et al., Reference Mouton-Liger, Paquet, Dumurgier, Bouras, Pradier, Gray and Hugon2012; Muche et al., Reference Muche, Arendt and Schliebs2017) (Fig. 6).

Fig. 6. Diagram representing the sequence of events on antiretroviral drugs promoting Aβ production. (1) Antiretroviral promote reactive oxygen species (ROS). (2) ROS induce lipid peroxidation. (3) BACE1 expression and activity increased by lipid peroxidation. (4) Amyloid peptide (Aβ) generated following the sequential cleavage of its precursor, the amyloid-β precursor protein (AβPP) by BACE1.

Further support for our postulate comes from histopathological studies that identified Aβ accumulation in the central nervous system of HIV-positive patients on ART (Esiri et al., Reference Esiri, Biddolph and Morris1998; Brew et al., Reference Brew, Pemberton, Blennow, Wallin and Hagberg2005; Green et al., Reference Green, Masliah, Vinters, Beizai, Moore and Achim2005). Our results, therefore, support evidence that ART may contribute to the formation of Aβ in the brains of HIV-infected persons and that the observed Aβ deposits may partially stem from ARV treatment. We have also previously shown that TDF and nevirapine upregulate neuroinflammatory cytokine and astrogliosis in vivo (Zulu et al., Reference Zulu, Simola, Mabandla and Daniels2018). As such our findings point to a possibility that ART may be one of the aetiological factors of HAND and suggest that it may be worthwhile to investigate whether the supplementation of ARVs with agents that reduce oxidative stress hold any benefit to HIV-infected individuals on ART with respect to the possible development of HAND. In conclusion, this study demonstrated that ARV drugs, TDF and nevirapine, induce lipid peroxidation. Also, nevirapine promotes Aβ by upregulating BACE1 and AβPP production in the hippocampus and subsequently cause spatial learning and memory impairments. Therefore, consideration should be given to HIV medication since it might be among the factors that contribute to the persistence of neurocognitive disorders in HIV-infected persons on cART.

Supplementary material

To view supplementary material for this article, please visit https://doi.org/10.1017/neu.2020.19

Authors contributions

SSZ, WMUD and MVM conceived of the presented idea.

SSZ and OA carried out the experiments.

SSZ wrote the manuscript with support from WMUD, OA and NS

NS, WMUD contributed to the interpretation of the results

Critical revision of the article was done by MVM and OA.

All authors discussed the results and contributed to the final manuscript.

Financial support

The authors wish to thank the National Research Foundation of South Africa and the College of Health Sciences from the University of KwaZulu-Natal for their financial support. NS wishes to thank the Italian Ministry of Foreign Affairs (MAE) for financial support.

Statement of interest

Authors declare no competing interests.

Ethical standards

The authors assert that all procedures contributing to this work comply with the ethical standards of the relevant national and institutional guides on the care and use of laboratory animals.

