Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Nov 15.
Published in final edited form as: Brain Res. 2019 Aug 21;1723:146398. doi: 10.1016/j.brainres.2019.146398

Dopaminergic impact of cART and anti-depressants on HIV neuropathogenesis in older adults

Stephanie M Matt 1, Peter J Gaskill 1,*
PMCID: PMC6766423  NIHMSID: NIHMS1539148  PMID: 31442412

Abstract

The success of combination antiretroviral therapy (cART) has transformed HIV infection into a chronic condition, resulting in an increase in the number of older, cART-treated adults living with HIV. This has increased the incidence of age-related, non-AIDS comorbidities in this population. One of the most common comorbidities is depression, which is also associated with cognitive impairment and a number of neuropathologies. In older people living with HIV, treating these overlapping disorders is complex, often creating pill burden or adverse drug-drug interactions that can exacerbate these neurologic disorders. Depression, NeuroHIV and many of the neuropsychiatric therapeutics used to treat them impact the dopaminergic system, suggesting that dopaminergic dysfunction may be a common factor in the development of these disorders. Further, changes in dopamine can influence the development of inflammation and the regulation of immune function, which are also implicated in the progression of NeuroHIV and depression. Little is known about the optimal clinical management of drug-drug interactions between cART drugs and antidepressants, particularly in regard to dopamine in older people living with HIV. This review will discuss those interactions, first examining the etiology of NeuroHIV and depression in older adults, then discussing the interrelated effects of dopamine and inflammation on these disorders, and finally reviewing the activity and interactions of cART drugs and antidepressants on each of these factors. Developing better strategies to manage these comorbidities is critical to the health of the aging, HIV-infected population, as the older population may be particularly vulnerable to drug-drug interactions affecting dopamine.

Keywords: Dopamine, NeuroHIV, Aging, Depression, Combination antiretroviral therapy, Antidepressants

1. Introduction

The use of combination antiretroviral therapy (cART) has greatly improved the lifespan and quality of life of HIV-infected individuals (Teeraananchai, 2017), transforming HIV infection into a chronic condition. However, the HIV-infected population is aging (Burgess et al., 2015; Negredo, 2017; Wing, 2016), increasing the disease burden as more infected individuals develop age-related impairments. The impact of chronic HIV infection and long-term cART on non-AIDS related comorbidities is not clear, but the increase in these comorbidities in older individuals living with HIV suggests a deleterious effect (Bhavan et al., 2008; Deeks, 2009; Simone and Appelbaum, 2008). Many of these comorbidities are neuropsychiatric disorders, among the most common of which is depression. The incidence of depression is between 20 and 70% of HIV-infected individuals (Campos et al., 2010; Savetsky, 2001), is the same in cART-naive and cART-treated populations (Bhatia and Munjal, 2014; Hobkirk, 2015; Pinheiro, 2016; Wang, 2018), and in both groups depression substantively worsens disease prognosis. Individuals over age 50 living with HIV show an increased risk for depression (Cahill and Valadéz, 2013; Milanini, 2017), and this disorder is commonly comorbid with cognitive impairment and neuropathology (Havlik et al., 2011; Arseniou et al., 2014). Treatment of these disorders is complex, often creating pill burden or deleterious drug-drug interactions, leading to problems with adherence and medication errors that can exacerbate disease symptoms and diminish quality of life.

Thus, developing better strategies to manage these comorbidities is critical to the health of the aging, HIV-infected population. Even with frequent co-administration of cART and antidepressants in older, HIV-infected patients, little information exists regarding the optimal clinical management of the interactions of these drugs within this population. Many of these drugs affect the dopaminergic system, and dopaminergic dysfunction is associated with the progression of both HIV and depression (Belujon and Grace, 2017; Nolan and Gaskill, 2018). Recent data suggest that dopamine may be an important regulator of inflammation (Nolan, 2018; Yan, 2015), which is also involved in the development of depression and HIV infection of the central nervous system (CNS), suggesting that dysregulated dopamine could exacerbate both of these diseases. Taken with the increase in dopamine-altering medications among older individuals, these data suggest that an older population may be particularly vulnerable to drug-drug interactions affecting CNS dopamine (see Fig. 1). To address this, we will first review the pathogenesis of NeuroHIV in the aging, HIV-infected population, focusing on the interaction with comorbid depression, and the issues surrounding its diagnosis and treatment. We will then discuss the overlapping impact of inflammation and dopamine on the etiology of depression and NeuroHIV, and how cART drugs and antidepressants alter these disorders. A more comprehensive understanding of the dopaminergic effects of cART-antidepressant crosstalk is important for the current HIV epidemic, as it could provide useful contraindication data, initiate drug repurposing, and spur the development of new therapeutic approaches for delaying or reducing HIV age-related comorbidities.

Fig. 1.

Fig. 1.

The Role of Dopamine in Mediating Antidepressant/cART Interactions in Older HIV-infected Individuals with Depression. This figure summarizes the interactions between dopamine and depression on aging, HIV-infected individuals with enhanced inflammation, demonstrating that cART drugs and antidepressants show significant influence on these factors. Synergistic effects of aging and HIV create a cycle of chronic inflammation that could result in significant age-related complications and decreased survival in HIV-infected older adults, relative to uninfected individuals. One of the most prevalent inflammation-mediated comorbidities in HIV-infected older adults is depression, which often necessitates complex treatment regimens of antidepressants in conjunction with cART drugs. This can cause pill burden, medication non-adherence or adverse drug-drug interactions that can exacerbate neuropathogenesis. Depression, NeuroHIV and many of the therapeutics used to treat them impact the dopaminergic system, suggesting that dopaminergic dysfunction may be a common factor in the development of these disorders. Further, changes in dopamine can influence the develop of inflammation and the regulation of immune function, which are also implicated in the development of NeuroHIV and depression.

2. Aging with HIV

Between 2015 and 2050, the proportion of people over 60 years old will nearly double from 12% to 22% (WHO, 2018). This trend is also prevalent in people living with HIV (Smit, 2015), as access to cART results in significantly longer life spans (High, 2012), transforming HIV from a terminal illness into a chronic disease. In HIV literature, individuals over the age of 50 are generally considered older adults (Blanco, 2012). Globally, more than 10% of the population living with HIV is aged 50 or older, and in higher income countries with better access to cART, this population makes up over 30% of infected individuals (UNAIDS, 2013.). Not only is the HIV-infected population aging, but older people represent an increasingly higher proportion of new HIV infections. In the US, around 18% of newly diagnosed patients are over the age of 50 years, and increasingly patients initially diagnosed at older ages are also diagnosed with a more advanced HIV infection (Tavoschi, 2017; Prevention, C.f.D.C.a., 2016). This may be due, in part, to a lack of awareness or education, as many older people consider HIV to be a “young person’s disease”, and there is less of a push for frequent disease screening in the 50+ population (Emlet, 2006; Sankar, 2011). However, immune function in older individuals is not as robust, and therefore the advanced disease presentation in this population may also be due to immunologic factors.

Older adults with HIV display more comorbidities and age-related conditions at a younger age than in the non-HIV population (Guaraldi, 2015; Vance, 2011), including many HIV-associated non-AIDS conditions that are becoming more frequent (Spano, 2016). Indeed, non-AIDS-related mortality is now more common than AIDS-related mortality as the major cause of death in the HIV-infected population (Alejos, 2014; Lifson, 2008). These conditions include diabetes (Abebe, 2016), hyperlipidemia (Lo, 2011), cardiovascular disease (Triant, 2013), and kidney disease (Maciel, 2018), but one of the largest problems associated with HIV infection in the aging population is neuropsychiatric diseases such as depression, anxiety and substance abuse (Watkins and Treisman, 2012).

Management of HIV in combination with these neuropsychiatric comorbidities is complicated by polypharmacy, drug–drug interactions and toxicity due to altered drug metabolism (Simone and Appelbaum, 2008; Greene, 2014). Older HIV-infected patients tend to use a higher number of medications at the same time (Marzolini, 2011), and one study found that 90% of patients over 50 had at least one concomitant medication and nearly 35% had three or more. Further, the same study found that age and duration of HIV infection were significantly associated with the number of concomitant medications (Serrao, 2019). Complex treatments with a high number of drugs leads to pill burden and is associated with a greater likelihood of medication errors and lack of adherence (Edelman, 2013). Polypharmacy is associated with an increase in inflammation and neuropsychiatric problems, such as depressive symptoms, in non-HIV-infected older adults (Ersoy and Engin, 2018), suggesting this association may also be present in the HlV-infected population. It has been estimated that nearly half of HIV-infected people aged 50 or older may be receiving concomitant drugs that are potentially prone to significant interactions with antiretroviral therapy, especially when the antiretroviral regimen includes boosted antiretroviral drugs (Marzolini, 2011). Historically, comparatively few HIV research studies specifically examined older adults, and these were often underrepresented, so relatively little information is available about optimal management for the older HIV population with neuropsychiatric conditions.

2.1. NeuroHIV in the aging population

The primary targets for HIV infection in the CNS are myeloid cells such as microglia and perivascular macrophages (Avalos, 2017.; Mallard and Williams, 2018; Rappaport and Volsky, 2015; Navia, 1986). Infection of the CNS occurs rapidly after initial infection in almost all individuals (Valcour, 2012; Davis, 1992), with HIV-infected monocytes crossing the blood brain barrier and differentiating into macrophages, which then disseminate virus throughout myeloid populations (Williams, 2014). Once infected, myeloid cells can produce new virus, act as viral reservoirs, and release neurotoxic factors that drive HIV neuropathology (Rappaport and Volsky, 2015; Williams and Hickey, 2002). Recent studies suggest that T-cells may also contribute to HIV infection of the CNS and act as reservoirs in this compartment, although this remains unclear (Honeycutt, 2018; Joseph, 2018; Spudich, 2019). Further, a number of studies suggest that astrocytes may be a target for HIV infection in the CNS (Churchill, 2009; Liu, 2004; Eugenin, 2011; Li, 2011; Reynolds, 2006; Carroll-Anzinger and Al-Harthi, 2006), but the importance of productive astrocyte infection in vivo, and how this influences the development of CNS infection, remains controversial (Russell, 2017; Ko, 2019; Al-Harthi and Nath, 2019). If infected, astrocytes could act as reservoirs, or amplify the spread neurotoxic signals (Eugenin and Berman, 2007; Eugenin and Berman, 2013; Carroll-Anzinger, 2007; Li et al., 2016), suggesting further examination of this cell type is needed to fully understand the development of CNS disease. However, for the purpose of this review, we will focus our discussion on myeloid cells as they are the most well characterized HIV targets in the CNS.

Prior to cART, this resulted in significant neuropathology, including microglial activation and the formation of multinucleated giant cells, reactive astrogliosis, inflammation, myelin loss, and regional atrophy, particularly in dopaminergic brain regions (Navia, 1986; Berger and Nath, 1997). While cART has greatly reduced and shifted these effects, more subtle forms of neuropathology are still seen in most infected individuals (Gelman, 2015). In the current era, cART-mediated suppression of viral replication has altered the severity, but not the prevalence, of HIV-associated neurocognitive disorders (HAND). Approximately 50% of all HIV-infected individuals still have some degree of neurological impairment, including, but not limited to, problems in attention, working memory, executive function, and processing speed, as well as behavioral changes and problems with motor coordination (Farhadian et al., 2017; Saylor, 2016). This constellation of symptoms, along with the underlying neuropathology, is known as NeuroHIV. The prevalence of these conditions in virally-suppressed individuals suggests that they are not caused directly by viral replication but are due to ancillary effects such as chronic neuroinflammation and subsequent disruptions in neurotransmission (Gannon et al., 2011; Tavazzi, 2014; Yuan, 2013; Yadav and Collman, 2009; Spudich, 2011; Anthony, 2005; Spudich, 2016). In older individuals, these effects could be compounded by age-associated inflammation, or “inflammaging,” which is a major risk factor for both morbidity and mortality in non-infected older adults (Franceschi and Campisi, 2014; Franceschi, 2007). Infection with HIV induces chronic immune activation similar to that seen healthy, aging adults, suggesting that aging and HIV synergistically enhance inflammation, particularly in the innate immune system (Zapata and Shaw, 2014; Martin, 2013). This could result in significant age-related complications and decreased survival in HIV-infected older adults, relative to uninfected individuals (Deeks, 2011; Grund, 2016).

The specific neuropathology present in older adults living with HIV is not well defined, but it is clear that these individuals are at greater risk for neurocognitive impairment, regardless of the duration of infection (Valcour, et al., 2004; Wendelken and Valcour, 2012). Advanced age increases susceptibility to more severe forms of impairment (Cañizares et al., 2014), and older HIV-infected individuals show an increased incidence of neuropathy (Watters, 2004) and dementia, with the risk for dementia being increased by more than three-fold for HIV-infected patients older than 50 years old compared to younger HIV-infected patients (Valcour, 2004). Older populations also show exacerbated extrapyramidal motor impairment, often associated with basal ganglia dysfunction and loss of dopamine, regardless of cART (Valcour, 2008; Tierney, 2019), and a greater neurocognitive impact in response to aging (Goodkin, 2017). Brain atrophy is increased with HIV, and older, cART-treated HIV-infected individuals show more rapid and progressive brain atrophy relative to healthy populations (Becker, 2011; Clifford, 2017).

Additionally, neuroimaging in HIV-infected individuals, particularly among older adults, shows elevations in abnormal white matter (Gongvatana, 2011), and metabolic abnormalities associated with a decline in neural efficiency (Ernst, 2009; Chang, 2013). These changes may be a reflection of the accelerated deterioration of dopaminergic circuits seen in HIV-infected individuals (Ipser, 2015; Anderson, 2016; Janssen, 2017; Ann, 2016), as well as in the specific sensitivity of dopaminergic neurons to HIV-associated neurotoxicity (Agrawal, 2010; Lopez, 1999; Hu, 2009). In addition, HIV itself may accelerate the aging process, as the increased rate of chronic comorbidities, along with persistent increased inflammatory markers, other senescent immune changes and frailty are present at younger ages compared to the non-HIV population (Deeks, 2009; Pathai, 2014). A recent study also showed that increased brain aging, predicted using neuroimaging, was related to cognitive deficits, despite effective viral suppression (Cole, 2017). However, the concept of accelerated aging in HIV-infected individuals is still controversial. While there is a higher occurrence of comorbidities in the HIV population for any given age, the rate of comorbidities does not increase with the amount of time a person is infected with HIV, suggesting that HIV does not appear to accelerate their occurrence over time (Rasmussen, 2015; Althoff, 2015). While these and other data suggest the cumulative effects of aging and HIV on brain function are substantial, the specific impact of these changes on the etiology of neuropsychiatric comorbidities is not clear.

2.2. NeuroHIV and comorbid depression in the aging population

Globally, major depressive disorder (MDD) is one of the most common neuropsychiatric disorders. This disorder is characterized by depressed mood, loss of interest in most activities, appetite and sleep disturbances, feelings of worthlessness and guilt, as well as suicidal thoughts and ideation (Richards, 2011). For HIV-infected individuals, depression is the most prevalent neuropsychiatric condition (Bhatia and Munjal, 2014; Rabkin, 2008), with 20–70% of people living with HIV showing depressive symptoms or MDD (Campos et al., 2010; Savetsky, 2001; Bing, 2001). Further, the severity of depression in people living with HIV is worse compared with the general population (Milanini, 2017; Do, 2014; Pence, 2018). The specific interaction between depression and aging with HIV is not well understood, but older HIV patients have an increased risk of depression compared with younger infected individuals (Hinkin, 2001; Grov, 2010), and the Research on Older Adults with HIV study found individuals with depression had a higher disease burden and an average of three other diseases (Havlik et al., 2011).

This same study also found significant correlations between Depression Scale scores and cognitive impairment in similar domains as non-infected individuals (Havlik et al., 2011), and other research shows around 33% of HIV-infected patients have deficits in subjective memory closely associated with depression and anxiety (Herrmann, 2019). Functional alterations of the medial prefrontal cortex and limbic structures such as the amygdala, hippocampus, parahippocampal cortex, and basal ganglia contribute to cognitive impairments in depression (Drevets, 2000; Sheline, 2000). These functional brain alterations overlap in part with the HIV-associated alterations in brain circuitry (Plessis, 2014), indicating HIV infection and depression could exacerbate each other, acting through common mechanisms to cause to behavioral and functional changes in these individuals.

However, the connection between depression and cognitive impairment in older people living with HIV is not clear and remains controversial in the cART era (Milanini, 2017). Studies in the Hawaii Aging with HIV cohort indicate that depression is significantly correlated with impaired neuropsychological performance in younger but not older individuals (Shimizu, 2011), and other research suggests that depression and cognitive impairment should be examined as independent processes (Cysique, 2007; Cole, 2007). For instance, although basal ganglia atrophy has been associated with both HIV infection and depression, no correlation was seen between basal ganglia atrophy and depression in HIV-infected individuals (Davison, 1997). Some studies have also noted gender- or ethnicity- specific impacts of depression in aging HIV populations (Brooks, 2012; Rubtsova, 2017). For example, unlike HIV-infected men, the depression scores of women with HIV increase yearly starting at age 40 (Aljassem, 2016). Another study found that with regards to the severity of depression, having a partner was protective for men as they aged, but not for women (Swendeman, 2018). Research into the links between NeuroHIV and depression in the aging population are relatively limited, and more data studying specific populations is urgently needed to fully understand these effects in this rapidly growing population.

Depression has been shown to be a predictor, cause and result of HIV disease progression. Depressed individuals living with HIV show increased plasma viral loads and rates of CD4+ T-cell loss, altered immune responses, and increased levels of inflammatory cytokines in both plasma and the CNS (Leserman, 2003; Rivera-Rivera, 2016). This leads to poorer viral suppression and cognitive impairment (Gonzalez, 2011), as well as more rapid HIV disease progression and higher mortality rates including higher rates of suicide (Ickovics, 2001; Dabaghzadeh, 2015). Depressive symptoms also reduce adherence to cART, and increase the risk of substance abuse, risky sexual behaviors and feelings of hopelessness (Hobkirk, 2015; Schuster et al., 2012). All of these effects significantly worsen individual clinical outcomes and promote the spread of HIV, which could be exacerbated in older individuals, making effective diagnosis and treatment of depression in this population a critical public health issue.

A major challenge in the current era is differentiating cognitive symptoms of depression from NeuroHIV, considering that in a high percentage of patients, the two disorders coexist. Depression may be an early manifestation of HIV-associated dementia (HAD), and depressive symptoms arising from HIV neurodegeneration may be associated with a risk of developing or worsening of cognitive impairment or HAD (Gibbie, 2006; Nanni, 2015; Castellon, 2006). Indeed, patients with cognitive decline and depression have significantly more functional impairment than those without depression (Fernandes Filho and de Melo, 2012), and there is a significant relationship between the extent of depression and the severity of loss of cognitive functioning in patients infected with HIV (Braganca and Palha, 2011). Further, the worsening of NeuroHIV could potentially increase risk for delayed diagnosis of other forms of dementia such as Alzheimer’s (Milanini and Valcour, 2017), although further studies are needed to assess this risk.

Compounding these issues, depressive symptoms are often overlooked, as almost 50% of HIV/AIDS patients suffering from depression are never diagnosed (Asch, 2003). In elderly patients with HIV, depression is often underdiagnosed and undertreated (Nyirenda, 2013; Zanjani et al., 2007), in part because older patients often present with a constellation of vague symptoms. These include somatic complaints such as headache and gastrointestinal disturbances, anger, and irritability instead of low mood (Krishnan, 2002), as well as poor sleep, appetite changes, lack of motivation, decreased concentration, fatigue, and weight loss, all of which can be caused by both depression and HIV. Further confusing the diagnostic process, the initial diagnosis of HIV often results in an “adjustment disorder”, which can present with depressive-like symptoms, and depressive-like symptoms can also be caused by substance intoxication, withdrawal and dementia (Angelino, 2002). Symptom confusion can be compounded by failure to use strict diagnostic criteria for MDD, such as the DSM-V or ICD-10, and instead defining depression based on physician reporting or structured screening surveys. Substitution of self-report or clinician rating is not an accurate substitute for neuropsychiatric testing (Justice, 2004; Underwood and Winston, 2016) and likely a major contributor to the large variation in depression prevalence seen in the literature. Because of these factors, it is difficult to delineate HIV symptoms from depression. This can contribute to delayed or inadequate diagnosis and treatment of this disorder in older HIV-infected adults (Malaspina, 2011; Cherner, 2004), exacerbating the development and spread of disease in this population.

2.3. Effects of cART in the aging population

Combination antiretroviral therapy (cART) – simultaneous treatment with several classes of antiretroviral drugs – was first developed in 1996 and rapidly became the primary treatment for HIV-infected individuals. Currently, the standard of care consists of three antiretroviral drugs from at least two different classes (Department of Health and Human Services, 2017), and many cART regimens are now available as a single tablet formulation (Astuti and Maggiolo, 2014). Some data show that older adults have a more robust viral suppression in response to cART (Silverberg, 2007; Orlando, 2006; Manfredi and Chiodo, 2000) although others do not see a difference between older and younger patients (Wellons, 2002; Greenbaum, 2008; Szadkowski, 2012). Some studies also show that despite effective cART, older adults have a blunted immune recovery relative to younger individuals, decreases in physical function, increased mortality, and faster disease progression (Khoury, 2017; Jourjy et al., 2015; Fatti, 2014). Notably, older patients have a number of distinct issues that can confound cART effectiveness. These are generally associated with medication compliance due to polypharmacy, as well as age-related cognitive and vision impairment, increased blood–brain barrier permeability, and diminished metabolism and kidney function (Burgess et al., 2015; Zeevi, 2010; Barclay, 2007; Winston and Underwood, 2015).

It is also likely that cART is less effective in older, HIV-infected individuals due to the additive effects of age-related conditions, including age-related inflammation (Deeks, 2011; Nasi, 2017). Treatment with cART generally reduces inflammation, decreasing T-cell activation and partially normalizing inflammation (Hileman and Funderburg, 2017; Jennes, 2004), especially early on in disease progression (de Paula, 2018; Allers, 2016). However, several studies show that cART has minimal effects on expression of several inflammatory markers, including IL-1β and IL-6 (Gay, 2011; Regidor, 2011; Osuji, 2018), and that there is no difference in inflammatory markers in patients regardless of time of initiation or duration of cART following infection (Ruggiero, 2015; Ruggiero, 2018; Amu, 2016). Further, two cohorts, the VACS-BC and APROCO-COPILOTE cohorts, showed elevated levels of inflammatory and immune activation markers in aging, long-term infected, cART-controlled HIV-infected patients (Bastard, 2015; Armah, 2012). This suggests that cART may have time and donor-dependent limits in its effectiveness in regard to immune recovery and control, and that these limits may be increased in older individuals with pre-existing inflammation and immune activation.

Several studies suggest that cART may specifically be less effective in controlling myeloid inflammatory activity. In a Thai cohort, early initiation of cART did not affect monocyte activation and levels of IL-6 and TNF-α remained elevated (Sereti, 2017), while in the MACS cohort, 1 year of cART normalized T-cell activity, but activation of monocytes and macrophages remained elevated (Wada, 2015). Persistent myeloid activation, measured by plasma sCD163, was shown in neurocognitively impaired, HIV-infected individuals on cART (Burdo, 2013), and a second group found that serum levels of sCD14, another myeloid activation marker, were elevated in adults over age 50 living with HIV on suppressive cART (Montoya, 2017). In the VACS-BC cohort, increased measures of monocyte activation (sCD14) and inflammation (IL-6) are correlated with increased mortality (So-Armah, 2016). In vitro, human monocyte-derived macrophages (MDM) from individuals on cART show elevated toll like receptor (TLR) responses and increased IL-6 and IL-1β production (Galvao-Lima, 2017; Merlini, 2016), and in THP-1 human macrophages, treatment with the cART drugs stavudine (d4T), zidovudine (AZT), nelfinavir (NFV) and lopinavir (LPV) increased production of IL-1β, TNF-α and ROS (Lagathu, 2007). Further the cART drug maraviroc (MVC), a CCR5 inhibitor, significantly increased activation of primary rat microglia exposed to gp120 (Lisi, 2012). These data suggest that myeloid cell-derived inflammation is not ameliorated by cART and suggest that this type of inflammation may be important to the development of age-related comorbidities in the aging, HIV-infected population (Lagathu, 2017).