References

Akay, C, Cooper, M, Odeleye, A, Jensen, BK, White, MG, Vassoler, F, Gannon, PJ, Mankowski, J, Dorsey, JL and Buch, AM (2014a) Antiretroviral drugs induce oxidative stress and neuronal damage in the central nervous system. Journal of Neurovirology 20, 3953.CrossRefGoogle ScholarPubMed
Akay, C, Cooper, M, Odeleye, A, Jensen, BK, White, MG, Vassoler, F, Gannon, PJ, Mankowski, J, Dorsey, JL, Buch, AM, Cross, SA, Cook, DR, Pena, MM, Andersen, ES, Christofidou-Solomidou, M, Lindl, KA, Zink, MC, Clements, J, Pierce, RC, Kolson, DL and Jordan-Sciutto, KL (2014b) Antiretroviral drugs induce oxidative stress and neuronal damage in the central nervous system. Journal of Neurovirology 20, 3953.CrossRefGoogle ScholarPubMed
Allavena, C, Le Moal, G, Michau, C, Chiffoleau, A and Raffi, F (2006) Neuropsychiatric adverse events after switching from an antiretroviral regimen containing efavirenz without tenofovir to an efavirenz regimen containing tenofovir: a report of nine cases. Antiviral Therapy 11, 263265.Google Scholar
Amacher, DE (2014) Female gender as a susceptibility factor for drug-induced liver injury. Human & Experimental Toxicology 33, 928939.CrossRefGoogle ScholarPubMed
Andras, IE and Toborek, M (2013) Amyloid beta accumulation in HIV-1-infected brain: the role of the blood brain barrier. IUBMB Life 65, 4349.CrossRefGoogle ScholarPubMed
Apostolova, N, Blas-Garcia, A and Esplugues, JV (2011) Mitochondrial interference by anti-HIV drugs: mechanisms beyond Pol-gamma inhibition. Trends in Pharmacological Sciences 32, 715725.CrossRefGoogle ScholarPubMed
Apostolova, N, Gomez-Sucerquia, LJ, Moran, A, Alvarez, A, Blas-Garcia, A and Esplugues, JV (2010) Enhanced oxidative stress and increased mitochondrial mass during Efavirenz-induced apoptosis in human hepatic cells. British Journal of Pharmacology 160, 20692084.CrossRefGoogle ScholarPubMed
Arimon, M, Takeda, S, Post, KL, Svirsky, S, Hyman, BT and Berezovska, O (2015) Oxidative stress and lipid peroxidation are upstream of amyloid pathology. Neurobiology of Disease 84, 109119.CrossRefGoogle ScholarPubMed
Ayala, A, Muñoz, MF and Argüelles, S (2014) Lipid peroxidation: production, metabolism, and signaling mechanisms of malondialdehyde and 4-Hydroxy-2-Nonenal. Oxidative Medicine and Cellular Longevity 2014, 360438.CrossRefGoogle ScholarPubMed
Bienstock, RJ and Copeland, WC (2004) Molecular insights into NRTI inhibition and mitochondrial toxicity revealed from a structural model of the human mitochondrial DNA polymerase. Mitochondrion 4, 203213.CrossRefGoogle ScholarPubMed
Birkus, G, Hajek, M, Kramata, P, Votruba, I, Holy, A and Otova, B (2002) Tenofovir diphosphate is a poor substrate and a weak inhibitor of rat DNA polymerases alpha, delta, and epsilon*. Antimicrob Agents Chemother 46, 16101613.CrossRefGoogle Scholar
Boelen, E, Stassen, FR, van der Ven, AJ, Lemmens, MA, Steinbusch, HP, Bruggeman, CA, Schmitz, C and Steinbusch, HW (2007) Detection of amyloid beta aggregates in the brain of BALB/c mice after Chlamydia pneumoniae infection. Acta Neuropathologica 114, 255261.CrossRefGoogle ScholarPubMed
Brew, BJ, Pemberton, L, Blennow, K, Wallin, A and Hagberg, L (2005) CSF amyloid beta42 and tau levels correlate with AIDS dementia complex. Neurology 65, 14901492.CrossRefGoogle ScholarPubMed
Brown, LA, Jin, J, Ferrell, D, Sadic, E, Obregon, D, Smith, AJ, Tan, J and Giunta, B (2014a) Efavirenz promotes β-Secretase expression and increased Aβ 1-40, 42 via oxidative stress and reduced microglial phagocytosis: implications for HIV associated neurocognitive disorders (HAND). PLoS One 9, e95500.CrossRefGoogle Scholar
Brown, LAM., Jin, J, Ferrell, D, Sadic, E, Obregon, D, Smith, AJ., Tan, J and Giunta, B (2014b) Efavirenz promotes β-Secretase expression and increased Aβ1-40,42 via oxidative stress and reduced microglial phagocytosis: implications for HIV associated neurocognitive disorders (HAND). PLoS One 9, e95500.CrossRefGoogle Scholar