3. Inflammation as a link between Depression, aging and NeuroHIV

There are a number of hypotheses regarding the etiology of depression, including nutritional deficiencies (Rao, 2008), abnormalities in circadian rhythm (Germain and Kupfer, 2008), impaired emotional processing (Carballedo, 2011), chronic inflammation (Hodes, 2015) and changes in monoamine levels, including serotonergic and adrenergic but also dopaminergic dysfunction of neural circuits (Belujon and Grace, 2017; Delgado, 2000; Dunlop and Nemeroff, 2007). While the precise etiology of depression is unclear, data indicate increased inflammation is a critical contributor to and target for treatment in this disease (Wohleb, 2016; Pfau et al., 2018; Miller and Raison, 2016; Kaufmann, 2017; Beydoun, 2019). Inflammation in both the CNS and peripheral regions are associated with depression, although each region’s role in initiating and sustaining depressive symptoms remains unclear. In older adults with HIV, emerging data indicate that the increased risk of neuropsychological comorbidities like depression is linked to the long-term use of cART, chronic inflammation, and persistent immune activation due to HIV infection (Schouten, 2014; Rivera-Rivera, 2014). Combined with age-associated increases in inflammation, these effects may also lead to higher rates of neuropsychiatric disorders in older HIV-infected individuals compared to healthy older adults (Rivera-Rivera, 2016; Effros, 2008; Leserman, 2008). Elevated levels of circulating inflammatory cytokines in HIV-infected individuals on cART (Regidor, 2011; Sereti, 2017; Funderburg, 2014; Gay, 2011; Shive, 2012), including IL-1β, TNF-α and IL-6, are associated with depressive symptoms in both rodents and humans (Hodes, 2015; Liu et al., 2012; Felger and Lotrich, 2013; Dowlati, 2010).

In many models, increased inflammation is associated with hyperactivity of the hypothalamic-pituitaryadrenal (HPA) axis and the release of glucocorticoids (Pariante and Lightman, 2008; Horowitz, 2013), which can be induced by both HIV infection and cART drugs (Collazos, 2003; Christeff, 1997). This suggests that HIV could disrupt the bidirectional immune interaction between the periphery and CNS (Wohleb, 2016; Raedler, 2011), triggering cascading inflammatory effects in both compartments. Inflammation resulting from depression has also been linked to activation of the indoleamine 2,3-dioxygenase (IDO) enzyme (Christmas et al., 2011; Dantzer, 2011), which initiates the metabolism of tryptophan into neurotoxic compound quinolinic acid. This could potentiate both depression and HIV neuropathogenesis, as IDO activity is increased in HIV-infected individuals and is only partially ameliorated by cART (Chen, 2014; Peltenburg, 2018; Jones, 2015). The Rhesus macaque model of NeuroAIDS shows myeloid lineage cells are the primary source of IDO in the brain, and that IDO expression is proportional to SIV viral load and IFN-γ (Burudi, 2002). Further, exposure to the HIV viral protein Tat in the brains of mice led to increased cytokine and IDO production along with development of depressive-like behaviors (Fu, 2011; Lawson et al., 2011).

A substantial amount of depression-associated inflammation is mediated by myeloid cell activation; microglia and perivascular macrophages in the CNS as well as different types of macrophages and monocytes in the periphery (Hodes, 2015; Wohleb, 2016; Yirmiya et al., 2015). Glucocorticoids increase the release of IL-1β, IL-6 and TNF-α from macrophages and monocytes (Busillo et al., 2011) and in social stress models of depression, extravasation of inflammatory monocytes and macrophages from the bone marrow is increased (Engler, 2004). Therapeutic administration of IFN-α, which increases macrophage inflammatory function (Raison et al., 2006), induces the development of depressive symptoms such as anhedonia, fatigue, and psychomotor retardation (Capuron, 2002; Raison, 2005). Further, the antidepressants fluoxetine and venlafaxine (Table 1) suppress immune activation and release of inflammatory factors such as IL-6 specifically by inhibiting macrophage function (Nazimek, 2017).

Table 1.

Compounds that Modulate the Dopaminergic System. A summary of compounds with varying mechanisms of action to increase or decrease dopaminergic activity. References in the table refer to the specific studies that demonstrate that these drugs change dopamine levels and/or dopamine turnover. As the aging HIV population uses a number of different drugs, many of which impact dopamine but are not always indicated to affect the dopaminergic system, caution should be used in taking them as they may affect the progression of NeuroHIV, depression, and other age-related comorbidities.

Generic Name Brand Names Mechanism Therapeutic Use References
Dopamine Precursors Levodopa Atamet, Duopa, Larodopa, Parcopa, Prolopa, Rytary, Stalevo, Sinemet Dopamine Precursor Parkinsons’ Disease (PD) (Yahr, 1969; Ng, 1970)
L-phenylalanine N/A Dopamine Precursor Dietary Supplement (Milner et al., 1986; Fernstrom and Fernstrom, 2007; Kapatos and Zigmond, 1977)
L-tyrosine N/A Dopamine Precursor Dietary Supplement (Fernstrom and Fernstrom, 2007; Kapatos and Zigmond, 1977; During et al., 1989)
Dopamine Receptor Agonists Amantadine Symmetrel Dopamine Receptor Agonist/Dopamine Reuptake Inhibitor PD, Antiviral (Von Vigtlander and Moore, 1971; Mizoguchi, 1994)
Apomorphine Apokyn Dopamine Receptor Agonist PD (Andén, 1967; de La Fuente-Fernandez, 2001)
Aripiprazole Abilify, Aripipex Dopamine Receptor Agonist# Schizophrenia, Bipolar Disorder, Major Depressive Disorder (MDD), Obsessive Compulsive Disorder (OCD), Autism (Li, 2004)
Bromocriptine Cycloset, Parlodel, Brotin Dopamine Receptor Agonist PD, Pituitary tumors, Type 2 Diabetes, Hyperprolactinaemia, Neuroleptic malignant syndrome (Brannan, 1993)
Lisuride Proclacam, Revanii, Dopergin Dopamine Receptor Agonist PD, Migraine (Kehr, 1977)
Memantine Axura, Akatinol, Abixa, Memox, Ebixa, Namenda Dopamine Receptor Agonist Alzheimers’ Disease (AD) (Spanagel et al., 1994)
Pramexipole Mirapex, Mirapexin, Sifrol Dopamine Receptor Agonist PD, Restless Leg Syndrome (RLS) (Carter and Müller, 1991)
Piribedil Pronoran, Trivastal Retard, Trastal, Trivastan, Clarium Dopamine Receptor Agonist Parkinsons’ Disease, Elderly dizziness and cognitive deficit, retinal ischemic manifestations (Gil-Loyzaga, 1994; Delbarre, 1995)
Rotigotine Neupro Dopamine Receptor Agonist MDD, PD, RLS, Willis-Ekborn Disease (Kehr, 2007)
Dopamine Receptor Antagonists Amisulpride Solian Dopamine Receptor Antagonist Schizophrenia, Dysthymia (Schoemaker, 1997)
Chlorpromazine Largactil, Thorazine Dopamine Receptor Antagonist ADHD, Bipolar Disorder, Nausea, Schizophrenia, Vomiting (Starke, 1978; Stamford et al., 1986)
Clozapine Clozaril Dopamine Receptor Antagonist Treatment-resistant Schizophrenia (Youngren, 1999; Meltzer, 1989; Drew, 1990)
Domperidone Motilium Dopamine Receptor Antagonist Nausea, vomiting, Gastroprokinetic, Galactagogue (Champion, 1988)
Fluphenazine Prolixin Dopamine Receptor Antagonist Schizophrenia (Bacopoulos et al., 1979; Goosey and Doggett, 1983)
Haloperidol Haldol Dopamine Receptor Antagonist Schizophrenia, Bipolar Disorder, Tourette Syndrome, Nausea, Vomiting, Delerium (Drew, 1990; Gudelsky and Porter, 1980; Bunney and Grace, 1978)
Loxapine Loxitane Dopamine Receptor Antagonist Schizophrenia, Bipolar Disorder (Li et al., 2003)
Metoclopramide Primperan, Reglan Dopamine Receptor Antagonist Nausea, Vomiting, Migraine (Peringer, 1976)
Molindone Moban Dopamine Receptor Antagonist Schizophrenia (McMillen and McDonald, 1983)
Olanzapine Zyprexa Dopamine Receptor Antagonist Schizophrenia, Bipolar Disorder (Zhang, 2000; Li, 1998)
Perphenazine Trilafon Dopamine Receptor Antagonist Schizophrenia, Bipolar Disorder, Depression (Mjörndal and Persson, 1990)
Pimozide Orap Dopamine Receptor Antagonist Schizophrenia, Tourette Syndrome (Matsuo, 2010; Parsons et al., 1993)
Prochlorperazine Compro Dopamine Receptor Antagonist Schizophrenia, Nausea, Migraine, Anxiety (Goosey and Doggett, 1983)
Quetiapine Seroquel Dopamine Receptor Antagonist Schizophrenia, Bipolar Disorder, Depression, Sleep Aid (Pira et al., 2004; Silverstone et al., 2012)
Risperidone Risperdal Dopamine Receptor Antagonist Schizophrenia, Bipolar Disorder, Autism (Hertel, 1996; Huang, 2006)
Sulpiride* Dogmatil Dopamine Receptor Antagonist Schizophrenia, Depression (Andersen and Gazzara, 1996)
Thiothixene Navane Dopamine Receptor Antagonist Schizophrenia, Bipolar Disorder (Bjerkenstedt, (1970), 1977)
Trifluoperazine Stelazine Dopamine Receptor Antagonist Schizophrenia, Generalized Anxiety Disorder (GAD) (Goosey and Doggett, 1983)
Ziprasidone Geodon Dopamine Receptor Antagonist Schizophrenia, Bipolar Disorder (Li et al., 2003)
Dopamine Reuptake Inhibitors Armodafanil Nuvigil, Acronite Dopamine Reuptake Inhibitor Sleep Disorders (Loland, 2012)
Benzatropin Cogentin Dopamine Reuptake Inhibitor PD, Dystonia (Goodale and Moore, 1975)
Bupropion Elontril, Wellbutrin, Zyban Dopamine Reuptake Inhibitor Depression, Smoking Cessation, ADHD (Ascher, 1995; Nomikos, 1992)
Dexmethylphenidate Attenade, Focalin Dopamine Reuptake Inhibitor ADHD (Volkow, 2001)
Diphenylpyraline Allergen, Arbid, Belfene, Diafen, Hispril, Histyn, Lergobine, Lyssipol, Mepiben, Neargal Dopamine Reuptake Inhibitor Allergies, PD (Oleson, 2012)
Esketamine Ketanest, Spravato Dopamine Reuptake Inhibitor Anesthesia, Treatment-resistant depression (Hashimoto, 2017)
Methylphenidate Methylin, Concerta, Medikinet, Ritalin, Equasym XL, Metadate, Quillivant XR Dopamine Reuptake Inhibitor ADHD (Heal, 2008)
Modafanil Alertec, Modavigil, Modiodal, Provigil, Modalert Dopamine Reuptake Inhibitor Sleep Disorders (Ferraro, 1996)
Nefazdone Serzone, Dutonin, Nefadar Dopamine Reuptake Inhibitor MDD (Olausson, 1998; Dremencov, 2005)
Nomifensine Merital, Alival Dopamine Reuptake Inhibitor Depression, Anxiety Disorders (Karoum, 1994)
Sertraline Zoloft Weak Dopamine Reuptake Inhibitor MDD, Anxiety Disorders, OCD (Kitaichi, 2010)
Tripelennamine Pyrbenzamine Weak Dopamine Reuptake Inhibitor Allergies (San-Martin-Clark, 1996; Oishi, 1994)
Venlafaxine Effexor, Lanvexin, Trevilor Weak Dopamine Reuptake Inhibitor MDD, Anxiety Disorders (Czubak, 2010)
Monoamine Oxidase Inhibitors Phenelzine Nardil, Nardelzine Non-Selective MAO-I MDD, Anxiety Disorders (Juorio et al., 1986)
Rasagiline Azilect Selective MAO-B Inhibitor PD (Finberg, 1998)
Selegiline Anipryl, Emsam, Deprenyl, Eldepryl, Zelapar, Selgene Selective MAO-B Inhibitor PD, MDD, ADHD (Kitaichi, 2013)
Tranylcypromine Parnate, Jatrosom Non-Selective MAO-I MDD, Mood Disorders, Anxiety Disorders (Shioda, 2004; Ainsworth, 1998)
Vesicular Monoamine Transporter 2 (VMAT2) Inhibitors Reserpine Serpalan, Serpasil VMAT2 Inhibitor High Blood Pressure (Elverfors and Nissbrandt, 1991; Okada, 1993)
Tetrabenazine Nitoman, Xenazine VMAT2 Inhibitor Huntington’s Disease, Tardive Dyskinesia, Tourette Syndrome, Hemiballismus (Owesson-White, 2012)
Other Drugs with Dopaminergic Mechanisms Agomelatine* Melitor, Thymanax, Valdoxan Dopamine Releasing Agent Depression (Millan, 2003)
Buspirone Buspar, Buspar Dividose, Vanspar Dopamine Releasing Agent Anxiety Disorders (McMillen and McDonald, 1983; Silverstone et al, 2012; Sakaue, 2000)
Clomipramine Anafranil, Clofranil Weak Dopamine Reuptake Inhibitor/Weak Dopamine Receptor Agonist Depression, OCD, Anxiety Disorders, Other Psychiatric Disorders (Ichikawa and Meltzer, 1995)
Desipramine Norpramin, Pertofrane Dopamine Releasing Agent Depression (Dremencov, 2005)
Disulfiram Antabuse, Antabus Prevents Dopamine Breakdown Cocaine Dependence, Cancer, Alcoholism (Devoto, 2012)
Donepezil Aricept Dopamine Releasing Agent AD (Zhang et al., 2004)
Entacapone Comtan Catechol-O-Methyl Transferase Inhibtor PD (Gerlach, 2004)
Fluoxetine Prozac, Sarafem Dopamine Releasing Agent MDD, OCD, Bulimia, Panic Disorder, Premenstrual Dysphoric Disorder (Zhang, 2000; Sakaue, 2000; Ichikawa and Meltzer, 1995)
Folate N/A Increases Dopamine Turnover Dietary Supplement, Depression (Miller, 2008)
Galantamine Nivalin, Razadyne, Razadyne ER, Reminyl, Lycoremine Dopamine Releasing Agent AD (Schilstrom, 2007)
Imipramine Tofranil Weak Dopamine Receptor Antagonist Depression (Ichikawa and Meltzer, 1995)
L-methylfolate N/A Increases Dopamine Turnover Dietary Supplement, Depression (Miller, 2008)
Lisdexamfetamine Vyvanse Dopamine Releasing Agent ADHD, Binge eating disorder (Rowley, 2012)
Metirosine Metyrosine Tyrosine Hydroxylase Inhibitor Hypertension, Headahce, Tachycardia, Constipation, Tremors (Naruse, 2018)
Mirtazapine Remeron Dopamine Releasing Agent Depression (Nakayama et al., 2004)
Moclobemide* Aurorix, Manerix Selective MAO-A Inhibitor Depression, Anxiety Disorders (Da Prada, 1989)
Paroxetine Paxil, Seroxat Dopamine Releasing Agent Depression, OCD, PTSD, Panic Disorder, Generalized Anxiety Disorder, Premenstrual Dysphoric Disorder (Nakayama, 2002)
Pergolide* Permax Dopamine Receptor Agonist PD (Herdon and Nahorski, 1987; Fuller and Snoddy, 1981)
Reboxetine Edronax Dopamine Releasing Agent Depression (Linner, 2001)
S-adenosyl methionine N/A Increases Dopamine Turnover Dietary Supplement, Depression (Miller, 2008; Schalinske and Smazal, 2015)
Tolcapone Tasmar Catechol-O-Methyl Transferase Inhibtor PD (Lapish, 2009; Tunbridge, 2004)
Trimipramine Surmontil, Rhotrimine, Stangyl Weak Dopamine Reuptake Depression (Taylor, 1996)
Drugs of Abuse Alcohol N/A Dopamine Releasing Agent N/A (Bosse and Mathews, 2011; Yan, 1999; Weiss, 1996)
Alpha-Pyrrolidinopentiophenone (PVP) N/A Dopamine Reuptake Inhibitor N/A (Hataoka et al., 2017; Glennon and Young, 2016)
Amphetamine N/A Dopamine Releasing Agent N/A (Moghaddam and Bunney, 1989; Tor-Agbidye et al., 2001)
Cocaine N/A Dopamine Reuptake Inhibitor N/A (Wightman, 2007; Shou, 2006; Phillips, 2003)
Heroin N/A Weak Dopamine Releasing Agent N/A (Hemby, 1995; Smith, 2006)
Ketamine Ketalar Dopamine Reuptake Inhibitor Depression (Kokkinou et al., 2018)
Lysergic Acid Diethylamide (LSD) N/A Dopamine Receptor Agonist N/A (Smith et al., 1975; Antkiewicz-Michaluk et al., 1997)
Methylenedioxymethamphetamine (MDMA) N/A Dopamine Releasing Agent N/A (Baumann et al., 2008; Gudelsky et al., 1994)
Methylenedioxypyrovalerone (MDPV) N/A Dopamine Reuptake Inhibitor N/A (Glennon and Young, 2016; Shekar, 2017)
Methamphetamine N/A Dopamine Releasing Agent N/A (Wilson, 1996; Kita, 2000)
Morphine MorphaBond ER, Arymo ER, Astramorph-PF Dopamine Releasing Agent Pain Reliever (Anagnostakis and Spyraki, 1994; Vander Weele, 2014)
Nicotine N/A Weak Dopamine Releasing Agent N/A (Wing, 2015; Di Chiara, 2000)
Oxycodone Xtampza ER, Oxaydo, Roxicodone, and Oxycontin Dopamine Releasing Agent Pain Reliever (Vander Weele, 2014; Zhang, 2009)
Phencyclidine (PCP) N/A Dopamine Reuptake Inhibitor N/A (Hondo, 1994)
Tetrahydrocannabinol (THC) N/A Dopamine Releasing Agent N/A (Chen, 1993; Pistis, 2002)
*

Not prescribed in the United States.

#

Acts as both a dopamine receptor agonist (presynaptic) and a dopamine receptor antagonist (postsynaptic).

In the CNS, rodent models of stress-induced depression show disturbances in microglial function (Tong, 2017; Kreisel, 2014) and neuroimaging studies in individuals with major depressive disorder show increased microglial density and inflammatory activity (Li et al., 2018; Li et al., 2018). Similar to what occurs in the non-HIV aged brain, immune cells such as microglia in HIV-infected persons may be “primed” to an activated phenotype in response to inflammatory stimuli in the periphery (Matt and Johnson, 2016; Tedaldi et al., 2015). This might occur through priming of the NLRP3 inflammasome, which is strongly correlated with the development of depression (Iwata et al., 2013; Wong, 2016) and increases depressive symptoms in stress-induced animal models (Alcocer-Gomez, 2016; Pan, 2014). The microglial inflammasome pathway is active during aging (Wang, 2018; Salminen, 2012), and is also activated by HIV infection (Chivero, 2017; Mamik, 2017; Walsh, 2014). Once primed by HIV and/or in response to aging, microglia may display an aberrant response to inflammatory stimuli, enhancing neuroinflammation by increasing the production of mediators such as IL-1β and IL-18, and contributing to the development or maintenance of depression. In line with this theory, the release of inflammatory mediators by activated myeloid cells in the CNS has been shown to be a better correlate of neuropathogenesis than the actual brain viral load (Kraft-Terry, 2010). Together these data suggest that depression, HIV and aging may act synergistically on a number of inflammatory pathways, many mediated by myeloid cells, to potentiate the development and maintenance of neurologic disease.

4. The role of dopamine in NeuroHIV and depression

Both depression and NeuroHIV are associated with changes in dopamine, an important CNS neurotransmitter associated with motor control, cognition, learning, reward and other processes (BrombergMartin et al., 2010). While both depression and NeuroHIV are also associated with other major neurotransmitter systems, such as serotonin, norepinephrine, glutamate, and GABA, the effects of these disorders on these systems have been discussed in greater detail elsewhere and refer the reader to a number of recent publications (Vázquez-Santiago, 2014; Sperner-Unterweger et al., 2014; Hammoud, 2010; Cody and Vance, 2016; Buzhdygan, 2016). Infection with HIV is strongly associated with dopaminergic dysfunction throughout disease progression. Studies show an increase in CNS dopamine in subcortical structures in early stage disease, and a reduction in CNS dopamine during more advanced infection. Increased dopamine is seen in the cerebrospinal fluid (CSF) of cART-naive, HIV-infected people in early stage disease (Scheller, 2010; Horn, 2013). while, individuals in later stages show decreases in CNS and CSF dopamine (Larsson, 1991; Berger, 1994; di Rocco, 2000; Koutsilieri et al., 2001; Kumar, 2011). In addition, HIV infection and neurodegenerative damage are particularly pronounced in dopaminergic areas, such as the caudate, putamen, globus pallidus, and substantia nigra (Navia, 1986; Becker, 2011; Kumar, 2009; Berger and Arendt, 2000; Aylward, 1995; Itoh et al., 2000; Gongvatana, 2013; Clifford, 2017).

Some studies also show CNS hypermetabolism in subcortical regions such as the basal ganglia in earlier stages of infection, and subcortical hypometabolism in later stage disease (Rottenberg, 1987; van Gorp, 1992; Rottenberg, 1996; Georgiou, 2008), changing CNS dopamine levels during these stages. These changes in dopaminergic tone may be due to the selective sensitivity of dopamine neurons to HIV-induced neurotoxicity (Schier, 2017; Nath, 2000; Aksenova, 2006). The death of dopaminergic neurons would decrease dopamine availability (Bennett et al., 1995; Cass, 2003; Maragos, 2002), and many HIV patients demonstrate parkinsonian symptoms associated with decreased dopamine, including bradykinesia and extrapyramidal symptoms (Koutsilieri, 2002). They are also sensitive to parkinsonism-inducing agents (Hriso, 1991; Edelstein and Knight, 1987), like antipsychotics that act as dopamine antagonists (Table 1), supporting the hypothesis that extensive dopaminergic degeneration takes place during HIV infection (Purohit et al., 2011). In addition, HIV proteins can bind to DAT and elevate dopamine levels by disrupting uptake (Zhu, 2011; Zhu, 2016; Zhu, 2009), further contributing to later stage HIV-induced neurological damage and neuropathogenesis.

In addition to mediating neurotoxicity, these changes in dopamine concentrations may also potentiate neuropathogenesis by modulating immune function (Matt and Gaskill, 2019; Gaskill, 2013; Pinoli et al., 2017). Data from our lab shows dopamine can significantly increase HIV infection (Gaskill, 2009; Gaskill, 2014), and promotes an inflammatory phenotype in primary human MDM, which occurs irrespective of cART treatment (Nolan and Gaskill, 2018; Nolan, 2018; Gaskill, 2012). Additionally, modulation of the dopaminergic system can alter production of inflammatory mediators such as nitric oxide, IL-6, and TNF-α in other myeloid cells and cell lines (Yamamoto, 2016; Huck, 2015; Bone, 2017). These data suggest changes in dopamine associated with HIV infection of the CNS may enhance infection and exacerbate neuroinflammation, playing an important role in the development of NeuroHIV in chronically infected individuals on cART.

These alterations in CNS dopamine could also substantially impact the development of comorbid depression in older people living with HIV. While the primary neurotransmitter circuits mediating depressive behaviors are often thought to be serotonergic and/or noradrenergic, changes in dopaminergic neurotransmission, and the interactions between these circuits are also important in depression (Belujon and Grace, 2017; Dunlop and Nemeroff, 2007; Tye, 2012). It has been postulated that the mesolimbic dopamine system is involved in the etiology of depression, with decreased activation of this system inducing anhedonia, reduced motivation, and decreased energy level in most depressed individuals (Dailly, 2004; Nestler and Carlezon, 2006). Depressed patients show decreases in jugular vein plasma dopamine and its metabolites (Lambert, 2000), and imaging studies show significantly lower dopamine transporter (DAT) binding compared with healthy subjects (Sarchiapone, 2006; Pizzagalli, 2019; Meyer, 2001), suggesting a compensatory downregulation to lower dopamine concentrations. Animal models of depression also show altered dopamine regulation is associated with depressive-like symptoms such as anhedonia, behavioral despair, and learned helplessness (Kram, 2002; Chaudhury, 2013; Di Chiara and Tanda, 1997; Perona, 2008; Chang and Grace, 2014).

Overall, these data suggest that changes in CNS dopamine could significantly impact the progression of both NeuroHIV and depression and indicate that further evaluation of the risks and benefits of dopamine-altering therapeutics is needed for proper treatment of the older, HIV-infected population. Further, other neurotransmitter systems such as serotonin, norepinephrine, GABA, and glutamate can impact the regulation of dopamine (Guiard, 2008; Nutt, 2008; Floresco, 2003; El Mansari, 2010). Further, these interactions change with aging (Mora et al., 2008), suggesting the use of antidepressants that impact more than one system provides an additional route by which dopamine could impact depressive symptoms. This also suggests that the neuroinflammation prominent in older HIV-infected individuals on cART could be exacerbated by the dopamine increases resulting from the use of many antidepressants, even those not primarily targeting dopamine (Table 1).

5. Dopamine-associated mechanisms by which antidepressants and cART potentiate disease in the aging population

Effective treatment of depressed, older adults living with HIV often requires a variety of different medications, including antidepressants, cART drugs and a number of other medications, as these individuals have a high disease burden (Havlik et al., 2011). Understanding the drug-drug interactions occurring in these therapeutic regimens is critical to effective treatment in this population, specifically the effects of cART on depression, and the effects of antidepressants on HIV disease. While many studies have examined cART interactions with other drugs, including antidepressants, very few studies have assessed the effects of cART on monoamine transmission, in particular dopamine. As dopamine is an inflammatory mediator in both the CNS and periphery (Matt and Gaskill, 2019) that is strongly connected to depression and HIV, a better understanding of the specific effects of dopamine-modulating therapeutics on the development and persistence of HIV and depression in older individuals is warranted.

5.1. Antidepressants and dopamine in the aging population

The specific antidepressant regimens for older adults are not different from those recommended for the general HIV-infected population, despite a higher prevalence of comorbidities and potential for drug-drug interactions. Although early antidepressants, such as tricyclic compounds, primarily targeted the dopaminergic system, in the current era, antidepressants that predominantly act as serotonin- and combined serotonin/noradrenaline-reuptake inhibitors (SSRIs and SNRIs) are drugs of choice in HIV-infected patients with depression (Caballero and Nahata, 2005). Notably, despite the focus on serotonin and/or norepinephrine, indirect effects of the dopaminergic system in the antidepressant-like activity of SSRIs have been demonstrated (Renard, 2001; Cervo et al., 1990), and many of these newer drugs also directly affect the dopaminergic system (Subbaiah, 2018) (Table 1).

A number of different types of antidepressants are efficacious in the HIV-infected population, although no single drug has been shown to be clearly superior (Yanofski and Croarkin, 2008). Many SSRIs and SNRIs such as fluoxetine, sertraline, reboxetine, and paroxetine have shown to be effective and well-tolerated in HIV-infected individuals in various stages of HIV progression (Carvalhal, 2003; Grassi, 1997; Ferrando et al., 1997). While several studies reported a potential benefit to tricyclic antidepressants (TCAs) and related compounds like mirtazipine (Rabkin, 1994; Elliott and Roy-Byrne, 2000), they are not recommended because of their many side-effects. In older populations, TCAs are contraindicated for precisely this reason, as older adults, particularly those with HIV, are sensitive to orthostatic hypotension, urinary retention, and neurocognitive impairment, as well as anticholinergic, hypertensive and sedating effects (Yanofski and Croarkin, 2008; American Geriatrics Society, 2012; McLennon et al., 2003). Evaluations of the effectiveness of antidepressants in older people living with HIV have not determined whether they improve cognition, although mixed results have been found for antidepressants improving cognitive symptoms in the general HIV population (Hinkin, 2001; Schifitto, 2007; Sacktor, 2018). However, treating depression in non-HIV individuals has generally been shown to improve cognition (Rosenblat et al., 2015), and further attempts to use antidepressants therapeutically should be evaluated for the aging HIV population in the future.

Notably, inflammatory symptoms related to reduced motivation and motor slowing, which are more common in older adults, are difficult to treat with standard antidepressants like SSRIs in patients with depression (Konsman et al., 2002; Goldsmith, 2016; Diniz, 2019). Dopaminemediated potentiation may be involved in these SSRI-resistant, inflammation-related symptoms in the HIV population, disrupting or counteracting the impact of these agents through immunomodulatory effects (Nolan and Gaskill, 2018), and age-related inflammation may exacerbate this even further. It may also be that there is only a small therapeutic window for these drugs to be effective because disrupted dopaminergic function is seen at all stages of HIV infection; high dopamine early on in infection and low during dopaminergic neuron loss in later stages of infection.

Given the changes in dopamine concentrations during HIV infection, augmentation strategies, such as the addition of an atypical antipsychotic to an SSRI treatment, are another way to optimize treatment in patients with treatment-resistant depression (Zhou, 2015). This type of depression is more common in the elderly population (Knöchel, 2015), and may benefit from a restoration of more optimal dopamine levels. Some SSRIs like fluoxetine or escitalopram induce a decrease in dopaminergic neuron firing in the ventral tegmental area (Dremencov et al., 2009; Di Mascio, 1998; Prisco and Esposito, 1995), and the combination of the antipsychotic aripiprazole with escitalopram reversed the inhibitory action of the antidepressant on firing (Chernoloz et al., 2009). The combination of fluoxetine with olanzapine, another atypical antipsychotic, also induced an increase in extracellular dopamine in the prefrontal cortex, which was higher than either drug alone (Zhang, 2000). Prior to cART, patients with AIDS showed improvements in delirium after treatment with dopamine receptor antagonists haloperidol and chlorpromazine (Breitbart, 1996), but effects of these antipsychotics on depressive symptoms in the cART-treated population has not been investigated. While these treatments may be more effective in HIV-infected individuals, careful evaluation is warranted in older adults, as this population is vulnerable to the adverse effects of these drugs (Gareri, 2014), and HIV patients in general have a higher susceptibility to neuroleptic-induced extrapyramidal symptoms (Hill and Lee, 2013; Dolder et al., 2004).

The reduced DAT and dopamine found in chronically infected HIV patients with more severe cognitive and motor deficits, who are now more likely to be older adults, also suggests that they may benefit from treatment with dopaminergic agents. One study showed that sustained-release bupropion, a dopamine/norepinephrine reuptake inhibitor, is effective and well-tolerated for the treatment of depression in HIV-infected patients, regardless of length of HIV progression (Currier et al., 2003). Phenelzine and tranylcypromine, both monoamine oxidase inhibitors (MAOIs), have been shown to be effective in subpopulations of HIV-infected patients with depression (Watkins et al., 2011). Triple reuptake inhibitors, such as venlafaxine, inhibit the reuptake of serotonin, norepinephrine, and dopamine (Marks et al., 2008), and have the potential to be more beneficial in HIV-infected individuals than current first line therapies such as SSRIs. However, a drug-interaction study found that venlafaxine decreased the plasma concentration of the protease inhibitor indinavir (Levin, 2001), suggesting it may interfere with the action of cART.

Studies have also found improvement on mood response rates and cognitive performance in HIV patients after treatment with methylphenidate (Hinkin, 2001), dextroamphetamine (Wagner and Rabkin, 2000) and selegiline (No author, 1998). The compounds folic acid, L-methylfolate, or S-adenosyl-methionine (Table 1), which boost tetra-hydrobiopterin (BH4) activity and facilitate the synthesis of dopamine (Miller, 2008), have all shown efficacy as antidepressants (Papakostas, 2010; Godfrey, 1990; Ginsberg et al., 2011; Galizia, 2016). Additionally, studies of modafinil in HIV patients over 6 months showed that those still taking modafinil had a decline in HIV RNA viral load, more energy and fewer depressive symptoms (Rabkin, 2010). However, a number of dopamine-altering antidepressants have also been shown to disrupt the immune system and alter the production of inflammatory modulators (Martino, 2012). In clinical trials in HIV-infected individuals, the MAOI selegiline showed no neurological benefit (Evans, 2007; Schifitto, 2009). A number of studies also indicate that selegiline and levodopa further exacerbate neurotoxicity and disease progression in a model of neuroAIDS (Czub, 2004; Czub, 2001). As these drugs increase dopamine levels, it is possible that increases in replication or inflammation (Nolan, 2018; Gaskill, 2009; Gaskill, 2014) could counteract any beneficial effects of this treatment. Overall, these studies suggest that the dopamine-mediated effects of antidepressants in older people living with HIV could substantially influence both depression and NeuroHIV, but that more research is needed to understand both the benefits and risks of using specific antidepressants in this population.

5.2. Impact of antidepressants on HIV pathogenesis

Few studies examine whether antidepressants directly affect the HIV replication process, but those that have been performed suggest that they may, although the specific effects and mechanisms remain unclear. Derivatives of paroxetine and femoxetine have been shown to directly inhibit HIV replication (Kristiansen and Hansen, 2000), and other studies report increased viral suppression and CD4+ T cells in HIV patients taking antidepressants (Pence, 2015). Citalopram decreases HIV infection in primary human macrophages and T-cell lines (Benton, 2010), possibly through a decrease in the expression of HIV receptor and coreceptor mRNA (Greeson, 2016). Serotonin itself has been shown to both increase and decrease HIV infection (Sidibe, 1996; Manéglier, 2008), elevated plasma serotonin is correlated with more effective viral suppression by cART (Miguez-Burbano, 2014), and prior to cART, the level of plasma serotonin in infected individuals is also inversely correlated with neuropsychiatric symptoms and disease severity (Launay, 1988; Launay, 1989).

As we and others have shown that dopamine can increase HIV replication, antidepressants that increase dopamine could also have a similar effect. In the cART era, and especially in the older, HIV-infected population, chronic inflammation is a central driver of disease, and antidepressants can also directly affect immune function. Reductions in cytokines such as IL-4, IL-6, IL-1β, TNF-α and IL-10 have been demonstrated after SSRI treatment (Wiedlocha, 2018; Kohler, 2018; Warner-Schmidt, 2011), although this does not always correlate with decreased depressive symptoms (Strawbridge, 2015; Hannestad et al., 2011). The TCAs clomipramine and imipramine as well as the SSRI fluoxetine may have anti-inflammatory and neuroprotective effects via modulation of glial activation (Hwang, 2008; Obuchowicz, 2014). Bupropion has been shown to suppress Th1 and Th17 immune responses (Ebbinghaus, 2012; Jha, 2017), and pramipexole, a dopamine agonist with evidence of efficacy in treatment-resistant depression (Fawcett, 2016), has been shown to inhibit the production of IL-17 (Lieberknecht, 2017). More research is needed to evaluate the specific effects of antidepressants on HIV replication and chronic inflammation, focused on both the impact on HIV pathogenesis and the effectiveness of antidepressants in the aging HIV population.

5.3. Neuropsychiatric effects of cART in the aging population

The primary function of cART drugs is viral suppression, but antiretroviral treatment can also ameliorate depressive symptoms and increase medication adherence in association with restored immune activity (Gutiérrez, 2014; Tsai, 2010). However, many other studies have found cART has no effect or adverse effects on depression. For example, initiation or changes in cART treatment can induce depression (Hill and Lee, 2013; Kaestner, 2012), and cART may also precipitate or worsen cognition, mood, and daily functioning (van der Lee, 2007; Ellis, 2010; Letendre, 2010). Depression can also inhibit the restoration of immune function, suggesting a reciprocal interaction between the effects of depression and cART on immune activation (Alciati, 2007; Hartzell et al., 2008). Further, as depression is associated with inflammation, the exacerbated myeloid inflammation associated with HIV, aging and cART may contribute to the development of this disease (Rivera-Rivera, 2016).

Neuropsychiatric symptoms from cART start anywhere within hours to months after initiation of cART, and can be transient or long-lasting, making the precise mechanisms underlying these side effects unclear (Kaestner, 2012; Gelmon, 1989). Several cART drugs in particular have been reported to induce neurological complications, although studies on the neuropsychiatric and inflammatory effects of specific cART drugs, are scarce, particularly in older adults. Specifically, efavirenz (EFV), a non-nucleoside reverse transcriptase inhibitor (NNRTI), causes neuropsychiatric adverse events (NPAEs) in a substantial number of patients, in particular increasing the risk for depression (Mollan, 2014; Silveira, 2012). In older adults, these effects may be partially mediated by slower metabolism of efavirenz, as this contributes to loss to care in an older population (Torgersen, 2019), and speed of efavirenz metabolism was positively correlated with better neuropsychological performance in older adults (Sandkovsky, 2017). In rats, efavirenz and efavirenz-containing regimens induce depressive-like behavior and lead to increases in IL-1β and TNF-α, and oxidative stress, suggesting an association with increased inflammatory activity (Akang, 2019; O’Mahony, 2005).

Two nucleotide reverse transcriptase inhibitors (NRTIs), including the first antiretroviral drug approved to treat HIV, zidovudine, are also associated NPAEs. Zidovudine has been shown to induce manic episodes following treatment, even in patients with no previous psychiatric history (Wright, 1989), while the NRTI abacavir (ABC) is associated with depression, fatigue, headache and psychotic symptoms (Colebunders, 2002; Foster et al., 2004). Recent studies show the second-generation integrase strand transfer inhibitor (INSTI) dolute-gravir (DTG), which is currently part of the recommended first line ART-regimen for adults, can also induce NPAEs. The effects of dolute-gravir include severe depression, as well as insomnia, anxiety and suicidality (Fettiplace, 2017; Scheper, 2018; Llibre, 2019), and one study observed 3-fold higher discontinuation rates for dolutegravir containing regimens in the older population because of NPAEs (Hoffmann, 2017). And while Truvada, a popular pre-exposure prophylaxis (PrEP) formulation has not been shown to cause depression (Defechereux, 2016), both of the drugs comprising this therapy, emtricitabine (FTC) and tenofovir disoproxil fumarate (TDF), have shown depressive side effects (Arribas, 2008; Pozniak, 2006). Altogether, these studies underscore the need for more research on the role of cART drugs in the development of NPAEs, particularly depression in the older, HIV-infected population.

5.4. Dopaminergic effects of cART

As alterations in dopaminergic neurotransmission are strongly correlated with depression, it is likely that changes in this neurotransmitter are involved in the neuropsychiatric impact of cART. Very few studies have directly examined the effects of cART on the dopaminergic system, but those that have do show some cART drugs could have a substantial impact on dopamine. Efavirenz, which is strongly implicated in a number of neuropsychiatric complications in people living with HIV (Mollan, 2014; Silveira, 2012), has been shown to block presynaptic DAT (Gatch, 2013), inhibit MAO-A, and upregulate MAO-B (Akang, 2019; Dalwadi, 2016). Further, acute treatment with efavirenz can increase striatal levels of dopamine and decrease turnover rate which was associated with anxiety-like behaviors in rats, and subchronic efavirenz reduced the striatal levels of dopamine and increased the turnover rate which was associated with depressive-like effects (Cavalcante, 2017). In mice prenatally exposed to zidovudine, D1-like receptor signaling is hyporesponsive (Venerosi, 2005), while a number of protease inhibitors (PIs), particularly lopinavir, inhibit the activity of PMAT, a secondary monoamine uptake system (Duan, 2015). Further, CCR5 can be modulated by dopamine (Basova, 2018), and in a primate model of Parkinson’s, the entry inhibitor (EI) maraviroc actually protected from depletion of neurotransmitters and deterioration of the substantia nigra, improving locomotor function (Mondal, 2019). These different alterations in the dopaminergic system and subsequent behaviors suggest that the dopamine-modulating effects of cART may be involved in the emergence or disappearance of different types of neuropsychiatric adverse effects in HIV-infected patients. As some studies have shown that effective viral suppression correlates with less dopaminergic injury (Wang, 2004), future studies are warranted to evaluate HIV patients before and after antiretroviral treatment to better determine the effect of cART on dopaminergic circuits.

5.5. Interactions of cART, antidepressants and dopamine in the aging population

Combining neuropsychiatric treatment and cART can be of great benefit with respect to both HIV infection and depression. HIV-infected patients suffering from depression and treated with antidepressants are more likely to receive appropriate care for HIV than untreated subjects (Sambamoorthi, 2000), and adherence to cART has been shown to be higher in patients that are adherent to antidepressants (Walkup, 2008; Yun, 2005). Many studies show effectively treating depressive symptoms in the infected population reduces the incidence of medical complications and improves prognosis and quality of life (Watkins et al., 2011; Fulk, 2004). However, pharmacological interactions between cART and antidepressants have to be more carefully considered in order to successfully treat both HIV infection and depression. Additional consideration is needed in aging HIV individuals, where pharmacokinetic alterations with aging can result in changes to both body composition and function of drug-eliminating organs (Mangoni and Jackson, 2004; Shi and Klotz, 2011). The majority of clinical studies of antiretroviral therapy have not specifically evaluated patients over 50 years old, and therefore do not show adverse effect rates in older versus younger patients. The available studies of drug interactions often focus on healthy, younger patients without polypharmacy and with minimal or limited comorbid conditions or organ compromise, generating conclusions that may not be applicable to an older population. Currently, the best recommendations for treatment with antidepressants in older individuals are to start at low doses, and titrate slowly, especially in those with advanced illness or complex medication regimens (Burgess et al., 2015; Arseniou et al., 2014). Other treatment strategies such as cognitive behavioral therapy to reduce depressive symptoms may also be beneficial in the aging HIV population, in order to avoid drug interactions (Spies et al., 2013).

While there are many reported interactions between cART and antidepressants (Lefkowitz, 2007), most are in reference to interference with cytochrome p450 enzymes, particularly in respect to changes in drug exposure (Gleason et al., 2013; Thompson, 2006). This is particularly true with the use of pharmacokinetic boosters such as ritonavir (RTV), as this increases exposure times for a large number of antidepressants (Yanofski and Croarkin, 2008). Treatment with NNRTIs such as efavirenz and etravirine (ETV) can also lead to subtherapeutic concentrations of antidepressants. For example, taking trazodone in combination with ritonavir slowed clearance of trazodone and resulted in nausea, dizziness, and hypotension (Greenblatt, 2003), while in vitro studies indicate ritonavir, nelfinavir, and efavirenz all inhibited bupropion hydroxylation, suggesting the potential for increased levels of bupropion (Hesse, 2001).

The available literature on both HIV and cART altering the dopaminergic system, along with the clear dopamine-modulating properties of antidepressants (Table 1), warrants consideration of cART-antidepressant effects on the dopaminergic system. Increased or decreased exposure to these drugs through cytochrome p450 interactions could negatively impact treatment outcomes due to too much or too little production of dopamine. Timing and dosing of both types of drugs is also of major importance because dopamine can fluctuate throughout the course of infection, so poorly timed changes due to therapeutic use could render the drugs ineffective or potentiate the impact of dopamine disease progression. For example, during initial infection, enhanced dopamine availability in subcortical regions due to hypermetabolism may increase HIV replication and inflammatory cytokine production, an effect that could be potentiated by the use of therapeutics that enhance dopamine release. Or use of anti-depressants that stimulate dopamine release later in disease, during dopamine hypometabolism, may be ineffective between of the degradation of the dopaminergic system. Alternatively, a combination of cART and antidepressants reduces dopamine levels during a later stage of HIV infection could further precipitate neuropathology and create a Parkinson’s-like state in sensitive individuals. In addition, the negative effects of drugs that enhance dopamine may not be limited to the brain, as we and others have recently discussed the important role of dopamine in peripheral homeostasis (Matt and Gaskill, 2019; Rubí and Maechler, 2010; Zhang, 2017), and the effects of dopamine-modulating drugs in the periphery is very poorly understood. Increases in dopamine in peripheral regions such as the kidneys and lungs could increase viral replication and accelerate the development of HIV-related pathologies in these organs, as well as affect inflammatory pathways in peripheral organs that can signal to the CNS to cause additional damage leading to neuropsychiatric symptoms (Matt and Gaskill, 2019). Therefore, the drug combinations in the clinical management of HIV-infected patients has to be reconsidered in light of their effects on different dopaminergic circuits and the immunologic and inflammatory impact of those changes.

6. Additional considerations

The number of possible interactions between anti-depressants, cART drugs and dopamine in the aging, HIV-infected population is vast and very poorly studied, so a substantial amount of research is still needed in this area. However, there are a few considerations that are particularly important in this population. First, the nexus of these factors should be considered in the context of substance abuse, because unlike the general population where substance abuse rates decline in people over age 50, older HIV-positive patients maintain steady rates of substance abuse and dependence (Justice, 2004; Pappas and Halkitis, 2011). Chronic alcohol abuse and illicit drug use are also associated with a greater number of comorbid conditions in older HIV patients, including depression and cognitive disorders (Chiesi, 1996; Sullivan, 2011). Although described extensively elsewhere (Nolan and Gaskill, 2018; Gaskill, 2013), it is important to emphasize that exposure of HIV-infected patients to drugs of abuse also increases extracellular dopamine in the CNS (Table 1) to the levels needed to increase HIV entry and replication as well as production of cytokines, creating a synergistic negative impact on the CNS. These changes in dopamine may be associated with the higher prevalence and increased severity of cognitive dysfunction and depression also seen among drug abusers (Durvasula and Miller, 2014; Springer et al., 2009). Some research has also found enhanced sensitivity to antidepressants in active drug-abusing, HIV-infected individuals, potentially potentiating these effects. For example, an enhanced sensitivity to benzodiazepines in cocaine abusers (Volkow, 1998), SSRI-associated increases in cocaine-induced toxicity through monoaminergic mechanisms that include dopamine (O’Dell et al., 2000). Further, patients with depression and psychiatric comorbidities relating to illicit drug use are less likely to receive and respond to antidepressant treatment (DiPrete, 2019), compounding disease progression.

Second, the role of dopamine in HIV neuropathogenesis suggests that other dopamine-modulating therapeutics not prescribed for depression but often prescribed for other age-related comorbidities (Table 1) have the also have the potential for adverse interactions. For example, metoclopramide, a dopamine D2-like receptor antagonist used to treat nausea and migraines, can induce depressive and extrapyramidal symptoms in individuals with HIV (Hollander, 1985; Dahl and Diskin, 2014). New antidepressants should also be evaluated for their dopamine-associated properties and potential effects on this population. For example, ketamine, similar to esketamine, an active enantiomer of ketamine, which was recently approved by the FDA to treat depression, significantly increases dopamine and dopamine receptor levels across the rodent brain (Belujon and Grace, 2017; Kokkinou et al., 2018; Ma, 2015). Agomelatine, a potent melatonin receptor agonist and selective antagonist of the 5-HT2C receptors which is used to treat MDD (Taylor, 2014), can also increase dopamine and norepinephrine (Millan, 2003). Agomelatine increases the number and bursting of spontaneously active dopaminergic neurons (Chenu et al., 2013), with effects on neuronal plasticity in the prefrontal cortex, hippocampus, and amygdala (Dagyte, 2010; Ladurelle, 2012), which are important for modulating dopaminergic activity.

Third, to leverage multiple studies to support accurate, population level analyses, future studies need to emphasize comparability between experimental methodology. The inclusion of psychiatric disorders and standardized measures of peripheral inflammation, such as C-reactive protein or IL-6 will be tremendously useful, as will the use of rigorously validated measurements for a diagnosis. As the number of different drugs being used proliferates, especially in aging populations, a medication history listing prescribed and over the counter medications will also be invaluable, as this may lead to identification of unknown drug-drug or drug-disease interactions. And finally, to take better advantage of the data resulting from large meta-analyses, greater emphasis should be placed on expansion of in vitro mechanistic studies assessing drug-drug interactions in the context of HIV and depression, and proper validation of these studies in human populations. Human studies should take better advantage of collecting PBMCs, CSF, postmortem tissue, or plasma, and assess parallel measurements of inflammatory markers, dopamine, dopamine metabolites, or phenylalanine/tyrosine ratios, which are related to dopamine production and have been shown to be altered in HIV patients (Cassol, 2014; Zangerle, 2010; Nixon and Landay, 2010). This could allow for better understanding of patients regarding their potential responses to dopaminergic treatment regimens.

7. Conclusion

Although the prognosis of people living with HIV has improved dramatically over the past few decades, increased rates of comorbidities such as depression and neurocognitive dysfunction as this population ages poses challenges to effective treatment. In the era of polypharmacy, the increased risk of drug-related complications and exacerbation of comorbidities must be considered when choosing an appropriate cART or anti-depressant regimen. Better clinical management with careful monitoring and emphasis on personalized medicine is required, but this will require an expansion of current research, as the studies to support these requirements do not currently exist. A better understanding of dopamine-associated impact of cART, anti-depressants and other neuropsychiatric therapies is critical to understanding the association of depressive-like symptoms with particular inflammatory or neuropathogenic effects. Specific studies will be required to define the impact of therapeutic changes on dopamine in the context of the abnormal dopaminergic signaling and immune responses present older HIV-infected individuals with co-morbid depression. Further, the association of HIV and depression with inflammation and dysfunction of dopamine must be carefully considered to minimize side effects. Active consideration of all of these factors could help to reduce drug-drug interactions, ameliorate inflammation and potentially identify novel therapies for both depression and HIV-neuropathogenesis. Further, these types of studies will greatly advance our understanding of the complex interplay by which depression and NeuroHIV interact, paving the way toward the development of personalized multidisease treatment regimens.

HIGHLIGHTS.

  • Depression, and its treatment, is a major factor in the progression of HIV.

  • Depression, NeuroHIV, and their therapies impact the dopaminergic system.

  • Dopamine associated inflammation may link the etiology of NeuroHIV and depression.

  • Dopamine changes due to cART-antidepressant interaction can exacerbate NeuroHIV.

  • Managing cART-antidepressant interactions is crucial to healthy aging with HIV.

Acknowledgements

We would like to state our tremendous appreciation to all the members of the Gaskill laboratory for their insights and critical feedback during the preparation of this manuscript. This work was supported by the National Institutes of Drug Abuse, R01DA039005 (PJG) and the Brody Family Medical Trust Fund fellowship (SMM), as well as support from the Department of Pharmacology and Physiology at Drexel University College of Medicine. This content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.