Bryant, AK, Ellis, RJ, Umlauf, A, Gouaux, B, Soontornniyomkij, V, Letendre, SL, Achim, CL, Masliah, E, Grant, I and Moore, DJ (2015) Antiretroviral therapy reduces neurodegeneration in HIV infection. AIDS 29, 323330.CrossRefGoogle ScholarPubMed
Ciccarelli, N, Fabbiani, M, di Giambenedetto, S, Fanti, I, Baldonero, E, Bracciale, L, Tamburrini, E, Cauda, R, de Luca, A and Silveri, MC (2011) Efavirenz associated with cognitive disorders in otherwise asymptomatic HIV-infected patients. Neurology 76, 14031409.CrossRefGoogle ScholarPubMed
Cote, HC, Magil, AB, Harris, M, Scarth, BJ, Gadawski, I, Wang, N, Yu, E, Yip, B, Zalunardo, N, Werb, R, Hogg, R, Harrigan, PR and Montaner, JS (2006) Exploring mitochondrial nephrotoxicity as a potential mechanism of kidney dysfunction among HIV-infected patients on highly active antiretroviral therapy. Antiviral Therapy 11, 7986.Google ScholarPubMed
Davies, S and Taylor, S (2013) Antiretroviral pharmacology. Medicine 41, 474478.CrossRefGoogle Scholar
de O’Leary, JC, Obregon, D, Fernandez, F, Tan, J and Giunta, B (2012) The impact of HAART on advanced brain aging: implications for mitochondrial dysfunction and APP processing. Journal of Antivirals and Antiretrovirals 10, 2.Google Scholar
de Oliveira, HM, Damiani, AP, Dias Rde, O, Romao, PR and Andrade, VM (2014) Effect of antiretroviral drugs on the DNA damage in mice. Environmental Toxicology and Pharmacology 37, 390395.CrossRefGoogle ScholarPubMed
di Domenico, F, Tramutola, A and Butterfield, DA (2017) Role of 4-hydroxy-2-nonenal (HNE) in the pathogenesis of alzheimer disease and other selected age-related neurodegenerative disorders. Free Radical Biology and Medicine 111, 253261.CrossRefGoogle ScholarPubMed
du Plessis, S, Perez, A, Fouche, JP, Phillips, N, Joska, JA, Vink, M, Myer, L, Zar, HJ, Stein, DJ and Hoare, J (2019) Efavirenz is associated with altered fronto-striatal function in HIV+ adolescents. Journal of Neurovirology 25, 783791.CrossRefGoogle ScholarPubMed
Ene, L, Duiculescu, D and Ruta, SM (2011) How much do antiretroviral drugs penetrate into the central nervous system?. Journal of Medicine and Life 4, 432439.Google ScholarPubMed
Esiri, MM, Biddolph, SC and Morris, CS (1998) Prevalence of Alzheimer plaques in AIDS. Journal of Neurology, Neurosurgery & Psychiatry 65, 2933.CrossRefGoogle Scholar
Ferrer, K and Rakhmanina, N (2013) Neuropsychiatric effects of tenofovir in comparison with other antiretroviral drugs. Neurobehavioral HIV Medicine 5, 110.Google Scholar
Gannon, PJ (2014) Assessing the Contribution Antiretroviral Therapy to Neuronal Damage and Death as a Mediator of Cognitive Decline in HIV-Associated Neurocognitive Disorders. Publicly Accessible Penn Dissertations, 1280.Google Scholar
Giunta, B, Ehrhart, J, Obregon, DF, Lam, L, Le, L, Jin, J, Fernandez, F, Tan, J and Shytle, RD (2011) Antiretroviral medications disrupt microglial phagocytosis of beta-amyloid and increase its production by neurons: implications for HIV-associated neurocognitive disorders. Molecular Brain 4, 23.CrossRefGoogle ScholarPubMed
Green, DA, Masliah, E, Vinters, HV, Beizai, P, Moore, DJ and Achim, CL (2005) Brain deposition of beta-amyloid is a common pathologic feature in HIV positive patients. AIDS 19, 407411.CrossRefGoogle ScholarPubMed
Hardy, J and Selkoe, DJ (2002) The amyloid hypothesis of Alzheimer’s disease: progress and problems on the road to therapeutics. Science 297, 353356.CrossRefGoogle ScholarPubMed
Howlett, DR, Richardson, JC, Austin, A, Parsons, AA, Bate, ST, Davies, DC and Gonzalez, MI (2004) Cognitive correlates of Aβ deposition in male and female mice bearing amyloid precursor protein and presenilin-1 mutant transgenes. Brain Research 1017, 130136.CrossRefGoogle ScholarPubMed