Abbreviations

AD

Alzheimer’s disease

ADHD

attention deficit hyperactivity disorder

AZT

zidovudine

BBB

blood brain barrier

BH4

tetrahydrobiopterin

cART

combination antiretroviral therapy

CNS

central nervous system

CSF

cerebrospinal fluid

d4T

stavudine

DAT

dopamine transporter

DTG

dolutegravir

EFV

efavirenz

ETV

etravirine

FTC

emtricitabine

GAD

generalized anxiety disorder

HAD

HIV-associated dementia

HAND

HIV-associated neurocognitive disorder

HPA

hypothalamic-pituitary axis

HVA

homovanillic acid

IDO

indoleamine 2,3-dioxygenase

IL

interleukin

ISTI

integrase strand transfer inhibitor

LPV

lopinavir

MAO

monoamine oxidase

MDD

major depressive disorder

MDM

monocyte derived macrophages

NFV

nelfinavir

NLRP3

NLR family pyrin domain containing 3

NNRTI

non-nucleoside reverse transcriptase inhibitor

NO

nitric oxide

NPAE

neuropsychiatric adverse event

OCD

obsessive compulsive disorder

PBMC

peripheral blood mononuclear cells

PD

Parkinson’s disease

PMAT

plasma membrane monoamine transporter

RLS

Restless leg syndrome

ROS

reactive oxygen species

RTV

ritonavir

SIV

simian immunodeficiency virus

SNRI

serotonin/norepinephrine reuptake inhibitor

SSRI

selective serotonin reuptake inhibitor

TCA

tricyclic antidepressant

TDF

tenofovir disoproxil fumarate

TH

tyrosine hydroxylase

VMAT2

vesicular monoamine transporter 2

References

  1. Abebe SM, et al. , 2016. Diabetes mellitus among HIV-infected individuals in follow-up care at University of Gondar Hospital Northwest Ethiopia. BMJ Open 6 (8) e011175–e11175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Agrawal L, et al. , 2010. Dopaminergic neurotoxicity of HIV-1 gp120: reactive oxygen species as signaling intermediates. Brain Res. 1306, 116–130. [DOI] [PubMed] [Google Scholar]
  3. Ainsworth K, et al. , 1998. Effect of antidepressant drugs on dopamine D1 and D2 receptor expression and dopamine release in the nucleus accumbens of the rat. Psychopharmacology 140 (4), 470–477. [DOI] [PubMed] [Google Scholar]
  4. Akang EN, 2019. Combination antiretroviral therapy (cART)-induced hippocampal disorders: highlights on therapeutic potential of Naringenin and Quercetin. IBRO Rep. 6, 137–146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Aksenova MV, et al. , 2006. HIV-1 Tat neurotoxicity in primary cultures of rat midbrain fetal neurons: changes in dopamine transporter binding and immunoreactivity. Neurosci. Lett 395 (3), 235–239. [DOI] [PubMed] [Google Scholar]
  6. Alciati A, et al. , 2007. Major depression-related immunological changes and combination antiretroviral therapy in HIV-seropositive patients. Hum. Psychopharmacol 22 (1), 33–40. [DOI] [PubMed] [Google Scholar]
  7. Alcocer-Gomez E, et al. , 2016. Stress-Induced depressive behaviors require a functional NLRP3 inflammasome. Mol. Neurobiol 53 (7), 4874–4882. [DOI] [PubMed] [Google Scholar]
  8. Alejos B, et al. , 2014. Overall and cause-specific mortality in HIV-positive subjects compared to the general population. J. Int. AIDS Soc 17 (4S3), 19711. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Al-Harthi L, Nath A, 2019. Letter to editor. J. Neuroimmune Pharmacol 14(1), 6. [DOI] [PubMed] [Google Scholar]
  10. Aljassem K, et al. , 2016. Gender differences in severity and correlates of depression symptoms in people living with HIV in Ontario, Canada. J. Int. Assoc. Provid. AIDS Care 15 (1), 23–35. [DOI] [PubMed] [Google Scholar]
  11. Allers K, et al. , 2016. The effect of timing of antiretroviral therapy on CD4+ T-cell reconstitution in the intestine of HIV-infected patients. Mucosal Immunol. 9 (1), 265–274. [DOI] [PubMed] [Google Scholar]
  12. Althoff KN, et al. , 2015. Comparison of risk and age at diagnosis of myocardial infarction, end-stage renal disease, and non-AIDS-defining cancer in HIV-infected versus uninfected adults. Clin. Infect. Dis 60 (4), 627–638. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. American Geriatrics Society, 2012. American Geriatrics Society updated Beers Criteria for potentially inappropriate medication use in older adults. J. Am. Geriatr. Soc 60 (4), 616–631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Amu S, et al. , 2016. Dysfunctional phenotypes of CD4+ and CD8+ T cells are comparable in patients initiating ART during early or chronic HIV-1 infection. Medicine (Baltimore) 95 (23), e3738. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Anagnostakis Y, Spyraki C, 1994. Effect of morphine applied by intrapallidal microdialysis on the release of dopamine in the nucleus accumbens. Brain Res. Bull 34 (3), 275–282. [DOI] [PubMed] [Google Scholar]
  16. Andén N-E, et al. , 1967. Evidence for dopamine receptor stimulation by apomorphine. J. Pharm. Pharmacol 19 (9), 627–629. [DOI] [PubMed] [Google Scholar]
  17. Andersen SL, Gazzara RA, 1996. Effects of (−)-sulpiride on dopamine release in striatum of developing rats: degree of depolarization influences responsiveness. J. Neurochem 67 (5), 1931–1937. [DOI] [PubMed] [Google Scholar]
  18. Anderson BA, et al. , 2016. Reward, attention, and HIV-related risk in HIV+ individuals. Neurobiol. Dis 92 (Pt B), 157–165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Angelino AF, 2002. Depression and adjustment disorder in patients with HIV disease. Top. HIV Med 10 (5), 31–35. [PubMed] [Google Scholar]
  20. Ann HW, et al. , 2016. Characteristics of resting-state functional connectivity in HIV-associated neurocognitive disorder. PLoS ONE 11 (4), e0153493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Anthony IC, et al. , 2005. Influence of HAART on HIV-related CNS disease and neuroinflammation. J. Neuropathol. Exp. Neurol 64 (6), 529–536. [DOI] [PubMed] [Google Scholar]
  22. Antkiewicz-Michaluk L, Romanska I, Vetulani J, 1997. Ca2+ channel blockade prevents lysergic acid diethylamide-induced changes in dopamine and serotonin metabolism. Eur. J. Pharmacol 332 (1), 9–14. [DOI] [PubMed] [Google Scholar]
  23. Armah KA, et al. , 2012. HIV status, burden of comorbid disease, and biomarkers of inflammation, altered coagulation, and monocyte activation. Clin. Infect. Dis. 55 (1), 126–136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Arribas JR, et al. , 2008. Tenofovir disoproxil fumarate, emtricitabine, and efavirenz compared with zidovudine/lamivudine and efavirenz in treatment-naive patients: 144-week analysis. J. Acquir. Immune Defic. Syndr 47 (1), 74–78. [DOI] [PubMed] [Google Scholar]
  25. Arseniou S, Arvaniti A, Samakouri M, 2014. HIV infection and depression. Psychiatry Clin. Neurosci. 68 (2), 96–109. [DOI] [PubMed] [Google Scholar]
  26. Asch SM, et al. , 2003. Underdiagnosis of depression in HIV: who are we missing? J. Gen. Intern. Med. 18 (6), 450–460. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Ascher JA, et al. , 1995. Bupropion: a review of its mechanism of antidepressant activity. J. Clin. Psychiatry 56 (9), 395–401. [PubMed] [Google Scholar]
  28. Astuti N, Maggiolo F, 2014. Single-tablet regimens in HIV therapy. Infect. Dis. Therapy 3 (1), 1–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Avalos CR, et al. , 2017. Brain macrophages in simian immunodeficiency virus-infected, antiretroviral-suppressed macaques: a functional latent reservoir. MBio 8 (4). [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Aylward EH, et al. , 1995. Magnetic resonance imaging measurement of gray matter volume reductions in HIV dementia. Am. J. Psychiatry 152 (7), 987–994. [DOI] [PubMed] [Google Scholar]
  31. Bacopoulos NG, Redmond DE, Roth RH, 1979. Serotonin and dopamine metabolites in brain regions and cerebrospinal fluid of a primate species: effects of ketamine and fluphenazine. J. Neurochem 32 (4), 1215–1218. [DOI] [PubMed] [Google Scholar]
  32. Barclay TR, et al. , 2007. Age-associated predictors of medication adherence in HIV-positive adults: health beliefs, self-efficacy, and neurocognitive status. Health Psychol. 26 (1), 40–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Basova L, et al. , 2018. Dopamine and its receptors play a role in the modulation of CCR5 expression in innate immune cells following exposure to Methamphetamine: implications to HIV infection. PLoS ONE 13 (6), e0199861. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Bastard JP, et al. , 2015. Increased systemic immune activation and inflammatory profile of long-term HIV-infected ART-controlled patients is related to personal factors, but not to markers of HIV infection severity. J. Antimicrob. Chemother 70 (6), 1816–1824. [DOI] [PubMed] [Google Scholar]
  35. Baumann MH, Clark RD, Rothman RB, 2008. Locomotor stimulation produced by 3,4-methylenedioxymethamphetamine (MDMA) is correlated with dialysate levels of serotonin and dopamine in rat brain. Pharmacol. Biochem. Behav 90 (2), 208–217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Becker JT, et al. , 2011. Subcortical brain atrophy persists even in HAART-regulated HIV disease. Brain Imaging Behav 5 (2), 77–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Belujon P, Grace AA, 2017. Dopamine system dysregulation in major depressive disorders. Int. J. Neuropsychopharmacol 20 (12), 1036–1046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Bennett BA, Rusyniak DE, Hollingsworth CK, 1995. HIV-1 gp120-induced neurotoxicity to midbrain dopamine cultures. Brain Res. 705 (1–2), 168–176. [DOI] [PubMed] [Google Scholar]
  39. Benton T, et al. , 2010. Selective serotonin reuptake inhibitor suppression of HIV infectivity and replication. Psychosom. Med 72 (9), 925–932. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Berger JR, et al. , 1994. Cerebrospinal fluid dopamine in HIV-1 infection. AIDS 8 (1), 67–71. [DOI] [PubMed] [Google Scholar]
  41. Berger JR, Arendt G, 2000. HIV dementia: the role of the basal ganglia and dopaminergic systems. J. Psychopharmacol 14 (3), 214–221. [DOI] [PubMed] [Google Scholar]
  42. Berger JR, Nath A, 1997. HIV dementia and the basal ganglia. Intervirology 40 (2–3), 122–131. [DOI] [PubMed] [Google Scholar]
  43. Beydoun MA, et al. , 2019. Systemic inflammation is associated with depressive symptoms differentially by sex and race: a longitudinal study of urban adults. Mol. Psychiatry [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Bhatia MS, Munjal S, 2014. Prevalence of depression in people living with HIV/AIDS undergoing ART and factors associated with it. J. Clin. Diagn. Res 8 (10), p. WC01–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Bhavan KP, Kampalath VN, Overton ET, 2008. The aging of the HIV epidemic. Curr. HIV/AIDS Rep 5 (3), 150–158. [DOI] [PubMed] [Google Scholar]
  46. Bing EG, et al. , 2001. Psychiatric disorders and drug use among human immunodeficiency virus-infected adults in the United States. Arch. Gen. Psychiatry 58 (8), 721–728. [DOI] [PubMed] [Google Scholar]
  47. Bjerkenstedt L, et al. , 1970,,. Monoamine metabolite levels in cerebrospinal fluid of psychotic women treated with melperone or thiothixene. Arch Psychiatr Nervenkr 224 (2), 107–118 1977. [DOI] [PubMed] [Google Scholar]
  48. Blanco JR, et al. , 2012. Definition of advanced age in HIV infection: looking for an age cut-off. AIDS Res. Hum. Retroviruses 28 (9), 1000–1006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Bone NB, et al. , 2017. Frontline Science: D1 dopaminergic receptor signaling activates the AMPK-bioenergetic pathway in macrophages and alveolar epithelial cells and reduces endotoxin-induced ALI. J. Leukoc. Biol 101 (2), 357–365. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Bosse KE, Mathews TA, 2011. Ethanol-induced increases in extracellular dopamine are blunted in brain-derived neurotrophic factor heterozygous mice. Neurosci. Lett 489 (3), 172–176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Braganca M, Palha A, 2011. HIV associated neurocognitive disorders. Actas Esp. Psiquiatry 39 (6), 374–383. [PubMed] [Google Scholar]
  52. Brannan T, et al. , 1993. Low and high dose bromocriptine have different effects on striatal dopamine release: an in vivo study. J. Neural Transm. Park Dis. Dement. Sect 6 (2), 81–87. [DOI] [PubMed] [Google Scholar]
  53. Breitbart W, et al. , 1996. A double-blind trial of haloperidol, chlorpromazine, and lorazepam in the treatment of delirium in hospitalized AIDS patients. Am. J. Psychiatry 153 (2), 231–237. [DOI] [PubMed] [Google Scholar]
  54. Bromberg-Martin ES, Matsumoto M, Hikosaka O, 2010. Dopamine in motivational control: rewarding, aversive, and alerting. Neuron 68 (5), 815–834. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Brooks JT, et al. , 2012. HIV infection and older Americans: the public health perspective. Am. J. Public Health 102 (8), 1516–1526. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Bunney BS, Grace AA, 1978. Acute and chronic haloperidol treatment: comparison of effects on nigral dopaminergic cell activity. Life Sci. 23 (16), 1715–1727. [DOI] [PubMed] [Google Scholar]
  57. Burdo TH, et al. , 2013. Elevated sCD163 in plasma but not cerebrospinal fluid is a marker of neurocognitive impairment in HIV infection. Aids 27 (9), 1387–1395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Burgess MJ, Zeuli JD, Kasten MJ, 2015. Managementof HIV/AIDS in older patients-drug/drug interactions and adherence to antiretroviral therapy. HIV AIDS (Auckl) 7, 251–264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Burudi EM, et al. , 2002. Regulation of indoleamine 2,3-dioxygenase expression in simian immunodeficiency virus-infected monkey brains. J. Virol 76 (23), 12233–12241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Busillo JM, Azzam KM, Cidlowski JA, 2011. Glucocorticoids sensitize the innate immune system through regulation of the NLRP3 inflammasome. J. Biol. Chem 286 (44), 38703–38713. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Buzhdygan T, et al. , 2016. Neuropsychological, neurovirological and neuroimmune aspects of abnormal GABAergic transmission in HIV infection. J. Neuroimmune Pharmacol 11 (2), 279–293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Caballero J, Nahata MC, 2005. Use of selective serotonin-reuptake inhibitors in the treatment of depression in adults with HIV. Ann. Pharmacother 39 (1), 141–145. [DOI] [PubMed] [Google Scholar]
  63. Cahill S, Valadéz R, 2013. Growing older with HIV/AIDS: new public health challenges. Am. J. Public Health 103 (3), e7–e15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Campos LN, Guimaraes MD, Remien RH, 2010. Anxiety and depression symptoms as risk factors for non-adherence to antiretroviral therapy in Brazil. AIDS Behav. 14 (2), 289–299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Cañizares S, Cherner M, Ellis RJ, 2014. HIV and aging: effects on the central nervous system. Semin. Neurol 34 (1), 27–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Capuron L, et al. , 2002. Neurobehavioral effects of interferon-alpha in cancer patients: phenomenology and paroxetine responsiveness of symptom dimensions. Neuropsychopharmacology 26 (5), 643–652. [DOI] [PubMed] [Google Scholar]
  67. Carballedo A, et al. , 2011. Functional connectivity of emotional processing in depression. J. Affect. Disord 134 (1), 272–279. [DOI] [PubMed] [Google Scholar]
  68. Carroll-Anzinger D, et al. , 2007. Human immunodeficiency virus-restricted replication in astrocytes and the ability of gamma interferon to modulate this restriction are regulated by a downstream effector of the Wnt signaling pathway. J. Virol 81 (11), 5864–5871. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Carroll-Anzinger D, Al-Harthi L, 2006. Gamma interferon primes productive human immunodeficiency virus infection in astrocytes. J. Virol 80 (1), 541–544. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Carter AJ, Müller RE, 1991. Pramipexole, a dopamine D2 autoreceptor agonist, decreases the extracellular concentration of dopamine in vivo. Eur. J. Pharmacol. 200 (1), 65–72. [DOI] [PubMed] [Google Scholar]
  71. Carvalhal AS, et al. , 2003. An open trial of reboxetine in HIV-seropositive outpatients with major depressive disorder. J. Clin. Psychiatry 64 (4), 421–424. [DOI] [PubMed] [Google Scholar]
  72. Cass WA, et al. , 2003. HIV-1 protein Tat potentiation of methamphetamine-induced decreases in evoked overflow of dopamine in the striatum of the rat. Brain Res. 984 (1–2), 133–142. [DOI] [PubMed] [Google Scholar]
  73. Cassol E, et al. , 2014. Cerebrospinal fluid metabolomics reveals altered waste clearance and accelerated aging in HIV patients with neurocognitive impairment. AIDS (London, England) 28 (11), 1579–1591. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Castellon SA, et al. , 2006. Components of depression in HIV-1 infection: their differential relationship to neurocognitive performance. J. Clin. Exp. Neuropsychol 28 (3), 420–437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Cavalcante GI, et al. , 2017. HIV antiretroviral drug Efavirenz induces anxiety-like and depression-like behavior in rats: evaluation of neurotransmitter alterations in the striatum. Eur. J. Pharmacol 799, 7–15. [DOI] [PubMed] [Google Scholar]
  76. Cervo L, Grignaschi G, Samanin R, 1990. The role of the mesolimbic dopaminergic system in the desipramine effect in the forced swimming test. Eur. J. Pharmacol 178 (1), 129–133. [DOI] [PubMed] [Google Scholar]
  77. Champion MC, 1988. Domperidone. General Pharmacol. 19 (4), 499–505. [DOI] [PubMed] [Google Scholar]
  78. Chang L, et al. , 2013. Lower cognitive reserve in the aging human immunodeficiency virus-infected brain. Neurobiol. Aging 34 (4), 1240–1253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Chang C-H, Grace AA, 2014. Amygdala-ventral pallidum pathway decreases dopamine activity after chronic mild stress in rats. Biol. Psychiatry 76 (3), 223–230. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Chaudhury D, et al. , 2013. Rapid regulation of depression-related behaviours by control of midbrain dopamine neurons. Nature 493 (7433), 532–536. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Chen J, et al. , 1993. Ventral tegmental microinjection of A9-tetrahydrocannabinol enhances ventral tegmental somatodendritic dopamine levels but not forebrain dopamine levels: evidence for local neural action by marijuana’s psychoactive ingredient. Brain Res. 621 (1), 65–70. [DOI] [PubMed] [Google Scholar]
  82. Chen J, et al. , 2014. Anti-retroviral therapy decreases but does not normalize indoleamine 2,3-dioxygenase activity in HIV-infected patients. PLoS ONE 9 (7), e100446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Chenu F, El Mansari M, Blier P, 2013. Electrophysiological effects of repeated administration of agomelatine on the dopamine, norepinephrine, and serotonin systems in the rat brain. Neuropsychopharmacology 38 (2), 275–284. [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Cherner M, et al. , 2004. Effects of HIV-1 infection and aging on neurobehavioral functioning: preliminary findings. Aids 18 (Suppl. 1), S27–S34. [PubMed] [Google Scholar]
  85. Chernoloz O, El Mansari M, Blier P, 2009. Sustained administration of pramipexole modifies the spontaneous firing of dopamine, norepinephrine, and serotonin neurons in the rat brain. Neuropsychopharmacology 34 (3), 651–661. [DOI] [PubMed] [Google Scholar]
  86. Chiesi A, et al. , 1996. Epidemiology of AIDS dementia complex in Europe. AIDS in Europe Study Group. J. Acquir. Immune Defic. Syndr. Hum. Retrovirol. 11 (1), 39–44. [DOI] [PubMed] [Google Scholar]
  87. Chivero ET, et al. , 2017. HIV-1 Tat primes and activates microglial NLRP3 inflammasome-mediated neuroinflammation. J. Neurosci. 37 (13), 3599–3609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Christeff N, et al. , 1997. Serum cortisol and DHEA concentrations during HIV infection. Psychoneuroendocrinology 22 (Suppl. 1), S11–S18. [DOI] [PubMed] [Google Scholar]
  89. Christmas DM, Potokar J, Davies SJ, 2011. A biological pathway linking inflammation and depression: activation of indoleamine 2,3-dioxygenase. Neuropsychiatr. Dis. Treat 7, 431–439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Churchill MJ, et al. , 2009. Extensive astrocyte infection is prominent in human immunodeficiency virus-associated dementia. Ann. Neurol 66 (2), 253–258. [DOI] [PubMed] [Google Scholar]
  91. Clifford KM, et al. , 2017. Progressive brain atrophy despite persistent viral suppression in HIV over age 60. J. Acquir. Immune Defic. Syndr. [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Clifford KM, et al. , 2017,,. Progressive brain atrophy despite persistent viral suppression in HIV patients older than 60 years. J. Acquir. Immune Defic. Syndr 76 (3), 289–297 1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Cody SL, Vance DE, 2016. The neurobiology of HIV and its impact on cognitive reserve: a review of cognitive interventions for an aging population. Neurobiol. Dis 92, 144–156. [DOI] [PubMed] [Google Scholar]
  94. Cole MA, et al. , 2007. Relationship between psychiatric status and frontal-subcortical systems in HIV-infected individuals. J. Int. Neuropsychol. Soc 13 (3), 549–554. [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Cole JH, et al. , 2017. Increased brain-predicted aging in treated HIV disease. Neurology 88 (14), 1349–1357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Colebunders R, et al. , 2002. Neuropsychiatric reaction induced by abacavir. Am. J. Med 113 (7), 616. [DOI] [PubMed] [Google Scholar]
  97. Collazos J, et al. , 2003. Serum cortisol in HIV-infected patients with and without highly active antiretroviral therapy. Aids 17 (1), 123–126. [DOI] [PubMed] [Google Scholar]
  98. Currier MB, Molina G, Kato M, 2003. A prospective trial of sustained-release bupropion for depression in HIV-seropositive and AIDS patients. Psychosomatics 44 (2), 120–125. [DOI] [PubMed] [Google Scholar]
  99. Cysique LA, et al. , 2007. Incident major depression does not affect neuropsychological functioning in HIV-infected men. J. Int. Neuropsychol. Soc 13 (1), 1–11. [DOI] [PubMed] [Google Scholar]
  100. Czub S, et al. , 2001. Enhancement of central nervous system pathology in early simian immunodeficiency virus infection by dopaminergic drugs. Acta Neuropathol. 101 (2), 85–91. [DOI] [PubMed] [Google Scholar]
  101. Czub S, et al. , 2004. Modulation of simian immunodeficiency virus neuropathology by dopaminergic drugs. Acta Neuropathol. 107 (3), 216–226. [DOI] [PubMed] [Google Scholar]
  102. Czubak A, et al. , 2010. Effect of venlafaxine and nicotine on the level of neurotransmitters and their metabolites in rat brains. J. Physiol. Pharmacol. 61 (3), 339–346. [PubMed] [Google Scholar]
  103. Da Prada M, et al. , 1989. Neurochemical profile of moclobemide, a short-acting and reversible inhibitor of monoamine oxidase type A. J. Pharmacol. Exp. Ther 248 (1), 400–414. [PubMed] [Google Scholar]
  104. Dabaghzadeh F, et al. , 2015. Associated factors of suicidal thoughts in HIV-positive individuals. Iran. J. Psychiatry 10 (3), 185–191. [PMC free article] [PubMed] [Google Scholar]
  105. Dagyte G, et al. , 2010. The novel antidepressant agomelatine normalizes hippocampal neuronal activity and promotes neurogenesis in chronically stressed rats. CNS Neurosci. Ther 16 (4), 195–207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Dahl E, Diskin AL, 2014. Long-lasting adverse effects after short-term low-dose treatment with metoclopramide for vomiting. Int. Marit. Health 65 (1), 16–19. [DOI] [PubMed] [Google Scholar]
  107. Dailly E, et al. , 2004. Dopamine, depression and antidepressants. Fundam. Clin. Pharmacol 18 (6), 601–607. [DOI] [PubMed] [Google Scholar]
  108. Dalwadi DA, et al. , 2016. Molecular mechanisms of serotonergic action of the HIV-1 antiretroviral efavirenz. Pharmacol. Res 110, 10–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Dantzer R, et al. , 2011. Inflammation-associated depression: from serotonin to kynurenine. Psychoneuroendocrinology 36 (3), 426–436. [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Davis LE, et al. , 1992. Early viral brain invasion in iatrogenic human immunodeficiency virus infection. Neurology 42 (9), 1736–1739. [DOI] [PubMed] [Google Scholar]
  111. Davison SE, et al. , 1997. A quantitative MRI study of the Basal Ganglia in depression in HIV seropositive men. J. NeuroAIDS 1 (3), 29–41. [DOI] [PubMed] [Google Scholar]
  112. de La Fuente-Fernandez R, et al. , 2001. Apomorphine-induced changes in synaptic dopamine levels: positron emission tomography evidence for presynaptic inhibition. J. Cereb. Blood Flow Metab 21 (10), 1151–1159. [DOI] [PubMed] [Google Scholar]
  113. de Paula HHS, et al. , 2018. Reduction of inflammation and T cell activation after 6 months of cART initiation during acute, but not in early chronic HIV-1 infection. Retrovirology 15 (1), 76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Deeks SG, 2009. Immune dysfunction, inflammation, and accelerated aging in patients on antiretroviral therapy. Top. HIV Med 17 (4), 118–123. [PubMed] [Google Scholar]
  115. Deeks SG, 2011. HIV infection, inflammation, immunosenescence, and aging. Annu. Rev. Med 62, 141–155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. Defechereux PA, et al. , 2016. Depression and oral FTC/TDF pre-exposure prophylaxis (PrEP) among men and transgender women who have sex with men (MSM/TGW). AIDS Behav. 20 (7), 1478–1488. [DOI] [PMC free article] [PubMed] [Google Scholar]
  117. Delbarre B, et al. , 1995. Effect of piribedil, a D-2 dopaminergic agonist, on dopamine, amino acids, and free radicals in gerbil brain after cerebral ischemia. Mol. Chem. Neuropathol 26 (1), 43–52. [DOI] [PubMed] [Google Scholar]
  118. Delgado PL, 2000. Depression: the case for a monoamine deficiency. J. Clin. Psychiatry 61 (Suppl. 6), 7–11. [PubMed] [Google Scholar]
  119. Department of Health and Human Services, 2017. Guidelines for the Use of Antiretroviral Agents in Adults and Adolescents with HIV.
  120. Devoto P, et al. , 2012. Disulfiram stimulates dopamine release from noradrenergic terminals and potentiates cocaine-induced dopamine release in the prefrontal cortex. Psychopharmacology 219 (4), 1153–1164. [DOI] [PubMed] [Google Scholar]
  121. Di Chiara G, 2000. Role of dopamine in the behavioural actions of nicotine related to addiction. Eur. J. Pharmacol 393 (1–3), 295–314. [DOI] [PubMed] [Google Scholar]
  122. Di Chiara G, Tanda G, 1997. Blunting of reactivity of dopamine transmission to palatable food: a biochemical marker of anhedonia in the CMS model? Psychopharmacology 134 (4), 351–353 discussion 371–7. [DOI] [PubMed] [Google Scholar]
  123. Di Mascio M, et al. , 1998. Selective serotonin reuptake inhibitors reduce the spontaneous activity of dopaminergic neurons in the ventral tegmental area. Brain Res. Bull. 46 (6), 547–554. [DOI] [PubMed] [Google Scholar]
  124. di Rocco A, et al. , 2000. Decreased homovanilic acid in cerebrospinal fluid correlates with impaired neuropsychologic function in HIV-1-infected patients. Clin. Neuropharmacol. 23 (4), 190–194. [DOI] [PubMed] [Google Scholar]
  125. Diniz BS, et al. , 2019. INFLAMMATION, dopaminergic decline, and psychomotor slowing as pathologic routes to late life depression. Am. J. Geriatric Psychiatry 27 (3), S32–S33. [Google Scholar]
  126. DiPrete BL, et al. , 2019. The depression treatment cascade: disparities by alcohol use, drug use, and panic symptoms among patients in routine HIV care in the United States. AIDS Behav. 23 (3), 592–601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  127. Do AN, et al. , 2014. Excess burden of depression among HIV-infected persons receiving medical care in the united states: data from the medical monitoring project and the behavioral risk factor surveillance system. PLoS ONE 9 (3), e92842. [DOI] [PMC free article] [PubMed] [Google Scholar]
  128. Dolder CR, Patterson TL, Jeste DV, 2004. HIV, psychosis and aging: past, present and future. AIDS 18, 35–42. [PubMed] [Google Scholar]
  129. Dowlati Y, et al. , 2010. A meta-analysis of cytokines in major depression. Biol. Psychiatry 67 (5), 446–457. [DOI] [PubMed] [Google Scholar]
  130. Dremencov E, et al. , 2005. Hyperfunctionality of serotonin-2C receptor-mediated inhibition of accumbal dopamine release in an animal model of depression is reversed by antidepressant treatment. Neuropharmacology 48 (1), 34–42. [DOI] [PubMed] [Google Scholar]
  131. Dremencov E, El Mansari M, Blier P, 2009. Effects of sustained serotonin reuptake inhibition on the firing of dopamine neurons in the rat ventral tegmental area. J. Psychiatry Neurosci 34 (3), 223–229. [PMC free article] [PubMed] [Google Scholar]
  132. Drevets WC, 2000. Neuroimaging studies of mood disorders. Biol. Psychiatry 48 (8), 813–829. [DOI] [PubMed] [Google Scholar]
  133. Drew KL, et al. , 1990. Regional specific effects of clozapine and haloperidol on GABA and dopamine release in rat basal ganglia. Eur. J. Pharmacol 187 (3), 385–397. [DOI] [PubMed] [Google Scholar]
  134. Duan H, et al. , 2015. Potent and selective inhibition of plasma membrane monoamine transporter by HIV protease inhibitors. Drug Metab. Dispos 43 (11), 1773–1780. [DOI] [PMC free article] [PubMed] [Google Scholar]
  135. Dunlop BW, Nemeroff CB, 2007. The role of dopamine in the pathophysiology of depression. JAMA Psychiatry 64 (3), 327–337. [DOI] [PubMed] [Google Scholar]
  136. During MJ, Acworth IN, Wurtman RJ, 1989. Dopamine release in rat striatum: physiological coupling to tyrosine supply. J. Neurochem. 52 (5), 1449–1454. [DOI] [PubMed] [Google Scholar]
  137. Durvasula R, Miller TR, 2014. Substance abuse treatment in persons with HIV/AIDS: challenges in managing triple diagnosis. Behav. Med. (Washington, D.C.) 40, 43–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  138. Ebbinghaus M, et al. , 2012. The anti-inflammatory effects of sympathectomy in murine antigen-induced arthritis are associated with a reduction of Th1 and Th17 responses. Ann. Rheum. Dis 71 (2), 253. [DOI] [PubMed] [Google Scholar]
  139. Edelman EJ, et al. , 2013. The next therapeutic challenge in HIV: polypharmacy. Drugs Aging 30 (8), 613–628. [DOI] [PMC free article] [PubMed] [Google Scholar]
  140. Edelstein H, Knight RT, 1987. Severe parkinsonism in two AIDS patients taking prochlorperazine. Lancet 2 (8554), 341–342. [DOI] [PubMed] [Google Scholar]
  141. Effros RB, et al. , 2008. Aging and infectious diseases: workshop on HIV infection and aging: what is known and future research directions. Clin. Infect. Dis 47 (4), 542–553. [DOI] [PMC free article] [PubMed] [Google Scholar]
  142. El Mansari M, et al. , 2010. Relevance of norepinephrine-dopamine interactions in the treatment of major depressive disorder. CNS Neurosci. Ther 16 (3), e1–e17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  143. Elliott AJ, Roy-Byrne PP, 2000. Mirtazapine for depression in patients with human immunodeficiency virus. J. Clin. Psychopharmacol. 20 (2), 265–267. [DOI] [PubMed] [Google Scholar]
  144. Ellis RJ, et al. , 2010. Continued high prevalence and adverse clinical impact of human immunodeficiency virus-associated sensory neuropathy in the era of combination antiretroviral therapy: the CHARTER Study. Arch. Neurol 67 (5), 552–558. [DOI] [PMC free article] [PubMed] [Google Scholar]
  145. Elverfors A, Nissbrandt H, 1991. Reserpine-insensitive dopamine release in the substantia nigra? Brain Res. 557 (1), 5–12. [DOI] [PubMed] [Google Scholar]
  146. Emlet CA, 2006. “You’re awfully old to have this disease”: experiences of stigma and ageism in adults 50 years and older living with HIV/AIDS. Gerontologist 46 (6), 781–790. [DOI] [PubMed] [Google Scholar]
  147. Engler H, et al. , 2004. Effects of repeated social stress on leukocyte distribution in bone marrow, peripheral blood and spleen. J. Neuroimmunol. 148 (1–2), 106–115. [DOI] [PubMed] [Google Scholar]
  148. Ernst T, et al. , 2009. Declined neural efficiency in cognitively stable human immunodeficiency virus patients. Ann. Neurol 65 (3), 316–325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  149. Ersoy S, Engin VS, 2018. Risk factors for polypharmacy in older adults in a primary care setting: a cross-sectional study. Clin. Interv. Aging 13, 2003–2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  150. Eugenin EA, et al. , 2011. Human immunodeficiency virus infection ofhuman astrocytes disrupts blood-brain barrier integrity by a gap junction-dependent mechanism. J. Neurosci 31 (26), 9456–9465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  151. Eugenin EA, Berman JW, 2007. Gap junctions mediate human immunodeficiency virus-bystander killing in astrocytes. J. Neurosci. 27 (47), 12844–12850. [DOI] [PMC free article] [PubMed] [Google Scholar]
  152. Eugenin EA, Berman JW, 2013. Cytochrome C dysregulation induced by HIV infection of astrocytes results in bystander apoptosis of uninfected astrocytes by an IP3 and calcium-dependent mechanism. J. Neurochem. 127 (5), 644–651. [DOI] [PMC free article] [PubMed] [Google Scholar]
  153. Evans SR, et al. , 2007. Selegiline transdermal system (STS) for HIV-associated cognitive impairment: open-label report of ACTG 5090. HIV Clin. Trials 8 (6), 437–446. [DOI] [PubMed] [Google Scholar]
  154. Farhadian S, Patel P, Spudich S, 2017. Neurological complications of HIV infection. Curr. Infect. Dis. Rep. 19 (12), 50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  155. Fatti G, et al. , 2014. Antiretroviral treatment outcomes amongst older adults in a large multicentre cohort in South Africa. PLoS ONE 9 (6), e100273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  156. Fawcett J, et al. , 2016. Clinical experience with high-dosage pramipexole in patients with treatment-resistant depressive episodes in unipolar and bipolar depression. Am. J. Psychiatry 173 (2), 107–111. [DOI] [PubMed] [Google Scholar]
  157. Felger JC, Lotrich FE, 2013. Inflammatory cytokines in depression: neurobiological mechanisms and therapeutic implications. Neuroscience 246, 199–229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  158. Fernandes Filho SM, de Melo HR, 2012. Frequency and risk factors for HIV-associated neurocognitive disorder and depression in older individuals with HIV in northeastern Brazil. Int. Psychogeriatr. 24 (10), 1648–1655. [DOI] [PubMed] [Google Scholar]
  159. Fernstrom JD, Fernstrom MH, 2007. Tyrosine, phenylalanine, and catecholamine synthesis and function in the brain. J. Nutr. 137 (6 Suppl 1), 1539S–1547S discussion 1548S. [DOI] [PubMed] [Google Scholar]
  160. Ferrando SJ, Goldman JD, Charness WE, 1997. Selective serotonin reuptake inhibitor treatment of depression in symptomatic HIV infection and AIDS: improvements in affective and somatic symptoms. Gen. Hosp. Psychiatry 19 (2), 89–97. [DOI] [PubMed] [Google Scholar]
  161. Ferraro L, et al. , 1996. The vigilance promoting drug modafinil increases dopamine release in the rat nucleus accumbens via the involvement of a local GABAergic mechanism. Eur. J. Pharmacol. 306 (1), 33–39. [DOI] [PubMed] [Google Scholar]
  162. Fettiplace A, et al. , 2017. Psychiatric symptoms in patients receiving dolutegravir. J. Acquir. Immune Defic. Syndr. 74 (4), 423–431. [DOI] [PMC free article] [PubMed] [Google Scholar]
  163. Finberg JP, et al. , 1998. Increased striatal dopamine production from L-DOPA following selective inhibition of monoamine oxidase B by R( + )-N-propargyl-1-aminoindan (rasagiline) in the monkey. J. Neural Transm Suppl 52, 279–285. [DOI] [PubMed] [Google Scholar]
  164. Floresco SB, et al. , 2003. Afferent modulation of dopamine neuron firing differentially regulates tonic and phasic dopamine transmission. Nat. Neurosci. 6, 968. [DOI] [PubMed] [Google Scholar]
  165. Foster R, Taylor C, Everall IP, 2004. More on abacavir-induced neuropsychiatric reactions. AIDS 18 (18), 2449. [PubMed] [Google Scholar]
  166. Franceschi C, et al. , 2007. Inflammaging and anti-inflammaging: a systemic perspective on aging and longevity emerged from studies in humans. Mech. Ageing Dev 128. [DOI] [PubMed] [Google Scholar]
  167. Franceschi C, Campisi J, 2014. Chronic inflammation (inflammaging) and its potential contribution to age-associated diseases. J. Gerontol. A Biol. Sci. Med. Sci 69 (Suppl 1), S4–S9. [DOI] [PubMed] [Google Scholar]
  168. Fu X, et al. , 2011. HIV-1 Tat activates indoleamine 2,3 dioxygenase in murine organotypic hippocampal slice cultures in a p38 mitogen-activated protein kinase-dependent manner. J. Neuroinflammation 8 (1), 88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  169. Fulk LJ, et al. , 2004. Depression in HIV-infected patients: allopathic, complementary, and alternative treatments. J. Psychosom. Res. 57 (4), 339–351. [DOI] [PubMed] [Google Scholar]
  170. Fuller RW, Snoddy HD, 1981. Repeated administration of pergolide to rats attenuates the acute elevation of serum corticosterone by pergolide. Pharmacol. Biochem. Behav. 15 (6), 933–936. [DOI] [PubMed] [Google Scholar]
  171. Funderburg NT, 2014. Markers of coagulation and inflammation often remain elevated in ART-treated HIV-infected patients. Curr. Opin. HIV AIDS 9 (1), 80–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  172. Galizia I, et al. , 2016. S-adenosyl methionine (SAMe) for depression in adults. Cochrane Database Syst. Rev 10 p. Cd011286. [DOI] [PMC free article] [PubMed] [Google Scholar]
  173. Galvao-Lima LJ, et al. , 2017. Classical and alternative macrophages have impaired function during acute and chronic HIV-1 infection. Braz. J. Infect. Dis. 21 (1), 42–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  174. Gannon P, Khan MZ, Kolson DL, 2011. Current understanding of HIV-associated neurocognitive disorders pathogenesis. Curr. Opin. Neurol 24 (3), 275–283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  175. Gareri P, et al. , 2014. Use of atypical antipsychotics in the elderly: a clinical review. Clin. Interv. Aging 9, 1363–1373. [DOI] [PMC free article] [PubMed] [Google Scholar]
  176. Gaskill PJ, et al. , 2009. Human immunodeficiency virus (HIV) infection of human macrophages is increased by dopamine: a bridge between HIV-associated neurologic disorders and drug abuse. Am. J. Pathol 175 (3), 1148–1159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  177. Gaskill PJ, et al. , 2012. Characterization and function of the human macrophage dopaminergic system: implications for CNS disease and drug abuse. J. Neuroinflammation 9, 203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  178. Gaskill PJ, et al. , 2013. Drug induced increases in CNS dopamine alter monocyte, macrophage and T cell functions: implications for HAND. J. Neuroimmune Pharmacol 8 (3), 621–642. [DOI] [PMC free article] [PubMed] [Google Scholar]
  179. Gaskill PJ, et al. , 2014. Dopamine receptor activation increases HIV entry into primary human macrophages. PLoS ONE 9 (9), e108232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  180. Gatch MB, et al. , 2013. The HIV antiretroviral drug efavirenz has LSD-like properties. Neuropsychopharmacology 38 (12), 2373–2384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  181. Gay C, et al. , 2011. Cross-sectional detection of acute HIV infection: timing of transmission, inflammation and antiretroviral therapy. PLoS ONE 6 (5), e19617. [DOI] [PMC free article] [PubMed] [Google Scholar]
  182. Gay CL, et al. , 2011. Efficacy of NNRTI-based antiretroviral therapy initiated during acute HIV infection. AIDS 25 (7), 941–949. [DOI] [PMC free article] [PubMed] [Google Scholar]
  183. Gelman BB, 2015. Neuropathology of HAND With suppressive antiretroviral therapy: encephalitis and neurodegeneration reconsidered. Curr. HIV/AIDS Rep 12 (2), 272–279. [DOI] [PMC free article] [PubMed] [Google Scholar]
  184. Gelmon K, et al. , 1989. Nature, time course and dose dependence of zidovudine-related side effects: results from the Multicenter Canadian Azidothymidine Trial. Aids 3 (9), 555–561. [DOI] [PubMed] [Google Scholar]
  185. Georgiou MF, et al. , 2008. Analysis of the effects of injecting drug use and HIV-1 infection on 18F-FDG PET brain metabolism. J. Nucl. Med 49 (12), 1999–2005. [DOI] [PubMed] [Google Scholar]
  186. Gerlach M, et al. , 2004. Entacapone increases and prolongs the central effects of l-DOPA in the 6-hydroxydopamine-lesioned rat. Naunyn Schmiedebergs Arch. Pharmacol. 370 (5), 388–394. [DOI] [PubMed] [Google Scholar]
  187. Germain A, Kupfer DJ, 2008. Circadian rhythm disturbances in depression. Hum. Psychopharmacol (7), 571–585. [DOI] [PMC free article] [PubMed] [Google Scholar]
  188. Gibbie T, et al. , 2006. Depression and neurocognitive performance in individuals with HIV/AIDS: 2-year follow-up. HIV Med 7 (2), 112–121. [DOI] [PubMed] [Google Scholar]
  189. Gil-Loyzaga P, et al. , 1994. Piribedil affects dopamine turnover in cochleas stimulated by white noise. Hear. Res 79 (1–2), 178–182. [DOI] [PubMed] [Google Scholar]
  190. Ginsberg LD, Oubre AY, Daoud YA, 2011. L-methylfolate plus SSRI or SNRI from treatment initiation compared to SSRI or SNRI monotherapy in a major depressive episode. Innov. Clin. Neurosci 8 (1), 19–28. [PMC free article] [PubMed] [Google Scholar]
  191. Gleason LJ, Luque AE, Shah K, 2013. Polypharmacy in the HIV-infected older adult population. Clin. Interv. Aging 8, 749–763. [DOI] [PMC free article] [PubMed] [Google Scholar]
  192. Glennon RA, Young R, 2016. Neurobiology of 3,4-methylenedioxypyrovalerone (MDPV) and α-pyrrolidinovalerophenone (α-PVP). Brain Res. Bull 126 (Pt 1), 111–126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  193. Godfrey PS, et al. , 1990. Enhancement of recovery from psychiatric illness by methylfolate. Lancet 336 (8712), 392–395. [DOI] [PubMed] [Google Scholar]
  194. Goldsmith DR, et al. , 2016. Inflammatory markers are associated with decreased psychomotor speed in patients with major depressive disorder. Brain Behav. Immun 56, 281–288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  195. Gongvatana A, et al. , 2011. Clinical contributors to cerebral white matter integrity in HIV-infected individuals. J. Neurovirol 17 (5), 477–486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  196. Gongvatana A, et al. , 2013. Progressive cerebral injury in the setting of chronic HIV infection and antiretroviral therapy. J. Neurovirol 19 (3), 209–218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  197. Gonzalez JS, et al. , 2011,. Depression and HIV/AIDS treatment nonadherence: a review and meta-analysis. J. Acquir. Immune Defic. Syndr 58 (2), 181–187 1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  198. Goodale DB, Moore KE, 1975. Benztropine-induced release of dopamine from brain in vivo. Neuropharmacology 14 (8), 585–589. [DOI] [PubMed] [Google Scholar]
  199. Goodkin K, et al. , 2017. Effect of ageing on neurocognitive function by stage of HIV infection: evidence from the Multicenter AIDS Cohort Study. Lancet HIV 4 (9), e411–e422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  200. Goosey MW, Doggett NS, 1983. Relationship between the ability of some neuroleptics to enhance striatal [3H]dopamine release and their lipophilicity. Biochem. Pharmacol 32 (16), 2411–2416. [DOI] [PubMed] [Google Scholar]
  201. Grassi B, et al. , 1997. Efficacy of paroxetine for the treatment of depression in the context of HIV infection. Pharmacopsychiatry 30 (2), 70–71. [DOI] [PubMed] [Google Scholar]
  202. Greenbaum AH, et al. , 2008. Effect of age and HAART regimen on clinical response in an urban cohort of HIV-infected individuals. Aids 22 (17), 2331–2339. [DOI] [PMC free article] [PubMed] [Google Scholar]
  203. Greenblatt DJ, et al. , 2003. Short-term exposure to low-dose ritonavir impairs clearance and enhances adverse effects of trazodone. J. Clin. Pharmacol 43 (4), 414–422. [DOI] [PubMed] [Google Scholar]
  204. Greene M, et al. , 2014. Polypharmacy, drug-drug interactions, and potentially inappropriate medications in older adults with human immunodeficiency virus infection. J. Am. Geriatr. Soc 62 (3), 447–453. [DOI] [PMC free article] [PubMed] [Google Scholar]
  205. Greeson JM, et al. , 2016. The selective serotonin reuptake inhibitor citalopram decreases human immunodeficiency virus receptor and coreceptor expression in immune cells. Biol. Psychiatry 80 (1), 33–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  206. Grov C, et al. , 2010. Loneliness and HIV-related stigma explain depression among older HIV-positive adults. AIDS Care 22 (5), 630–639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  207. Grund B, et al. , 2016. Relevance of interleukin-6 and D-dimer for serious Non-AIDS morbidity and death among hiv-positive adults on suppressive antiretroviral therapy. PLoS ONE 11 (5), e0155100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  208. Guaraldi G, et al. , 2015. Aging with HIV vs. HIV seroconversion at older age: a diverse population with distinct comorbidity profiles. PLoS ONE 10 (4), e0118531. [DOI] [PMC free article] [PubMed] [Google Scholar]
  209. Gudelsky GA, Porter JC, 1980. Release of dopamine from tuberoinfundibular neurons into pituitary stalk blood after prolactin or haloperidol administration*. Endocrinology 106 (2), 526–529. [DOI] [PubMed] [Google Scholar]
  210. Gudelsky GA, Yamamoto BK, Nash JF, 1994. Potentiation of 3,4-methylenedioxymethamphetamine-induced dopamine release and serotonin neurotoxicity by 5-HT2 receptor agonists. Eur. J. Pharmacol 264 (3), 325–330. [DOI] [PubMed] [Google Scholar]
  211. Guiard BP, et al. , 2008. Functional interactions between dopamine, serotonin and norepinephrine neurons: an in-vivo electrophysiological study in rats with monoaminergic lesions. Int. J. Neuropsychopharmacol 11 (5), 625–639. [DOI] [PubMed] [Google Scholar]
  212. Gutiérrez F, et al. , 2014. Risk of clinically significant depression in HIV-infected patients: effect of antiretroviral drugs. HIV Med. 15 (4), 213–223. [DOI] [PubMed] [Google Scholar]
  213. Hammoud DA, et al. , 2010. Imaging serotonergic transmission with [11C]DASB-PET in depressed and non-depressed patients infected with HIV. NeuroImage 49 (3), 2588–2595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  214. Hannestad J, DellaGioia N, Bloch M, 2011. The effect of antidepressant medication treatment on serum levels of inflammatory cytokines: a meta-analysis. Neuropsychopharmacology 36 (12), 2452–2459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  215. Hartzell JD, Janke IE, Weintrob AC, 2008. Impact of depression on HIV outcomes in the HAART era. J. Antimicrob. Chemother. 62 (2), 246–255. [DOI] [PubMed] [Google Scholar]
  216. Hashimoto K, et al. , 2017. Reduction of dopamine D(2/3) receptor binding in the striatum after a single administration of esketamine, but not R-ketamine: a PET study in conscious monkeys. Eur. Arch. Psychiatry Clin. Neurosci 267 (2), 173–176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  217. Hataoka K, Kaizaki-Mitsumoto A, Numazawa S, 2017. Alpha-PVP induces the rewarding effect via activating dopaminergic neuron. J. Toxicol. Sci 42 (5), 539–543. [DOI] [PubMed] [Google Scholar]
  218. Havlik RJ, Brennan M, Karpiak SE, 2011. Comorbidities and depression in older adults with HIV. Sex Health 8 (4), 551–559. [DOI] [PubMed] [Google Scholar]
  219. Heal DJ, et al. , 2008. New perspectives from microdialysis studies in freely-moving, spontaneously hypertensive rats on the pharmacology of drugs for the treatment of ADHD. Pharmacol. Biochem. Behav 90 (2), 184–197. [DOI] [PubMed] [Google Scholar]