Hui, L, Ye, Y, Soliman, ML, Lakpa, KL, Miller, NM, Afghah, Z, Geiger, JD and Chen, X (2019) Antiretroviral drugs promote amyloidogenesis by De-acidifying endolysosomes. Journal of Neuroimmune Pharmacology, 110.CrossRefGoogle Scholar
Kohler, JJ, Hosseini, SH, Hoying-Brandt, A, Green, E, Johnson, DM, Russ, R, Tran, D, Raper, CM, Santoianni, R and Lewis, W (2009) Tenofovir renal toxicity targets mitochondria of renal proximal tubules. Laboratory Investigation 89, 513519.CrossRefGoogle ScholarPubMed
Kruger, NJ (2009) The Bradford method for protein quantitation. The Protein Protocols Handbook 15, 1724.CrossRefGoogle Scholar
Lebrecht, D, Venhoff, AC, Kirschner, J, Wiech, T, Venhoff, N and Walker, UA (2009) Mitochondrial tubulopathy in tenofovir disoproxil fumarate-treated rats. Journal of Acquired Immune Deficiency Syndromes 51, 258263.CrossRefGoogle ScholarPubMed
Little, CS, Joyce, TA, Hammond, CJ, Matta, H, Cahn, D, Appelt, DM and Balin, BJ (2014) Detection of bacterial antigens and Alzheimer’s disease-like pathology in the central nervous system of BALB/c mice following intranasal infection with a laboratory isolate of Chlamydia pneumoniae. Frontiers in Aging Neuroscience 6, 304.CrossRefGoogle ScholarPubMed
Mariani, E, Polidori, M, Cherubini, A and Mecocci, P (2005) Oxidative stress in brain aging, neurodegenerative and vascular diseases: an overview. Journal of Chromatography B 827, 6575.CrossRefGoogle ScholarPubMed
Mcarthur, JC, Steiner, J, Sacktor, N and Nath, A (2010) Human immunodeficiency virus-associated neurocognitive disorders: mind the gap. Annals of Neurology 67, 699714.Google ScholarPubMed
Montgomery, MK, Buttemer, WA and Hulbert, AJ (2012) Does the oxidative stress theory of aging explain longevity differences in birds? II. Antioxidant systems and oxidative damage. Experimental Gerontology 47, 211222.CrossRefGoogle ScholarPubMed
Mouton-Liger, F, Paquet, C, Dumurgier, J, Bouras, C, Pradier, L, Gray, F and Hugon, J (2012) Oxidative stress increases BACE1 protein levels through activation of the PKR-eIF2alpha pathway. Biochimica et Biophysica Acta 1822, 885896.CrossRefGoogle ScholarPubMed
Muche, A, Arendt, T and Schliebs, R (2017) Oxidative stress affects processing of amyloid precursor protein in vascular endothelial cells. PLoS One 12, e0178127.CrossRefGoogle ScholarPubMed
Nooka, S and Ghorpade, A (2017) HIV-1-associated inflammation and antiretroviral therapy regulate astrocyte endoplasmic reticulum stress responses. Cell Death Discovery 3, 1706117061.CrossRefGoogle ScholarPubMed
Ortega, M and Ances, BM (2014) Role of HIV in amyloid metabolism. Journal of Neuroimmune Pharmacology 9, 483491.CrossRefGoogle ScholarPubMed
Reddy, PH, Reddy, TP, Manczak, M, Calkins, MJ, Shirendeb, U and Mao, P (2011) Dynamin-related protein 1 and mitochondrial fragmentation in neurodegenerative diseases. Brain Research Reviews 67, 103118.CrossRefGoogle ScholarPubMed
Romão, PRT, Lemos, JC, Moreira, J, de Chaves, G, Moretti, M, Castro, AA, Andrade, VM, Boeck, CR, Quevedo, J and Gavioli, EC (2011) Anti-HIV drugs nevirapine and efavirenz affect anxiety-related behavior and cognitive performance in mice. Neurotoxicity Research 19, 7380.CrossRefGoogle ScholarPubMed
Sacktor, N, Lyles, RH, Skolasky, R, Kleeberger, C, Selnes, OA, Miller, EN, Becker, JT, Cohen, B and Mcarthur, JC (2001) HIV-associated neurologic disease incidence changes: multicenter AIDS cohort study, 1990–1998. Neurology 56, 257260.CrossRefGoogle ScholarPubMed
Schank, JC and Mcclintock, MK (1992) A coupled-oscillator model of ovarian-cycle synchrony among female rats. Journal of Theoretical Biology 157, 317362.CrossRefGoogle ScholarPubMed
Shah, A, Gangwani, MR, Chaudhari, NS, Glazyrin, A, Bhat, HK and Kumar, A (2016) Neurotoxicity in the post-HAART Era: caution for the antiretroviral therapeutics. Neurotoxicity Research 30, 677697.CrossRefGoogle ScholarPubMed
Sharma, B (2014) Oxidative stress in HIV patients receiving antiretroviral therapy. Current HIV Research 12, 1321.CrossRefGoogle ScholarPubMed