  220. Hemby SE, et al. , 1995. The effects of intravenous heroin administration on extracellular nucleus accumbens dopamine concentrations as determined by in vivo microdialysis. J. Pharmacol. Exp. Ther 273 (2), 591. [PubMed] [Google Scholar]
  221. Herdon H, Nahorski SR, 1987. Comparison between radiolabelled and endogenous dopamine release from rat striatal slices: effects of electrical field stimulation and regulation by D2-autoreceptors. Naunyn Schmiedebergs Arch. Pharmacol 335(3), 238–242. [DOI] [PubMed] [Google Scholar]
  222. Herrmann S, et al. , 2019. Screening for HIV-associated neurocognitive impairment: relevance of psychological factors and era of commencement of antiretroviral therapy. J. Assoc. Nurses AIDS Care 30 (1), 42–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  223. Hertel P, et al. , 1996. Risperidone: regional effects in vivo on release and metabolism of dopamine and serotonin in the rat brain. Psychopharmacology 124 (1–2), 74–86. [DOI] [PubMed] [Google Scholar]
  224. Hesse LM, et al. , 2001. Ritonavir, efavirenz, and nelfinavir inhibit CYP2B6 activity in vitro: potential drug interactions with bupropion. Drug Metab. Dispos 29 (2), 100–102. [PubMed] [Google Scholar]
  225. High KP, et al. , 2012. HIV and aging: state of knowledge and areas of critical need for research. a report to the NIH Office of AIDS Research by the HIV and Aging Working Group. J. Acquir. Immune Defic. Syndr. 60 (Suppl. 1), S1–S18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  226. Hileman CO, Funderburg NT, 2017. Inflammation, immune activation, and antiretroviral therapy in HIV. Curr. HIV/AIDS Rep 14 (3), 93–100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  227. Hill L, Lee KC, 2013. Pharmacotherapy considerations in patients with HIV and psychiatric disorders: focus on antidepressants and antipsychotics. Ann. Pharmacother. 47 (1), 75–89. [DOI] [PubMed] [Google Scholar]
  228. Hinkin CH, et al. , 2001. Methylphenidate improves HIV-1-associated cognitive slowing. J. Neuropsychiatry Clin. Neurosci 13 (2), 248–254. [DOI] [PubMed] [Google Scholar]
  229. Hobkirk AL, et al. , 2015. Psychometric validation of the BDI-II among HIV-positive CHARTER study participants. Psychol. Assess. 27 (2), 457–466. [DOI] [PMC free article] [PubMed] [Google Scholar]
  230. Hodes GE, et al. , 2015. Neuroimmune mechanisms of depression. Nat. Neurosci 18 (10), 1386–1393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  231. Hoffmann C, et al. , 2017. Higher rates of neuropsychiatric adverse events leading to dolutegravir discontinuation in women and older patients. HIV Med. 18 (1), 56–63. [DOI] [PubMed] [Google Scholar]
  232. Hollander H, et al. , 1985. Extrapyramidal symptoms in AIDS patients given low-dose metoclopramide or chlorpromazine. Lancet 2 (8465), 1186. [DOI] [PubMed] [Google Scholar]
  233. Hondo H, et al. , 1994. Effect of phencyclidine on dopamine release in the rat prefrontal cortex; an in vivo microdialysis study. Brain Res. 633 (1–2), 337–342. [DOI] [PubMed] [Google Scholar]
  234. Honeycutt JB, et al. , 2018. T cells establish and maintain CNS viral infection in HIV-infected humanized mice. J. Clin. Invest 128 (7), 2862–2876. [DOI] [PMC free article] [PubMed] [Google Scholar]
  235. Horn A, et al. , 2013. Increases in CSF dopamine in HIV patients are due to the dopamine transporter 10/10-repeat allele which is more frequent in HIV-infected individuals. J. Neural Transm. 120 (10), 1411–1419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  236. Horowitz MA, et al. , 2013. Glucocorticoids and inflammation: a double-headed sword in depression? How do neuroendocrine and inflammatory pathways interact during stress to contribute to the pathogenesis of depression? Mod Trends Pharmacopsychiatry 28, 127–143. [DOI] [PubMed] [Google Scholar]
  237. Hriso E, et al. , 1991. Extrapyramidal symptoms due to dopamine-blocking agents in patients with AIDS encephalopathy. Am. J. Psychiatry 148 (11), 1558–1561. [DOI] [PubMed] [Google Scholar]
  238. Hu S, et al. , 2009. Preferential sensitivity of human dopaminergic neurons to gp120-induced oxidative damage. J. Neurovirol 15 (5–6), 401–410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  239. Huang M, et al. , 2006. Augmentation by citalopram of risperidone-induced monoamine release in rat prefrontal cortex. Psychopharmacology 185 (3), 274–281. [DOI] [PubMed] [Google Scholar]
  240. Huck JHJ, et al. , 2015. De novo expression of dopamine D2 receptors on microglia after stroke. J. Cereb. Blood Flow Metab 35 (11), 1804–1811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  241. Hwang J, et al. , 2008. Inhibition of glial inflammatory activation and neurotoxicity by tricyclic antidepressants. Neuropharmacology 55 (5), 826–834. [DOI] [PubMed] [Google Scholar]
  242. Ichikawa J, Meltzer HY, 1995. Effect of antidepressants on striatal and accumbens extracellular dopamine levels. Eur. J. Pharmacol 281 (3), 255–261. [DOI] [PubMed] [Google Scholar]
  243. Ickovics JR, et al. , 2001. Mortality, CD4 cell count decline, and depressive symptoms among HIV-seropositive women: longitudinal analysis from the HIV Epidemiology Research Study. JAMA 285 (11), 1466–1474. [DOI] [PubMed] [Google Scholar]
  244. Ipser JC, et al. , 2015. HIV infection is associated with attenuated frontostriatal intrinsic connectivity: a preliminary study. J. Int. Neuropsychol. Soc 21 (3), 203–213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  245. Itoh K, Mehraein P, Weis S, 2000. Neuronal damage of the substantia nigra in HIV-1 infected brains. Acta Neuropathol. 99 (4), 376–384. [DOI] [PubMed] [Google Scholar]
  246. Iwata M, Ota KT, Duman RS, 2013. The inflammasome: pathways linking psychological stress, depression, and systemic illnesses. Brain Behav. Immun 31, 105–114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  247. Janssen MAM, et al. , 2017. Resting-state subcortical functional connectivity in HIV-infected patients on long-term cART. Brain Imaging Behav. 11 (5), 1555–1560. [DOI] [PMC free article] [PubMed] [Google Scholar]
  248. Jennes W, et al. , 2004. HIV-specific T helper responses and frequency of exposure among HIV-exposed seronegative female sex workers in Abidjan Cote d’Ivoire. J. Infect. Dis 189 (4), 602–610. [DOI] [PubMed] [Google Scholar]
  249. Jha MK, et al. , 2017. Interleukin 17 selectively predicts better outcomes with bupropion-SSRI combination: Novel T cell biomarker for antidepressant medication selection. Brain Behav. Immun 66, 103–110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  250. Jones SP, et al. , 2015. Expression of the kynurenine pathway in human peripheral blood mononuclear cells: implications for inflammatory and neurodegenerative disease. PLoS ONE 10 (6), e0131389. [DOI] [PMC free article] [PubMed] [Google Scholar]
  251. Joseph SB, et al. , 2018. HIV-1 RNA detected in the CNS after years of suppressive antiretroviral therapy can originate from a replicating CNS reservoir or clonally expanded cells. Clin. Infect. Dis [DOI] [PMC free article] [PubMed] [Google Scholar]
  252. Jourjy J, Dahl K, Huesgen E, 2015. Antiretroviral treatment efficacy and safety in older HIV-infected adults. Pharmacotherapy 35 (12), 1140–1151. [DOI] [PubMed] [Google Scholar]
  253. Juorio AV, Greenshaw AJ, Boulton AA, 1986. Effects of acute and chronic phenelzine on regional monoamine metabolism in rats and its potentiation by deuterium substitution. Naunyn Schmiedebergs Arch. Pharmacol 333 (3), 240–245. [DOI] [PubMed] [Google Scholar]
  254. Justice AC, et al. , 2004. Psychiatric and neurocognitive disorders among HIV-positive and negative veterans in care: veterans aging cohort five-site study. AIDS 18, 49–59. [PubMed] [Google Scholar]
  255. Kaestner F, et al. , 2012. Depression associated with antiretroviral drug therapy in HIV: case report and overview. Int. J. STD AIDS 23 (6), e14–e19. [DOI] [PubMed] [Google Scholar]
  256. Kapatos G, Zigmond M, 1977. Dopamine biosynthesis from L-tyrosine and L-phenylalanine in rat brain synaptosomes: preferential use of newly accumulated precursors. J. Neurochem 28 (5), 1109–1119. [DOI] [PubMed] [Google Scholar]
  257. Karoum F, et al. , 1994. Regional effects of amphetamine, cocaine, nomifensine and GBR 12909 on the dynamics of dopamine release and metabolism in the rat brain. Br. J. Pharmacol 113 (4), 1391–1399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  258. Kaufmann FN, et al. , 2017. NLRP3 inflammasome-driven pathways in depression: clinical and preclinical findings. Brain Behav. Immun 64, 367–383. [DOI] [PubMed] [Google Scholar]
  259. Kehr W, 1977. Effect of lisuride and other ergot derivatives on monoaminergic mechanisms in rat brain. Eur. J. Pharmacol 41 (3), 261–273. [DOI] [PubMed] [Google Scholar]
  260. Kehr J, et al. , 2007. Continuous delivery of rotigotine decreases extracellular dopamine suggesting continuous receptor stimulation. J. Neural Transm. (Vienna) 114 (8), 1027–1031. [DOI] [PubMed] [Google Scholar]
  261. Khoury AL, et al. , 2017. Diminished physical function in older HIV-infected adults in the Southeastern U.S. despite successful antiretroviral therapy. PLoS ONE 12 (6), e0179874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  262. Kita T, et al. , 2000. Methamphetamine-induced striatal dopamine release, behavior changes and neurotoxicity in BALB/c mice. Int. J. Dev. Neurosci 18 (6), 521–530. [DOI] [PubMed] [Google Scholar]
  263. Kitaichi Y, et al. , 2010. Sertraline increases extracellular levels not only of serotonin, but also of dopamine in the nucleus accumbens and striatum of rats. Eur. J. Pharmacol 647 (1–3), 90–96. [DOI] [PubMed] [Google Scholar]
  264. Kitaichi Y, et al. , 2013. Selegiline remarkably improved stage 5 treatment-resistant major depressive disorder: a case report. Neuropsychiatr. Dis. Treat. 9, 1591–1594. [DOI] [PMC free article] [PubMed] [Google Scholar]
  265. Knöchel C, et al. , 2015. Treatment-resistant late-life depression: challenges and perspectives. Curr. Neuropharmacol 13 (5), 577–591. [DOI] [PMC free article] [PubMed] [Google Scholar]
  266. Ko A, et al. , 2019. Macrophages but not astrocytes harbor HIV DNA in the brains of HIV-1-infected aviremic individuals on suppressive antiretroviral therapy. J. Neuroimmune Pharmacol 14 (1), 110–119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  267. Kohler CA, et al. , 2018. Peripheral alterations in cytokine and chemokine levels after antidepressant drug treatment for major depressive disorder: systematic review and meta-analysis. Mol. Neurobiol 55 (5), 4195–4206. [DOI] [PubMed] [Google Scholar]
  268. Kokkinou M, Ashok AH, Howes OD, 2018. The effects of ketamine on dopaminergic function: meta-analysis and review of the implications for neuropsychiatric disorders. Mol. Psychiatry 23 (1), 59–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  269. Konsman JP, Parnet P, Dantzer R, 2002. Cytokine-induced sickness behaviour: mechanisms and implications. Trends Neurosci. 25 (3), 154–159. [DOI] [PubMed] [Google Scholar]
  270. Koutsilieri E, et al. , 2002. Parkinsonism in HIV dementia. J. Neural Transm. (Vienna) 109 (5–6), 767–775. [DOI] [PubMed] [Google Scholar]
  271. Koutsilieri E, ter Meulen V, Riederer P, 2001. Neurotransmission in HIV associated dementia: a short review. J. Neural Transm 108 (6), 767–775. [DOI] [PubMed] [Google Scholar]
  272. Kraft-Terry SD, et al. , 2010. HIV-1 neuroimmunity in the era of antiretroviral therapy. Neurobiol. Dis 37 (3), 542–548. [DOI] [PMC free article] [PubMed] [Google Scholar]
  273. Kram ML, et al. , 2002. Dopamine receptors and learned helplessness in the rat: an autoradiographic study. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 26 (4), 639–645. [DOI] [PubMed] [Google Scholar]
  274. Kreisel T, et al. , 2014. Dynamic microglial alterations underlie stress-induced depressive-like behavior and suppressed neurogenesis. Mol. Psychiatry 19 (6), 699–709. [DOI] [PubMed] [Google Scholar]
  275. Krishnan KR, et al. , 2002. Comorbidity of depression with other medical diseases in the elderly. Biol. Psychiatry 52 (6), 559–588. [DOI] [PubMed] [Google Scholar]
  276. Kristiansen JE, Hansen JB, 2000. Inhibition of HIV replication by neuroleptic agents and their potential use in HIV infected patients with AIDS related dementia. Int. J. Antimicrob. Agents 14 (3), 209–213. [DOI] [PubMed] [Google Scholar]
  277. Kumar AM, et al. , 2009. Human immunodeficiency virus type 1 in the central nervous system leads to decreased dopamine in different regions of postmortem human brains. J. Neurovirol 15 (3), 257–274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  278. Kumar AM, et al. , 2011. Human immunodeficiency virus infection in the CNS and decreased dopamine availability: relationship with neuropsychological performance. J. Neurovirol. 17 (1), 26–40. [DOI] [PubMed] [Google Scholar]
  279. Ladurelle N, et al. , 2012. Agomelatine (S20098) modulates the expression of cytoskeletal microtubular proteins, synaptic markers and BDNF in the rat hippocampus, amygdala and PFC. Psychopharmacology 221 (3), 493–509. [DOI] [PubMed] [Google Scholar]
  280. Lagathu C, et al. , 2007. Some HIV antiretrovirals increase oxidative stress and alter chemokine, cytokine or adiponectin production in human adipocytes and macrophages. Antivir. Ther 12 (4), 489–500. [PubMed] [Google Scholar]
  281. Lagathu C, et al. , 2017. Basic science and pathogenesis of ageing with HIV: potential mechanisms and biomarkers. Aids 31 (Suppl. 2), S105–S119. [DOI] [PubMed] [Google Scholar]
  282. Lambert G, et al. , 2000. Reduced brain norepinephrine and dopamine release in treatment-refractory depressive illness: evidence in support of the catecholamine hypothesis of mood disorders. JAMA Psychiatry 57 (8), 787–793. [DOI] [PubMed] [Google Scholar]
  283. Lapish CC, et al. , 2009. Tolcapone enhances food-evoked dopamine efflux and executive memory processes mediated by the rat prefrontal cortex. Psychopharmacology 202 (1–3), 521–530. [DOI] [PubMed] [Google Scholar]
  284. Larsson M, et al. , 1991. Cerebrospinal fluid catecholamine metabolites in HIV-infected patients. J. Neurosci. Res. 28 (3), 406–409. [DOI] [PubMed] [Google Scholar]
  285. Launay J-M, et al. , 1988. Decreased whole blood 5-hydroxytryptamine (serotonin) in AIDS patients. JAIDS J. Acquir. Immune Defic. Syndr 1 (4), 324–325. [PubMed] [Google Scholar]
  286. Launay JM, et al. , 1989. Serotonin and human immunodeficiency viruses. Nouv. Rev. Fr. Hematol. 31 (2), 159–161. [PubMed] [Google Scholar]
  287. Lawson MA, Kelley KW, Dantzer R, 2011. Intracerebroventricular administration of HIV-1 Tat induces brain cytokine and indoleamine 2,3-dioxygenase expression: a possible mechanism for AIDS comorbid depression. Brain Behav. Immun 25 (8), 1569–1575. [DOI] [PMC free article] [PubMed] [Google Scholar]
  288. Lefkowitz RJ, 2007. Seven transmembrane receptors: a brief personal retrospective. Biochim. Biophys. Acta, Mol. Cell. Biol. Lipids 1768 (4), 748–755. [DOI] [PubMed] [Google Scholar]
  289. Leserman J, 2003. HIV disease progression: depression, stress, and possible mechanisms. Biol. Psychiatry 54 (3), 295–306. [DOI] [PubMed] [Google Scholar]
  290. Leserman J, 2008. Role of depression, stress, and trauma in HIV disease progression. Psychosom. Med 70 (5), 539–545. [DOI] [PubMed] [Google Scholar]
  291. Letendre SL, et al. , 2010. Neurologic complications of HIV disease and their treatment. Top HIV Med 18 (2), 45–55. [PMC free article] [PubMed] [Google Scholar]
  292. Levin GM, et al. , 2001. A pharmacokinetic drug-drug interaction study of venlafaxine and indinavir. Psychopharmacol. Bull 35 (2), 62–71. [PubMed] [Google Scholar]
  293. Li XM, et al. , 1998. Olanzapine increases in vivo dopamine and norepinephrine release in rat prefrontal cortex, nucleus accumbens and striatum. Psychopharmacology 136(2), 153–161. [DOI] [PubMed] [Google Scholar]
  294. Li Z, et al. , 2004. Aripiprazole, a novel antipsychotic drug, preferentially increases dopamine release in the prefrontal cortex and hippocampus in rat brain. Eur. J. Pharmacol 493 (1–3), 75–83. [DOI] [PubMed] [Google Scholar]
  295. Li W, et al. , 2011. IFN-γ mediates enhancement of HIV replication in astrocytes by inducing an antagonist of the β-catenin pathway (DKK1) in a STAT 3-dependent manner. J. Immunol 186(12), 6771. [DOI] [PMC free article] [PubMed] [Google Scholar]
  296. Li GH, Henderson L, Nath A, 2016. Astrocytes as an HIV reservoir: mechanism of HIV infection. Curr. HIV Res. 14 (5), 373–381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  297. Li Z, Ichikawa J, Meltzer HY, 2003. A comparison of the effects of loxapine with ziprasidone and thioridazine on the release of dopamine and acetylcholine in the prefrontal cortex and nucleus accumbens. Psychopharmacology 167(3), 315–323. [DOI] [PubMed] [Google Scholar]
  298. Li H, Sagar AP, Keri S, 2018. Microglial markers in the frontal cortex are related to cognitive dysfunctions in major depressive disorder. J. Affect. Disord. 241, 305–310. [DOI] [PubMed] [Google Scholar]
  299. Li H, Sagar AP, Keri S, 2018. Translocator protein (18kDa TSPO) binding, a marker of microglia, is reduced in major depression during cognitive-behavioral therapy. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 83, 1–7. [DOI] [PubMed] [Google Scholar]
  300. Lieberknecht V, et al. , 2017. Pramipexole, a dopamine D2/D3 receptor-preferring agonist, prevents experimental autoimmune encephalomyelitis development in mice. Mol. Neurobiol 54 (2), 1033–1045. [DOI] [PubMed] [Google Scholar]
  301. Lifson AR, et al. , 2008. Determination of the underlying cause of death in three multicenter international HIV clinical trials. HIV Clin. Trials 9 (3), 177–185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  302. Linner L, et al. , 2001. Reboxetine modulates the firing pattern of dopamine cells in the ventral tegmental area and selectively increases dopamine availability in the prefrontal cortex. J. Pharmacol. Exp. Ther 297 (2), 540–546. [PubMed] [Google Scholar]
  303. Lisi L, et al. , 2012. Modulatory effects of the CCR5 antagonist maraviroc on microglial pro-inflammatory activation elicited by gp120. J. Neurochem. 120 (1), 106–114. [DOI] [PubMed] [Google Scholar]
  304. Liu Y, et al. , 2004. CD4-independent infection of astrocytes by human immunodeficiency virus type 1: requirement for the human mannose receptor. J. Virol. 78 (13), 7288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  305. Liu Y, Ho RC, Mak A, 2012. Interleukin (IL)-6, tumour necrosis factor alpha (TNF-alpha) and soluble interleukin-2 receptors (sIL-2R) are elevated in patients with major depressive disorder: a meta-analysis and meta-regression. J. Affect. Disord 139(3), 230–239. [DOI] [PubMed] [Google Scholar]
  306. Llibre JM, et al. , 2019. Discontinuation of dolutegravir, elvitegravir/cobicistat and raltegravir because of toxicity in a prospective cohort. HIV Med. 20 (3), 237–247. [DOI] [PubMed] [Google Scholar]
  307. Lo J, 2011. Dyslipidemia and lipid management in HIV-infected patients. Curr. Opin. Endocrinol. Diabetes Obes 18 (2), 144–147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  308. Loland CJ, et al. , 2012. R-modafinil (armodafinil): a unique dopamine uptake inhibitor and potential medication for psychostimulant abuse. Biol. Psychiatry 72 (5), 405–413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  309. Lopez OL, et al. , 1999. Dopamine systems in human immunodeficiency virus-associated dementia. Neuropsychiatry Neuropsychol. Behav. Neurol 12 (3), 184–192. [PubMed] [Google Scholar]
  310. Ma Q, et al. , 2015. Long-term efavirenz use is associated with worse neurocognitive functioning in HIV-infected patients. J. Neurovirol. [DOI] [PMC free article] [PubMed] [Google Scholar]
  311. Maciel RA, et al. , 2018. Comorbidity is more common and occurs earlier in persons living with HIV than in HIV-uninfected matched controls, aged 50 years and older: a cross-sectional study. Int. J. Infect. Dis 70, 30–35. [DOI] [PubMed] [Google Scholar]
  312. Malaspina L, et al. , 2011. Successful cognitive aging in persons living with HIV infection. J. Neurovirol 17 (1), 110–119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  313. Mallard J, Williams K, 2018. An SIV macaque model of SIV and HAND: the need for adjunctive therapies in HIV that target activated monocytes and macrophages. J. Neurovirol. 24 (2), 213–219. [DOI] [PubMed] [Google Scholar]
  314. Mamik MK, et al. , 2017. HIV-1 viral protein R activates NLRP3 inflammasome in microglia: implications for HIV-1 associated neuroinflammation. J. Neuroimmune Pharmacol. 12 (2), 233–248. [DOI] [PubMed] [Google Scholar]
  315. Manéglier B, et al. , 2008. Serotonin decreases HIV-1 replication in primary cultures of human macrophages through 5-HT(1A) receptors. Br. J. Pharmacol 154 (1), 174–182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  316. Manfredi R, Chiodo F, 2000. A case-control study of virological and immunological effects of highly active antiretroviral therapy in HIV-infected patients with advanced age. Aids 14 (10), 1475–1477. [DOI] [PubMed] [Google Scholar]
  317. Mangoni AA, Jackson SHD, 2004. Age-related changes in pharmacokinetics and pharmacodynamics: basic principles and practical applications. Br. J. Clin. Pharmacol 57 (1), 6–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  318. Maragos WF, et al. , 2002. Human immunodeficiency virus-1 Tat protein and methamphetamine interact synergistically to impair striatal dopaminergic function. J. Neurochem 83 (4), 955–963. [DOI] [PubMed] [Google Scholar]
  319. Marks DM, Pae C-U, Patkar AA, 2008. Triple reuptake inhibitors: the next generation of antidepressants. Curr. Neuropharmacol 6 (4), 338–343. [DOI] [PMC free article] [PubMed] [Google Scholar]
  320. Martin GE, et al. , 2013. Age-associated changes in monocyte and innate immune activation markers occur more rapidly in HIV infected women. PLoS ONE 8 (1), e55279. [DOI] [PMC free article] [PubMed] [Google Scholar]
  321. Martino M, et al. , 2012. Immunomodulation mechanism of antidepressants: interactions between serotonin/norepinephrine balance and Th1/Th2 balance. Curr. Neuropharmacol 10 (2), 97–123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  322. Marzolini C, et al. , 2011. Ageing with HIV: medication use and risk for potential drug-drug interactions. J. Antimicrob. Chemother. 66 (9), 2107–2111. [DOI] [PubMed] [Google Scholar]
  323. Matsuo T, et al. , 2010. Haloperidol, spiperone, pimozide and aripiprazole reduce intracellular dopamine content in PC12 cells and rat mesencephalic cultures: Implication of inhibition of vesicular transport. Eur. J. Pharmacol 640 (1–3), 68–74. [DOI] [PubMed] [Google Scholar]
  324. Matt SM, Gaskill PJ, 2019. where is dopamine and how do immune cells see it?: Dopamine-mediated immune cell function in health and disease. J. Neuroimmune Pharmacol [DOI] [PMC free article] [PubMed] [Google Scholar]
  325. Matt SM, Johnson RW, 2016. Neuro-immune dysfunction during brain aging: new insights in microglial cell regulation. Curr. Opin. Pharmacol. 26, 96–101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  326. McLennon SM, Smith R, Orrick JJ, 2003. Recognizing and preventing drug interactions in older adults with HIV. J. Gerontol. Nurs 29 (4), 5–12. [DOI] [PubMed] [Google Scholar]
  327. McMillen BA, McDonald CC, 1983. Selective effects of buspirone and molindone on dopamine metabolism and function in the striatum and frontal cortex of the rat. Neuropharmacology 22 (3), 273–278. [DOI] [PubMed] [Google Scholar]
  328. Meltzer HY, 1989. Clinical studies on the mechanism of action of clozapine: the dopamine-serotonin hypothesis of schizophrenia. Psychopharmacology 99 (1), S18–S27. [DOI] [PubMed] [Google Scholar]
  329. Merlini E, et al. , 2016. Stimulation of PBMC and monocyte-derived macrophages via toll-like receptor activates innate immune pathways in HIV-infected patients on virally suppressive combination antiretroviral therapy. Front. Immunol 7, 614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  330. Meyer JH, et al. , 2001. Lower dopamine transporter binding potential in striatum during depression. NeuroReport 12 (18), 4121–4125. [DOI] [PubMed] [Google Scholar]
  331. Miguez-Burbano MJ, et al. , 2014. Are Serotonin Alterations the Link between Thrombocytopenia and Poor Immune Status among HIV Infected Individuals? J. AIDS Clin. Res. 2 (2). [DOI] [PMC free article] [PubMed] [Google Scholar]
  332. Milanini B, et al. , 2017. Psychiatric symptom burden in older people living with HIV with and without cognitive impairment: the UCSF HIV over 60 cohort study. AIDS Care 29 (9), 1178–1185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  333. Milanini B, Valcour V, 2017. Differentiating HIV-associated neurocognitive disorders from Alzheimer’s disease: an emerging issue in geriatric NeuroHIV. Curr. HIV/AIDS Rep 14 (4), 123–132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  334. Millan MJ, et al. , 2003. The novel melatonin agonist agomelatine (S20098) is an antagonist at 5-Hydroxytryptamine <sub >2C </sub > receptors, blockade of which enhances the activity of frontocortical dopaminergic and adrenergic pathways. J. Pharmacol. Exp. Ther 306 (3), 954. [DOI] [PubMed] [Google Scholar]
  335. Miller AL, 2008. The methylation, neurotransmitter, and antioxidant connections between folate and depression. Altern. Med. Rev 13 (3), 216–226. [PubMed] [Google Scholar]
  336. Miller AH, Raison CL, 2016. The role of inflammation in depression: from evolutionary imperative to modern treatment target. Nat. Rev. Immunol 16 (1), 22–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  337. Milner JD, Irie K, Wurtman RJ, 1986. Effects of phenylalanine on the release of endogenous dopamine from rat striatal slices. J. Neurochem 47 (5), 1444–1448. [DOI] [PubMed] [Google Scholar]
  338. Mizoguchi K, et al. , 1994. Amantadine increases the extracellular dopamine levels in the striatum by re-uptake inhibition and by N-methyl-D-aspartate antagonism. Brain Res. 662 (1–2), 255–258. [DOI] [PubMed] [Google Scholar]
  339. Mjörndal T, Persson S, 1990. Time-related changes in serum perphenazine, striatal 3H-spiperone binding and regional brain acid metabolites of dopamine and 5-hydroxy-tryptamine after a single dose of perphenazine. Fundam. Clin. Pharmacol 4 (2), 213–222. [DOI] [PubMed] [Google Scholar]
  340. Moghaddam B, Bunney BS, 1989. Differential effect of cocaine on extracellular dopamine levels in rat medial prefrontal cortex and nucleus accumbens: comparison to amphetamine. Synapse 4 (2), 156–161. [DOI] [PubMed] [Google Scholar]
  341. Mollan KR, et al. , 2014. Association between efavirenz as initial therapy for HIV-1 infection and increased risk for suicidal ideation or attempted or completed suicide: an analysis of trial data. Ann. Intern. Med. 161 (1), 1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  342. Mondal S, et al. , 2019. Low-dose maraviroc, an antiretroviral drug, attenuates the infiltration of T cells into the central nervous system and protects the nigrostriatum in hemiparkinsonian monkeys. J. Immunol p. ji1800587. [DOI] [PMC free article] [PubMed] [Google Scholar]
  343. Montoya JL, et al. , 2017. Coagulation imbalance and neurocognitive functioning in older HIV-positive adults on suppressive antiretroviral therapy. Aids 31 (6), 787–795. [DOI] [PMC free article] [PubMed] [Google Scholar]
  344. Mora F, Segovia G, del Arco A, 2008. Glutamate-dopamine-GABA interactions in the aging basal ganglia. Brain Res. Rev 58 (2), 340–353. [DOI] [PubMed] [Google Scholar]
  345. Nakayama K, 2002. Effect of paroxetine on extracellular serotonin and dopamine levels in the prefrontal cortex. Naunyn Schmiedebergs Arch. Pharmacol 365 (2), 102–105. [DOI] [PubMed] [Google Scholar]
  346. Nakayama K, Sakurai T, Katsu H, 2004. Mirtazapine increases dopamine release in prefrontal cortex by 5-HT1A receptor activation. Brain Res. Bull 63 (3), 237–241. [DOI] [PubMed] [Google Scholar]
  347. Nanni MG, et al. , 2015. Depression in HIV infected patients: a review. Curr. Psychiatry Rep 17 (1), 530. [DOI] [PubMed] [Google Scholar]
  348. Naruse M, et al. , 2018. Efficacy and safety of metyrosine in pheochromocytoma/paraganglioma: a multi-center trial in Japan. Endocr. J 65 (3), 359–371. [DOI] [PubMed] [Google Scholar]
  349. Nasi M, et al. , 2017. Ageing and inflammation in patients with HIV infection. Clin. Exp. Immunol 187 (1), 44–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  350. Nath A, et al. , 2000. Neurotoxicity and dysfunction of dopaminergic systems associated with AIDS dementia. J. Psychopharmacol 14 (3), 222–227. [DOI] [PubMed] [Google Scholar]
  351. Navia BA, et al. , 1986. The AIDS dementia complex: II. Neuropathology. Ann. Neurol 19 (6), 525–535. [DOI] [PubMed] [Google Scholar]
  352. Nazimek K, et al. , 2017. The role of macrophages in anti-inflammatory activity of antidepressant drugs. Immunobiology 222 (6), 823–830. [DOI] [PubMed] [Google Scholar]
  353. Negredo E, et al. , 2017. Aging in HIV-infected subjects: a new scenario and a new view. Biomed. Res. Int 2017, 5897298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  354. Nestler EJ, Carlezon WA Jr., 2006. The mesolimbic dopamine reward circuit in depression. Biol. Psychiatry 59 (12), 1151–1159. [DOI] [PubMed] [Google Scholar]
  355. Ng KY, et al. , 1970. L-Dopa-induced release of cerebral monoamines. Science 170 (3953), 76–77. [DOI] [PubMed] [Google Scholar]
  356. Nixon DE, Landay AL, 2010. Biomarkers of immune dysfunction in HIV. Curr. Opin. HIV AIDS 5 (6), 498–503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  357. No author, 1998. A randomized, double-blind, placebo-controlled trial of deprenyl and thioctic acid in human immunodeficiency virus-associated cognitive impairment. Dana Consortium on the Therapy of HIV Dementia and Related Cognitive Disorders. Neurology 50 (3), 645–651. [DOI] [PubMed] [Google Scholar]
  358. Nolan RA, et al. , 2018. Role of macrophage dopamine receptors in mediating cytokine production: implications for neuroinflammation in the context of HIV-associated neurocognitive disorders. J. Neuroimmune Pharmacol [DOI] [PMC free article] [PubMed] [Google Scholar]
  359. Nolan R, Gaskill PJ, 2018. The role of catecholamines in HIV neuropathogenesis. Brain Res. [DOI] [PMC free article] [PubMed] [Google Scholar]
  360. Nomikos GG, et al. , 1992. Effects of chronic bupropion on interstitial concentrations of dopamine in rat nucleus accumbens and striatum. Neuropsychopharmacology 7 (1), 7–14. [PubMed] [Google Scholar]
  361. Nutt DJ, 2008. Relationship of neurotransmitters to the symptoms of major depressive disorder. J. Clin. Psychiatry 69 (Suppl E1), 4–7. [PubMed] [Google Scholar]
  362. Nyirenda M, et al. , 2013. Prevalence and correlates of depression among HIV-infected and -affected older people in rural South Africa. J. Affect. Disord 151 (1), 31–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  363. Obuchowicz E, et al. , 2014. Imipramine and fluoxetine inhibit LPS-induced activation and affect morphology of microglial cells in the rat glial culture. Pharmacol. Rep. 66 (1), 34–43. [DOI] [PubMed] [Google Scholar]
  364. O’Dell LE, George FR, Ritz MC, 2000. Antidepressant drugs appear to enhance cocaine-induced toxicity. Exp. Clin. Psychopharmacol. 8 (1), 133–141. [DOI] [PubMed] [Google Scholar]
  365. Oishi R, et al. , 1994. Comparison of the effects of eleven histamine H1-receptor antagonists on monoamine turnover in the mouse brain. Naunyn Schmiedebergs Arch. Pharmacol 349 (2), 140–144. [DOI] [PubMed] [Google Scholar]
  366. Okada K, et al. , 1993. Effects of reserpine on the content and uptake of dopamine and noradrenaline in rabbit arteries. Clin. Exp. Pharmacol. Physiol 20 (4), 261–267. [DOI] [PubMed] [Google Scholar]
  367. Olausson P, et al. , 1998. Nefazodone attenuates the behavioral and neurochemical effects of ethanol. Alcohol 15 (1), 77–86. [DOI] [PubMed] [Google Scholar]
  368. Oleson EB, et al. , 2012. Effects of the histamine H1 receptor antagonist and benztropine analog diphenylpyraline on dopamine uptake, locomotion and reward. Eur. J. Pharmacol 683 (1–3), 161–165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  369. O’Mahony SM, et al. , 2005. Efavirenz induces depressive-like behaviour, increased stress response and changes in the immune response in rats. NeuroImmunoModulation 12 (5), 293–298. [DOI] [PubMed] [Google Scholar]
  370. Orlando G, et al. , 2006. Antiretroviral treatment and age-related comorbidities in a cohort of older HIV-infected patients. HIV Med. 7 (8), 549–557. [DOI] [PubMed] [Google Scholar]
  371. Osuji FN, et al. , 2018. The effects of highly active antiretroviral therapy on the serum levels of pro-inflammatory and anti-inflammatory cytokines in HIV infected subjects. J. Biomed. Sci. 25 (1), 88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  372. Owesson-White CA, et al. , 2012. Sources contributing to the average extracellular concentration of dopamine in the nucleus accumbens. J. Neurochem. 121 (2), 252–262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  373. Pan Y, et al. , 2014. Microglial NLRP3 inflammasome activation mediates IL-1beta-related inflammation in prefrontal cortex of depressive rats. Brain Behav. Immun. 41, 90–100. [DOI] [PubMed] [Google Scholar]
  374. Papakostas GI, et al. , 2010. S-adenosyl methionine (SAMe) augmentation of serotonin reuptake inhibitors for antidepressant nonresponders with major depressive disorder: a double-blind, randomized clinical trial. Am. J. Psychiatry 167 (8), 942–948. [DOI] [PubMed] [Google Scholar]
  375. Pappas MK, Halkitis PN, 2011. Sexual risk taking and club drug use across three age cohorts of HIV-positive gay and bisexual men in New York City. AIDS Care 23 (11), 1410–1416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  376. Pariante CM, Lightman SL, 2008. The HPA axis in major depression: classical theories and new developments. Trends Neurosci. 31 (9), 464–468. [DOI] [PubMed] [Google Scholar]
  377. Parsons LH, Schad CA, Justice JB Jr., 1993. Co-administration of the D2 antagonist pimozide inhibits up-regulation of dopamine release and uptake induced by repeated cocaine. J. Neurochem 60 (1), 376–379. [DOI] [PubMed] [Google Scholar]
  378. Pathai S, et al. , 2014. Is HIV a model of accelerated or accentuated aging? J. Gerontol. A Biol. Sci. Med. Sci 69 (7), 833–842. [DOI] [PMC free article] [PubMed] [Google Scholar]
  379. Peltenburg NC, et al. , 2018. Persistent metabolic changes in HIV-infected patients during the first year of combination antiretroviral therapy. Sci. Rep. 8 (1), 16947. [DOI] [PMC free article] [PubMed] [Google Scholar]
  380. Pence BW, et al. , 2015. The effect of antidepressant treatment on HIV and depression outcomes: results from a randomized trial. AIDS (London, England) 29 (15), 1975–1986. [DOI] [PMC free article] [PubMed] [Google Scholar]
  381. Pence BW, et al. , 2018. Association of increased chronicity of depression with HIV appointment attendance, treatment failure, and mortality among HIV-infected adults in the united statesassociation between depression and HIV treatment failure and mortalityassociation between depression and HIV treatment failure and mortality. JAMA Psychiatry 75 (4), 379–385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  382. Peringer E, et al. , 1976. Metoclopramide and dopamine receptor blockade. Neuropharmacology 15 (8), 463–469. [DOI] [PubMed] [Google Scholar]
  383. Perona MTG, et al. , 2008. Animal models of depression in dopamine, serotonin, and norepinephrine transporter knockout mice: prominent effects of dopamine transporter deletions. Behav. Pharmacol 19 (5–6), 566–574. [DOI] [PMC free article] [PubMed] [Google Scholar]
  384. Pfau ML, Menard C, Russo SJ, 2018. Inflammatory mediators in mood disorders: therapeutic opportunities. Annu. Rev. Pharmacol. Toxicol. 58, 411–428. [DOI] [PMC free article] [PubMed] [Google Scholar]
  385. Phillips PE, et al. , 2003. Subsecond dopamine release promotes cocaine seeking. Nature 422 (6932), 614–618. [DOI] [PubMed] [Google Scholar]
  386. Pinheiro CA, et al. , 2016. Depression and diagnosis of neurocognitive impairment in HIV-positive patients. Braz. J. Med. Biol. Res 49 (10), e5344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  387. Pinoli M, Marino F, Cosentino M, 2017. Dopaminergic regulation of innate immunity: a review. J. Neuroimmune Pharmacol 12 (4), 602–623. [DOI] [PubMed] [Google Scholar]
  388. Pira L, Mongeau R, Pani L, 2004. The atypical antipsychotic quetiapine increases both noradrenaline and dopamine release in the rat prefrontal cortex. Eur. J. Pharmacol 504 (1–2), 61–64. [DOI] [PubMed] [Google Scholar]
  389. Pistis M, et al. , 2002. A9-Tetrahydrocannabinol decreases extracellular GABA and increases extracellular glutamate and dopamine levels in the rat prefrontal cortex: an in vivo microdialysis study. Brain Res. 948 (1), 155–158. [DOI] [PubMed] [Google Scholar]
  390. Pizzagalli DA, et al. , 2019. Assessment of striatal dopamine transporter binding in individuals with major depressive disorder: in vivo positron emission tomography and postmortem evidence. JAMA Psychiatry. [DOI] [PMC free article] [PubMed] [Google Scholar]
  391. Plessis SD, et al. , 2014. HIV infection and the fronto-striatal system: a systematic review and meta-analysis of fMRI studies. AIDS 28 (6), 803–811. [DOI] [PubMed] [Google Scholar]
  392. Pozniak AL, et al. , 2006. Tenofovir disoproxil fumarate, emtricitabine, and efavirenz versus fixed-dose zidovudine/lamivudine and efavirenz in antiretroviral-naive patients: virologic, immunologic, and morphologic changes-a 96-week analysis. J. Acquir. Immune Defic. Syndr. 43 (5), 535–540. [DOI] [PubMed] [Google Scholar]
  393. Prevention, C.f.D.C.a., 2016. HIV among people aged 50 and older. pp. 25.
  394. Prisco S, Esposito E, 1995. Differential effects of acute and chronic fluoxetine administration on the spontaneous activity of dopaminergic neurones in the ventral tegmental area. Br. J. Pharmacol 116 (2), 1923–1931. [DOI] [PMC free article] [PubMed] [Google Scholar]
  395. Purohit V, Rapaka R, Shurtleff D, 2011. Drugs of abuse, dopamine, and HIV-associated neurocognitive disorders/HIV-associated dementia. Mol. Neurobiol 44 (1), 102–110. [DOI] [PubMed] [Google Scholar]
  396. Rabkin JG, et al. , 1994. Effect of imipramine on mood and enumerative measures of immune status in depressed patients with HIV illness. Am. J. Psychiatry 151 (4), 516–523. [DOI] [PubMed] [Google Scholar]
  397. Rabkin JG, 2008. HIV and depression: 2008 review and update. Curr. HIV/AIDS Rep 5(4), 163–171. [DOI] [PubMed] [Google Scholar]
  398. Rabkin JG, et al. , 2010. Modafinil treatment for fatigue in HIV/AIDS: a randomized placebo-controlled study. J. Clin. Psychiatry 71 (6), 707–715. [DOI] [PMC free article] [PubMed] [Google Scholar]
  399. Raedler TJ, 2011. Inflammatory mechanisms in major depressive disorder. Curr. Opin. Psychiatry 24 (6), 519–525. [DOI] [PubMed] [Google Scholar]
  400. Raison CL, et al. , 2005. Neuropsychiatric adverse effects of interferon-alpha: recognition and management. CNS Drugs 19 (2), 105–123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  401. Raison CL, Capuron L, Miller AH, 2006. Cytokines sing the blues: inflammation and the pathogenesis of depression. Trends Immunol. 27 (1), 24–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  402. Rao TSS, et al. , 2008. Understanding nutrition, depression and mental illnesses. Indian J. Psychiatry 50 (2), 77–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  403. Rappaport J, Volsky DJ, 2015. Role of the macrophage in HIV-associated neurocognitive disorders and other comorbidities in patients on effective antiretroviral treatment. J. Neurovirol 21 (3), 235–241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  404. Rasmussen LD, et al. , 2015. Time trends for risk of severe age-related diseases in individuals with and without HIV infection in Denmark: a nationwide population-based cohort study. Lancet HIV 2 (7), e288–e298. [DOI] [PubMed] [Google Scholar]
  405. Regidor DL, et al. , 2011. Effect of highly active antiretroviral therapy on biomarkers of B-lymphocyte activation and inflammation. Aids 25 (3), 303–314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  406. Renard CE, et al. , 2001. Is dopamine implicated in the antidepressant-like effects of selective serotonin reuptake inhibitors in the mouse forced swimming test? Psychopharmacology 159 (1), 42–50. [DOI] [PubMed] [Google Scholar]
  407. Reynolds JL, et al. , 2006. Proteomic analysis of the effects of cocaine on the enhancement of HIV-1 replication in normal human astrocytes (NHA). Brain Res. 1123 (1), 226–236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  408. Richards D, 2011. Prevalence and clinical course of depression: a review. Clin. Psychol. Rev 31 (7), 1117–1125. [DOI] [PubMed] [Google Scholar]
  409. Rivera-Rivera Y, et al. , 2014. Depression correlates with increased plasma levels of inflammatory cytokines and a dysregulated oxidant/antioxidant balance in HIV-1-infected subjects undergoing antiretroviral therapy. J. Clin. Cell. Immunol 5(6), 1000276. [DOI] [PMC free article] [PubMed] [Google Scholar]
  410. Rivera-Rivera Y, et al. , 2016. Impact of depression and inflammation on the progression of HIV disease. J. Clin. Cell Immunol. 7 (3). [DOI] [PMC free article] [PubMed] [Google Scholar]
  411. Rosenblat JD, Kakar R, McIntyre RS, 2015. The cognitive effects of antidepressants in major depressive disorder: a systematic review and meta-analysis of randomized clinical trials. Int. J. Neuropsychopharmacol 19 (2) p. pyv082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  412. Rottenberg DA, et al. , 1987. The metabolic pathology of the AIDS dementia complex. Ann. Neurol 22 (6), 700–706. [DOI] [PubMed] [Google Scholar]
  413. Rottenberg DA, et al. , 1996. Abnormal cerebral glucose metabolism in HIV-1 seropositive subjects with and without dementia. J. Nucl. Med 37 (7), 1133–1141. [PubMed] [Google Scholar]
  414. Rowley HL, et al. , 2012. Lisdexamfetamine and immediate release d-amfetamine – differences in pharmacokinetic/pharmacodynamic relationships revealed by striatal microdialysis in freely-moving rats with simultaneous determination of plasma drug concentrations and locomotor activity. Neuropharmacology 63 (6), 1064–1074. [DOI] [PubMed] [Google Scholar]
  415. Rubí B, Maechler P, 2010. Minireview: new roles for peripheral dopamine on metabolic control and tumor growth: let’s seek the balance. Endocrinology 151 (12), 5570–5581. [DOI] [PubMed] [Google Scholar]
  416. Rubtsova AA, et al. , 2017. Healthy aging in older women living with HIV Infection: a systematic review of psychosocial factors. Curr. HIV/AIDS Rep 14 (1), 17–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  417. Ruggiero A, et al. , 2015. During stably suppressive antiretroviral therapy integrated HIV-1 DNA load in peripheral blood is associated with the frequency of CD8 cells expressing HLA-DR/DP/DQ. EBioMedicine 2 (9), 1153–1159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  418. Ruggiero A, et al. , 2018. Factors associated with persistence of plasma HIV-1 RNA during long-term continuously suppressive firstline antiretroviral Therapy. Open Forum Infect. Dis 5 (2) p. ofy032–ofy032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  419. Russell RA, et al. , 2017. Astrocytes resist HIV-1 fusion but engulf infected macrophage material. Cell reports 18 (6), 1473–1483. [DOI] [PMC free article] [PubMed] [Google Scholar]
  420. Sacktor N, et al. , 2018. Paroxetine and fluconazole therapy for HIV-associated neurocognitive impairment: results from a double-blind, placebo-controlled trial. J. Neurovirol 24 (1), 16–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  421. Sakaue M, et al. , 2000. Postsynaptic 5-hydroxytryptamine1A receptor activation increases in vivo dopamine release in rat prefrontal cortex. Br. J. Pharmacol 129 (5), 1028–1034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  422. Salminen A, et al. , 2012. Mitochondrial dysfunction and oxidative stress activate inflammasomes: impact on the aging process and age-related diseases. Cell. Mol. Life Sci 69 (18), 2999–3013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  423. Sambamoorthi U, et al. , 2000. Antidepressant treatment and health services utilization among HIV-infected medicaid patients diagnosed with depression. J. Gen. Intern. Med 15 (5), 311–320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  424. Sandkovsky U, et al. , 2017. Impact of efavirenz pharmacokinetics and pharmacogenomics on neuropsychological performance in older HIV-infected patients. J. Antimicrob. Chemother 72 (1), 200–204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  425. Sankar A, et al. , 2011. What do we know about older adults and HIV? A review of social and behavioral literature. AIDS Care 23 (10), 1187–1207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  426. San-Martin-Clark O, et al. , 1996. Effects of trepelennamine on brain monoamine turnover in morphine dependent and abstinent mice. Psychopharmacology 123 (3), 297–302. [DOI] [PubMed] [Google Scholar]
  427. Sarchiapone M, et al. , 2006. Dopamine transporter binding in depressed patients with anhedonia. Psychiatry Res. 147 (2–3), 243–248. [DOI] [PubMed] [Google Scholar]
  428. Savetsky JB, et al. , 2001. In: Evolution of Depressive Symptoms in Human Immunodeficiency Virus-infected Patients Entering Primary Care. Lippincott Williams & Wilkins, US, pp. 76–83. [DOI] [PubMed] [Google Scholar]
  429. Saylor D, et al. , 2016. HIV-associated neurocognitive disorder-pathogenesis and prospects for treatment. Nat Rev Neurol 12 (4), 234–248. [DOI] [PMC free article] [PubMed] [Google Scholar]
  430. Schalinske K, Smazal A, 2015. Oral S-adenosylmethionine (SAM) Administration Increases Whole Brain Concentrations of Dopamine and Norepinephrine in Rats. FASEB J. 29 (1_supplement), p. 134.3. [Google Scholar]
  431. Scheller C, et al. , 2010. Increased dopaminergic neurotransmission in therapy-naive asymptomatic HIV patients is not associated with adaptive changes at the dopaminergic synapses. J. Neural Transm 117 (6), 699–705. [DOI] [PubMed] [Google Scholar]
  432. Scheper H, et al. , 2018. Severe depression as a neuropsychiatric side effect induced by dolutegravir. HIV Med 19 (4), e58–e59. [DOI] [PubMed] [Google Scholar]
  433. Schier CJ, et al. , 2017. Selective vulnerability of striatal D2 versus D1 dopamine receptor-expressing medium spiny neurons in HIV-1 Tat transgenic male mice. J. Neurosci 37 (23), 5758–5769. [DOI] [PMC free article] [PubMed] [Google Scholar]
  434. Schifitto G, et al. , 2007. A multicenter trial of selegiline transdermal system for HIV-associated cognitive impairment. Neurology 69 (13), 1314–1321. [DOI] [PubMed] [Google Scholar]
  435. Schifitto G, et al. , 2009. Selegiline and oxidative stress in HIV-associated cognitive impairment. Neurology 73 (23), 1975–1981. [DOI] [PMC free article] [PubMed] [Google Scholar]
  436. Schilstrom B, et al. , 2007. Galantamine enhances dopaminergic neurotransmission in vivo via allosteric potentiation of nicotinic acetylcholine receptors. Neuropsychopharmacology 32 (1), 43–53. [DOI] [PubMed] [Google Scholar]
  437. Schoemaker H, et al. , 1997. Neurochemical characteristics of amisulpride, an atypical dopamine D2/D3 receptor antagonist with both presynaptic and limbic selectivity. J. Pharmacol. Exp. Ther 280 (1), 83–97. [PubMed] [Google Scholar]