Solomon, IH, de Girolami, U, Chettimada, S, Misra, V, Singer, EJ and Gabuzda, D (2017) Brain and liver pathology, amyloid deposition, and interferon responses among older HIV-positive patients in the late HAART era. BMC Infectious Diseases 17, 151.Google ScholarPubMed
Streck, EL, Ferreira, GK, Scaini, G, Rezin, GT, Goncalves, CL, Jeremias, IC, Zugno, AI, Ferreira, GC, Moreira, J, Fochesato, CM and Romao, PR (2011) Non-nucleoside reverse transcriptase inhibitors efavirenz and nevirapine inhibit cytochrome C oxidase in mouse brain regions. Neurochemical Research 36, 962966.CrossRefGoogle ScholarPubMed
Tamagno, E, Guglielmotto, M, Aragno, M, Borghi, R, Autelli, R, Giliberto, L, Muraca, G, Danni, O, Zhu, X, Smith, MA, Perry, G, Jo, DG, Mattson, MP and Tabaton, M (2008) Oxidative stress activates a positive feedback between the gamma- and beta-secretase cleavages of the beta-amyloid precursor protein. Journal of Neurochemistry 104, 683695.Google ScholarPubMed
Tamagno, E, Guglielmotto, M, Monteleone, D and Tabaton, M (2012) Amyloid-β production: major link between oxidative stress and BACE1. Neurotoxicity Research 22, 208219.CrossRefGoogle ScholarPubMed
Tamagno, E, Parola, M, Bardini, P, Piccini, A, Borghi, R, Guglielmotto, M, Santoro, G, Davit, A, Danni, O, Smith, MA, Perry, G and Tabaton, M (2005) Beta-site APP cleaving enzyme up-regulation induced by 4-hydroxynonenal is mediated by stress-activated protein kinases pathways. Journal of Neurochemistry 92, 628636.Google ScholarPubMed
Turner, RS, Chadwick, M, Horton, WA, Simon, GL, Jiang, X and Esposito, G (2016) An individual with human immunodeficiency virus, dementia, and central nervous system amyloid deposition. Alzheimer’s & Dementia: Diagnosis, Assessment & Disease Monitoring 4, 15.Google ScholarPubMed
Vassar, R, Bennett, BD, Babu-Khan, S, Kahn, S, Mendiaz, EA, Denis, P, Teplow, DB, Ross, S, Amarante, P, Loeloff, R, Luo, Y, Fisher, S, Fuller, J, Edenson, S, Lile, J, Jarosinski, MA, Biere, AL, Curran, E, Burgess, T, Louis, JC, Collins, F, Treanor, J, Rogers, G and Citron, M (1999) Beta-secretase cleavage of Alzheimer’s amyloid precursor protein by the transmembrane aspartic protease BACE. Science 286, 735741.CrossRefGoogle ScholarPubMed
Vorhees, CV and Williams, MT (2006) Morris water maze: procedures for assessing spatial and related forms of learning and memory. Nature Protocols 1, 848858.CrossRefGoogle ScholarPubMed
Wang, J, Tanila, H, Puoliväli, J, Kadish, I and Van Groen, T (2003) Gender differences in the amount and deposition of amyloidβ in APPswe and PS1 double transgenic mice. Neurobiology of Disease 14, 318327.CrossRefGoogle ScholarPubMed
Wise, ME, Mistry, K and Reid, S (2002) Drug points: neuropsychiatric complications of nevirapine treatment. Bmj 324, 879.CrossRefGoogle ScholarPubMed
Woods, SP, Moore, DJ, Weber, E and Grant, I (2009) Cognitive neuropsychology of HIV-associated neurocognitive disorders. Neuropsychology Review 19, 152168.CrossRefGoogle ScholarPubMed
Zopf, Y, Rabe, C, Neubert, A, Gaßmann, KG, Rascher, W, Hahn, EG, Brune, K and Dormann, H (2008) Women encounter ADRs more often than do men. European Journal of Clinical Pharmacology 64, 999.CrossRefGoogle ScholarPubMed
Zuena, AR, Giuli, C, Venerosi Pesciolini, A, Tramutola, A, Ajmone-Cat, MA, Cinque, C, Alema, GS, Giovine, A, Peluso, G, Minghetti, L, Nicolai, R, Calamandrei, G and Casolini, P (2013) Transplacental exposure to AZT induces adverse neurochemical and behavioral effects in a mouse model: protection by L-acetylcarnitine. PLoS One 8, e55753.CrossRefGoogle Scholar
Zulu, SS, Simola, N, Mabandla, MV and Daniels, WMU. (2018) Effect of long-term administration of antiretroviral drugs (Tenofovir and Nevirapine) on neuroinflammation and neuroplasticity in mouse hippocampi. Journal of Chemical Neuroanatomy 94, 8692.CrossRefGoogle ScholarPubMed
Figure 0

Fig. 1. The effect of chronic administration of TDF and nevirapine on spatial learning and memory in mice. (A) Learning in the Morris water maze, that is, time to locate the hidden platform. Nevirapine-treated mice showed significantly greater escape latencies in the hidden platform test compared to control mice (*p < 0.05; **p < 0.01) or TDF (##p < 0.01). (B) Probe trials to assess spatial memory retrieval, that is, time spent in the target quadrant in the Morris water maze. Nevirapine-treated mice spent significantly less time in the target quadrant compared to control group (**p < 0.01). Data are presented as means ± SEM (n = 10 per group).

Figure 1

Fig. 2. Long-term administration of antiretroviral drugs, TDF and nevirapine, increased the concentration of 4-hydroxynonenal in the hippocampus. The TDF group (*p < 0.05) and the nevirapine group (***p < 0.001) had significantly higher levels than the control group. Data are presented as means ± SEM (n = 6 per group).

Figure 2

Fig. 3. Effect of TDF and nevirapine treatment on BACE1 expression in the hippocampus of mice. BACE1 expression was measured by Western blot and band intensity was normalized against β-actin. Results revealed a significant increase in BACE1 expression in the nevirapine-treated group when compared to the control group (**p < 0.01) and TDF (#p < 0.05) groups. Data are presented as means ± SEM (n = 6 per group).

Figure 3

Fig. 4. The effects of TDF and nevirapine treatment on amyloid-β 1-42 peptide concentration in the hippocampus. Data showed significantly higher levels of Aβ1-42 in the hippocampus of TDF-treated group compared to the control (*p < 0.05) and nevirapine group (**p < 0.01). Data are presented as means ± SEM (n = 6 per group).

Figure 4

Fig. 5. Effect of chronic administration of antiretroviral drugs, TDF and nevirapine, on Aβ deposition in the dorsal hippocampus (scale bar = 200  µm). Immunohistochemical staining for Aβ showed no significant increase in Aβ deposit number in the TDF-treated group compared to control animals. A significant increase in the number of Aβ deposition was observed in the nevirapine-treated group when compared to the control group (*p  <  0.05). Data are presented as means ± SEM (n  =  4 per group).

Figure 5

Fig. 6. Diagram representing the sequence of events on antiretroviral drugs promoting Aβ production. (1) Antiretroviral promote reactive oxygen species (ROS). (2) ROS induce lipid peroxidation. (3) BACE1 expression and activity increased by lipid peroxidation. (4) Amyloid peptide (Aβ) generated following the sequential cleavage of its precursor, the amyloid-β precursor protein (AβPP) by BACE1.

Supplementary material: File

Zulu et al. supplementary material

Figure S1

Download Zulu et al. supplementary material(File)
File 20.9 KB