  438. Schouten J, et al. , 2014. Cross-sectional comparison of the prevalence of age-associated comorbidities and their risk factors between HIV-infected and uninfected individuals: the AGEhIV cohort study. Clin. Infect. Dis 59 (12), 1787–1797. [DOI] [PubMed] [Google Scholar]
  439. Schuster R, Bornovalova M, Hunt E, 2012. The influence of depression on the progression of HIV: direct and indirect effects. Behav. Modif 36 (2), 123–145. [DOI] [PubMed] [Google Scholar]
  440. Sereti I, et al. , 2017. Persistent, albeit reduced, chronic inflammation in persons starting antiretroviral therapy in acute HIV infection. Clin. Infect. Dis 64 (2), 124–131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  441. Serrão R, et al. , 2019. Non-AIDS-related comorbidities in people living with HIV-1 aged 50 years and older: The AGING POSITIVE study. Int. J. Infect. Dis 79, 94–100. [DOI] [PubMed] [Google Scholar]
  442. Shekar A, et al. , 2017. Atypical dopamine efflux caused by 3,4-methylenedioxypyrovalerone (MDPV) via the human dopamine transporter. J. Chem. Neuroanat 83–84, 69–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  443. Sheline YI, 2000. 3D MRI studies of neuroanatomic changes in unipolar major depression: the role of stress and medical comorbidity. Biol. Psychiatry 48 (8), 791–800. [DOI] [PubMed] [Google Scholar]
  444. Shi S, Klotz U, 2011. Age-related changes in pharmacokinetics. Curr. Drug Metab.. 12 (7), 601–610. [DOI] [PubMed] [Google Scholar]
  445. Shimizu SM, et al. , 2011. The impact of depressive symptoms on neuropsychological performance tests in HIV-infected individuals: a study of the hawaii aging with HIV cohort. World J. AIDS 1 (4), 139–145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  446. Shioda K, et al. , 2004. Extracellular serotonin, dopamine and glutamate levels are elevated in the hypothalamus in a serotonin syndrome animal model induced by tranylcypromine and fluoxetine. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 28(4), 633–640. [DOI] [PubMed] [Google Scholar]
  447. Shive CL, et al. , 2012. HIV-1 is not a major driver of increased plasma IL-6 levels in chronic HIV-1 disease. J. Acquir. Immune Defic. Syndr 61 (2), 145–152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  448. Shou M, et al. , 2006. Monitoring dopamine in vivo by microdialysis sampling and online CE-laser-induced fluorescence. Anal. Chem. 78 (19), 6717–6725. [DOI] [PubMed] [Google Scholar]
  449. Sidibe S, et al. , 1996. Effects of serotonin and melanin on in vitro HIV-1 infection. J. Biol. Regul. Homeost. Agents 10 (1), 19–24. [PubMed] [Google Scholar]
  450. Silveira MP, et al. , 2012. Depressive symptoms in HIV-infected patients treated with highly active antiretroviral therapy. Braz. J. Psychiatry 34 (2), 162–167. [DOI] [PubMed] [Google Scholar]
  451. Silverberg MJ, et al. , 2007. Risk of cancers during interrupted antiretroviral therapy in the SMART study. Aids 21 (14), 1957–1963. [DOI] [PubMed] [Google Scholar]
  452. Silverstone PH, Lalies MD, Hudson AL, 2012. Quetiapine and buspirone both elevate cortical levels of noradrenaline and dopamine in vivo, but do not have synergistic effects. Front. Psychiatry 3, 82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  453. Simone MJ, Appelbaum J, 2008. HIV in older adults. Geriatrics 63 (12), 6–12. [PubMed] [Google Scholar]
  454. Smit M, et al. , 2015. Future challenges for clinical care of an ageing population infected with HIV: a modelling study. Lancet Infect. Dis. 15 (7), 810–818. [DOI] [PMC free article] [PubMed] [Google Scholar]
  455. Smith JE, et al. , 2006. Self-administered heroin and cocaine combinations in the rat: additive reinforcing effects-supra-additive effects on nucleus accumbens extracellular dopamine. Neuropsychopharmacology 31 (1), 139–150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  456. Smith RC, Boggan WO, Freedman DX, 1975. Effects of single and multiple dose LSD on endogenous levels of brain tyrosine and catecholamines. Psychopharmacologia 42 (3), 271–276. [DOI] [PubMed] [Google Scholar]
  457. So-Armah KA, et al. , 2016,. Do biomarkers of inflammation, monocyte activation, and altered coagulation explain excess mortality between HIV infected and uninfected people? J. Acquir. Immune Defic. Syndr 72 (2), 206–213 1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  458. Spanagel R, Eilbacher B, Wilke R, 1994. Memantine-induced dopamine release in the prefrontal cortex and striatum of the rat-a pharmacokinetic microdialysis study. Eur. J. Pharmacol 262 (1–2), 21–26. [DOI] [PubMed] [Google Scholar]
  459. Spano JP, et al. , 2016. Non-AIDS-related malignancies: expert consensus review and practical applications from the multidisciplinary CANCERVIH Working Group. Ann. Oncol 27 (3), 397–408. [DOI] [PubMed] [Google Scholar]
  460. Sperner-Unterweger B, Kohl C, Fuchs D, 2014. Immune changes and neurotransmitters: possible interactions in depression? Prog. Neuro-Psychopharmacol. Biol. Psychiatry 48, 268–276. [DOI] [PubMed] [Google Scholar]
  461. Spies G, Asmal L, Seedat S, 2013. Cognitive-behavioural interventions for mood and anxiety disorders in HIV: a systematic review. J. Affect. Disord. 150 (2), 171–180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  462. Springer SA, Chen S, Altice F, 2009. Depression and symptomatic response among HIV-infected drug users enrolled in a randomized controlled trial of directly administered antiretroviral therapy. AIDS care 21 (8), 976–983. [DOI] [PMC free article] [PubMed] [Google Scholar]
  463. Spudich S, et al. , 2011. Central nervous system immune activation characterizes primary human immunodeficiency virus 1 infection even in participants with minimal cerebrospinal fluid viral burden. J. Infect. Dis 204 (5), 753–760. [DOI] [PMC free article] [PubMed] [Google Scholar]
  464. Spudich SS, 2016. Immune activation in the central nervous system throughout the course of HIV infection. Curr. Opin. HIV AIDS 11 (2), 226–233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  465. Spudich S, et al. , 2019. Persistent HIV-infected cells in cerebrospinal fluid are associated with poorer neurocognitive performance. J. Clin. Invest 130, 3339–3346. [DOI] [PMC free article] [PubMed] [Google Scholar]
  466. Stamford JA, Kruk ZL, Millar J, 1986. Sub-second striatal dopamine release measured by in vivo voltammetry. Brain Res. 381 (2), 351–355. [DOI] [PubMed] [Google Scholar]
  467. Starke K, et al. , 1978. Effect of dopamine receptor agonists and antagonists on release of dopamine in the rabbit caudate nucleus in vitro. Naunyn Schmiedebergs Arch. Pharmacol 305 (1), 27–36. [DOI] [PubMed] [Google Scholar]
  468. Strawbridge R, et al. , 2015. Inflammation and clinical response to treatment in depression: a meta-analysis. Eur. Neuropsychopharmacol. 25 (10), 1532–1543. [DOI] [PubMed] [Google Scholar]
  469. Subbaiah MAM, 2018. Triple reuptake inhibitors as potential therapeutics for depression and other disorders: design paradigm and developmental challenges. J. Med. Chem. 61 (6), 2133–2165. [DOI] [PubMed] [Google Scholar]
  470. Sullivan LE, et al. , 2011. Alcohol consumption and depressive symptoms over time: a longitudinal study of patients with and without HIV infection. Drug Alcohol Depend. 117 (2–3), 158–163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  471. Swendeman D, et al. , 2018. Gender disparities in depression severity and coping among people living with HIV/AIDS in Kolkata, India. PLoS ONE 13 (11), e0207055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  472. Szadkowski L, et al. , 2012. Short communication: effects of age on virologic suppression and CD4 cell response in HIV-positive patients initiating combination antiretroviral therapy. AIDS Res. Hum. Retroviruses 28 (12), 1579–1583. [DOI] [PubMed] [Google Scholar]
  473. Tavazzi E, et al. , 2014. Brain inflammation is a common feature of HIV-infected patients without HIV encephalitis or productive brain infection. Curr. HIV Res 12 (2), 97–110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  474. Tavoschi L, et al. , 2017. New HIV diagnoses among adults aged 50 years or older in 31 European countries, 2004–15: an analysis of surveillance data. Lancet HIV 4 (11), e514–e521. [DOI] [PubMed] [Google Scholar]
  475. Taylor G, et al. , 1996. Male reproductive systems under chronic fluoxetine or trimipramine treatment. Physiol. Behav 59 (3), 479–485. [DOI] [PubMed] [Google Scholar]
  476. Taylor D, et al. , 2014. Antidepressant efficacy of agomelatine: meta-analysis of published and unpublished studies. BMJ 348, g1888. [DOI] [PMC free article] [PubMed] [Google Scholar]
  477. Tedaldi EM, Minniti NL, Fischer T, 2015. HIV-associated neurocognitive disorders: the relationship of HIV infection with physical and social comorbidities. Biomed Res. Int 2015, 641913. [DOI] [PMC free article] [PubMed] [Google Scholar]
  478. Teeraananchai S, et al. , 2017. Life expectancy of HIV-positive people after starting combination antiretroviral therapy: a meta-analysis. HIV Med 18 (4), 256–266. [DOI] [PubMed] [Google Scholar]
  479. Thompson A, et al. , 2006. Psychotropic medications and HIV. Clin. Infect. Dis 42 (9), 1305–1310. [DOI] [PubMed] [Google Scholar]
  480. Tierney SM, et al. , 2019. Extrapyramidal motor signs in older adults with HIV disease: frequency, 1-year course, and associations with activities of daily living and quality of life. J. Neurovirol 25 (2), 162–173. [DOI] [PubMed] [Google Scholar]
  481. Tong L, et al. , 2017. Microglia Loss contributes to the development of major depression induced by different types of chronic stresses. Neurochem. Res 42 (10), 2698–2711. [DOI] [PubMed] [Google Scholar]
  482. Tor-Agbidye J, Yamamoto B, Bowyer JF, 2001. Seizure activity and hyperthermia potentiate the increases in dopamine and serotonin extracellular levels in the amygdala during exposure to d-amphetamine. Toxicol. Sci 60 (1), 103–111. [DOI] [PubMed] [Google Scholar]
  483. Torgersen J, et al. , 2019. Impact of efavirenz metabolism on loss to care in older HIV + Africans. Eur. J. Drug Metab. Pharmacokinet 44 (2), 179–187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  484. Triant VA, 2013. Cardiovascular disease and HIV infection. Curr. HIV/AIDS Rep 10 (3), 199–206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  485. Tsai AC, et al. , 2010. A marginal structural model to estimate the causal effect of antidepressant medication treatment on viral suppression among homeless and marginally housed persons with HIV. Arch. Gen. Psychiatry 67 (12), 1282–1290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  486. Tunbridge EM, et al. , 2004. Catechol-< em >O< /em > -methyltransferase inhibition improves set-shifting performance and elevates stimulated dopamine release in the rat prefrontal cortex. J. Neurosci 24 (23), 5331. [DOI] [PMC free article] [PubMed] [Google Scholar]
  487. Tye KM, et al. , 2012. Dopamine neurons modulate neural encoding and expression of depression-related behaviour. Nature 493, 537. [DOI] [PMC free article] [PubMed] [Google Scholar]
  488. UNAIDS, 2013. HIV and aging: A special supplement to the UNAIDS report on the global AIDS epidemic 2013.
  489. Underwood J, Winston A, 2016. Guidelines for evaluation and management of cognitive disorders in HIV-positive individuals. Curr. HIV/AIDS Rep 13 (5), 235–240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  490. Valcour VG, et al. , 2004. Cognitive impairment in older HIV-1-seropositive individuals: prevalence and potential mechanisms. AIDS (London, England) 18 (Suppl 1), S79–S86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  491. Valcour V, et al. , 2004. Higher frequency of dementia in older HIV-1 individuals: the Hawaii Aging with HIV-1 Cohort. Neurology 63 (5), 822–827. [DOI] [PMC free article] [PubMed] [Google Scholar]
  492. Valcour V, et al. , 2008. Aging exacerbates extrapyramidal motor signs in the era of highly active antiretroviral therapy. J. Neurovirol 14 (5), 362–367. [DOI] [PMC free article] [PubMed] [Google Scholar]
  493. Valcour V, et al. , 2012. Central nervous system viral invasion and inflammation during acute HIV infection. J. Infect. Dis 206 (2), 275–282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  494. van der Lee MJ, et al. , 2007. Interaction study of the combined use of paroxetine and fosamprenavir-ritonavir in healthy subjects. Antimicrob. Agents Chemother 51 (11), 4098–4104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  495. van Gorp WG, et al. , 1992. Cerebral metabolic dysfunction in AIDS: findings in a sample with and without dementia. J. Neuropsychiatry Clin. Neurosci. 4 (3), 280–287. [DOI] [PubMed] [Google Scholar]
  496. Vance DE, et al. , 2011. Aging with HIV: a cross-sectional study of comorbidity prevalence and clinical characteristics across decades of life. J. Assoc. Nurses AIDS Care 22 (1), 17–25. [DOI] [PubMed] [Google Scholar]
  497. Vander Weele CM, et al. , 2014. Rapid dopamine transmission within the nucleus accumbens: dramatic difference between morphine and oxycodone delivery. Eur. J. Neurosci 40 (7), 3041–3054. [DOI] [PMC free article] [PubMed] [Google Scholar]
  498. Vazquez-Santiago FJ, et al. , 2014. Glutamate metabolism and HIV-associated neurocognitive disorders. J. Neurovirol. 20 (4), 315–331. [DOI] [PMC free article] [PubMed] [Google Scholar]
  499. Venerosi A, et al. , 2005. Neurobehavioral effects of prenatal exposure to AZT: a preliminary investigation with the D1 receptor agonist SKF 38393 in mice. Neurotoxicol. Teratol 27 (1), 169–173. [DOI] [PubMed] [Google Scholar]
  500. Volkow ND, et al. , 1998. Enhanced sensitivity to benzodiazepines in active cocaine-abusing subjects: a PET study. Am. J. Psychiatry 155 (2), 200–206. [DOI] [PubMed] [Google Scholar]
  501. Volkow ND, et al. , 2001. Therapeutic doses of oral methylphenidate significantly increase extracellular dopamine in the human brain. J. Neurosci. 21 (2), p. RC121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  502. Von Vigtlander PF, Moore KE, 1971. Dopamine: release from the brain in vivo by amantadine. Science 174 (4007), 408. [DOI] [PubMed] [Google Scholar]
  503. Wada NI, et al. , 2015. The effect of HAART-induced HIV suppression on circulating markers of inflammation and immune activation. AIDS (London, England) 29 (4), 463–471. [DOI] [PMC free article] [PubMed] [Google Scholar]
  504. Wagner GJ, Rabkin R, 2000. Effects of dextroamphetamine on depression and fatigue in men with HIV: a double-blind, placebo-controlled trial. J. Clin. Psychiatry 61 (6), 436–440. [DOI] [PubMed] [Google Scholar]
  505. Walkup J, et al. , 2008. Antidepressant treatment and adherence to combination antiretroviral therapy among patients with AIDS and diagnosed depression. Psychiatr. Q 79 (1), 43–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  506. Walsh JG, et al. , 2014. Rapid inflammasome activation in microglia contributes to brain disease in HIV/AIDS. Retrovirology 11, 35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  507. Wang GJ, et al. , 2004. Decreased brain dopaminergic transporters in HIV-associated dementia patients. Brain 127 (Pt 11), 2452–2458. [DOI] [PubMed] [Google Scholar]
  508. Wang Z, et al. , 2018. Critical role of NLRP3-caspase-1 pathway in age-dependent isoflurane-induced microglial inflammatory response and cognitive impairment. J. Neuroinflammation 15 (1), 109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  509. Wang T, et al. , 2018. Prevalence of depression or depressive symptoms among people living with HIV/AIDS in China: a systematic review and meta-analysis. BMC Psychiatry 18 (1), 160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  510. Warner-Schmidt JL, et al. , 2011. Antidepressant effects of selective serotonin reuptake inhibitors (SSRIs) are attenuated by antiinflammatory drugs in mice and humans. Proc. Natl. Acad. Sci. U.S.A 108 (22), 9262–9267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  511. Watkins CC, Pieper AA, Treisman GJ, 2011. Safety considerations in drug treatment of depression in HIV-positive patients. Drug Saf. 34 (8), 623–639. [DOI] [PubMed] [Google Scholar]
  512. Watkins CC, Treisman GJ, 2012. Neuropsychiatric complications of aging with HIV. J. Neurovirol 18 (4), 277–290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  513. Watters MR, et al. , 2004. Symptomatic distal sensory polyneuropathy in HIV after age 50. Neurology 62 (8), 1378–1383. [DOI] [PubMed] [Google Scholar]
  514. Weiss F, et al. , 1996. Ethanol self-administration restores withdrawal-associated deficiencies in accumbal dopamine and 5-hydroxytryptamine release in dependent rats. J. Neurosci. 16 (10), 3474–3485. [DOI] [PMC free article] [PubMed] [Google Scholar]
  515. Wellons MF, et al. , 2002. HIV infection: treatment outcomes in older and younger adults. J. Am. Geriatr. Soc 50 (4), 603–607. [DOI] [PubMed] [Google Scholar]
  516. Wendelken LA, Valcour V, 2012. Impact of HIV and aging on neuropsychological function. J. NeuroVirol. 18 (4), 256–263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  517. WHO, 2018. Ageing and health.
  518. Wiedlocha M, et al. , 2018. Effect of antidepressant treatment on peripheral inflammation markers - a meta-analysis. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 80 (Pt C), 217–226. [DOI] [PubMed] [Google Scholar]
  519. Wightman RM, et al. , 2007. Dopamine release is heterogeneous within microenvironments of the rat nucleus accumbens. Eur. J. Neurosci. 26 (7), 2046–2054. [DOI] [PubMed] [Google Scholar]
  520. Williams DW, et al. , 2014. Monocytes mediate HIV neuropathogenesis: mechanisms that contribute to HIV associated neurocognitive disorders. Curr. HIV Res [DOI] [PMC free article] [PubMed] [Google Scholar]
  521. Williams KC, Hickey WF, 2002. Central nervous system damage, monocytes and macrophages, and neurological disorders in AIDS. Annu. Rev. Neurosci 25, 537–562. [DOI] [PubMed] [Google Scholar]
  522. Wilson JM, et al. , 1996. Striatal dopamine nerve terminal markers in human, chronic methamphetamine users. Nat. Med 2 (6), 699–703. [DOI] [PubMed] [Google Scholar]
  523. Wing VC, et al. , 2015. Measuring cigarette smoking-induced cortical dopamine release: A [11C]FLB-457 PET study. Neuropsychopharmacology 40 (6), 1417–1427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  524. Wing EJ, 2016. HIV and aging. Int. J. Infect. Dis 53, 61–68. [DOI] [PubMed] [Google Scholar]
  525. Winston A, Underwood J, 2015. Emerging concepts on the use of antiretroviral therapy in older adults living with HIV infection. Curr. Opin. Infect. Dis. 28 (1), 17–22. [DOI] [PubMed] [Google Scholar]
  526. Wohleb ES, et al. , 2016. Integrating neuroimmune systems in the neurobiology of depression. Nat. Rev. Neurosci. 17 (8), 497–511. [DOI] [PubMed] [Google Scholar]
  527. Wong ML, et al. , 2016. Inflammasome signaling affects anxiety- and depressive-like behavior and gut microbiome composition. Mol. Psychiatry 21 (6), 797–805. [DOI] [PMC free article] [PubMed] [Google Scholar]
  528. Wright JM, et al. , 1989. Zidovudine-related mania. Med. J. Aust 150 (6), 339–341. [DOI] [PubMed] [Google Scholar]
  529. Yadav A, Collman RG, 2009. CNS inflammation and macrophage/microglial biology associated with HIV-1 infection. J. Neuroimmune Pharmacol 4 (4), 430–447. [DOI] [PMC free article] [PubMed] [Google Scholar]
  530. Yahr MD, et al. , 1969. Treatment of parkinsonism with levodopa. Arch. Neurol. 21 (4), 343–354. [DOI] [PubMed] [Google Scholar]
  531. Yamamoto S, et al. , 2016. Haloperidol suppresses NF-kappaB to inhibit lipopoly-saccharide-induced pro-inflammatory response in RAW 264 cells. Med. Sci. Monit. 22, 367–372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  532. Yan Q-S, 1999. Extracellular dopamine and serotonin after ethanol monitored with 5-minute microdialysis. Alcohol 19 (1), 1–7. [DOI] [PubMed] [Google Scholar]
  533. Yan Y, et al. , 2015. Dopamine Controls Systemic Inflammation through Inhibition of NLRP3 Inflammasome. Cell 160 (1), 62–73. [DOI] [PubMed] [Google Scholar]
  534. Yanofski J, Croarkin P, 2008. Choosing antidepressants for HIV and AIDS patients: insights on safety and side effects. Psychiatry (Edgmont) 5 (5), 61–66. [PMC free article] [PubMed] [Google Scholar]
  535. Yirmiya R, Rimmerman N, Reshef R, 2015. Depression as a microglial disease. Trends Neurosci. 38 (10), 637–658. [DOI] [PubMed] [Google Scholar]
  536. Youngren KD, et al. , 1999. Clozapine preferentially increases dopamine release in the rhesus monkey prefrontal cortex compared with the caudate nucleus. Neuropsychopharmacology 20 (5), 403–412. [DOI] [PubMed] [Google Scholar]
  537. Yuan L, et al. , 2013. Cytokines in CSF correlate with HIV-associated neurocognitive disorders in the post-HAART era in China. J. Neurovirol. 19 (2), 144–149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  538. Yun LW, et al. , 2005. Antidepressant treatment improves adherence to antiretroviral therapy among depressed HIV-infected patients. J. Acquir. Immune Defic. Syndr. 38 (4), 432–438. [DOI] [PubMed] [Google Scholar]
  539. Zangerle R, et al. , 2010. Increased blood phenylalanine to tyrosine ratio in HIV-1 infection and correction following effective antiretroviral therapy. Brain Behav. Immun 24 (3), 403–408. [DOI] [PubMed] [Google Scholar]
  540. Zanjani F, Saboe K, Oslin D, 2007. Age difference in rates of mental health/substance abuse and behavioral care in HIV-positive adults. AIDS Patient Care STDS 21 (5), 347–355. [DOI] [PubMed] [Google Scholar]
  541. Zapata HJ, Shaw AC, 2014. Aging of the human innate immune system in HIV infection. Curr. Opin. Immunol 29, 127–136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  542. Zeevi N, et al. , 2010. The blood-brain barrier: geriatric relevance of a critical brain-body interface. J. Am. Geriatr. Soc 58 (9), 1749–1757. [DOI] [PMC free article] [PubMed] [Google Scholar]
  543. Zhang W, et al. , 2000. Synergistic effects of olanzapine and other antipsychotic agents in combination with fluoxetine on norepinephrine and dopamine release in rat prefrontal cortex. Neuropsychopharmacology 23 (3), 250–262. [DOI] [PubMed] [Google Scholar]
  544. Zhang Y, et al. , 2009. Behavioral and neurochemical changes induced by oxycodone differ between adolescent and adult mice. Neuropsychopharmacology 34 (4), 912–922. [DOI] [PMC free article] [PubMed] [Google Scholar]
  545. Zhang X, et al. , 2017. Potential roles of peripheral dopamine in tumor immunity. J. Cancer 8 (15), 2966–2973. [DOI] [PMC free article] [PubMed] [Google Scholar]
  546. Zhang L, Zhou FM, Dani JA, 2004. Cholinergic drugs for Alzheimer’s disease enhance in vitro dopamine release. Mol. Pharmacol. 66 (3), 538–544. [DOI] [PubMed] [Google Scholar]
  547. Zhou X, et al. , 2015. Atypical antipsychotic augmentation for treatment-resistant depression: a systematic review and network meta-analysis. J. Neuropsychopharmacol 18 (11) p. pyv060–pyv060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  548. Zhu J, et al. , 2009. HIV-1 Tat protein-induced rapid and reversible decrease in [3H] dopamine uptake: dissociation of [3H]dopamine uptake and [3H]2beta-carbomethoxy-3-beta-(4-fluorophenyl)tropane (WIN 35,428) binding in rat striatal synaptosomes. J. Pharmacol. Exp. Ther. 329 (3), 1071–1083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  549. Zhu J, et al. , 2011. Recombinant human immunodeficiency virus-1 transactivator of transcription1–86 allosterically modulates dopamine transporter activity. Synapse 65 (11), 1251–1254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  550. Zhu J, et al. , 2016. HIV-1 transgenic rats display an increase in [(3)H]dopamine uptake in the prefrontal cortex and striatum. J. Neurovirol 22 (3), 282–292. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES