Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 May 29.
Published in final edited form as: Neurosci Lett. 2021 Mar 29;754:135863. doi: 10.1016/j.neulet.2021.135863

HIV-1 and drug abuse comorbidity: Lessons learned from the animal models of NeuroHIV

Susmita Sil 1,, Annadurai Thangaraj 1,, Ernest T Chivero 1, Fang Niu 1, Muthukumar Kannan 1, Ke Liao 1, Peter S Silverstein 2, Palsamy Periyasamy 1,*, Shilpa Buch 1,*
PMCID: PMC8108725  NIHMSID: NIHMS1692830  PMID: 33794296

Abstract

Various research studies that have investigated the association between HIV infection and addiction underpin the role of various drugs of abuse in impairing immunological and non-immunological pathways of the host system, ultimately leading to augmentation of HIV infection and disease progression. These studies have included both in vitro and in vivo animal models wherein investigators have assessed the effects of various drugs on several disease parameters to decipher the impact of drugs on both HIV infection and progression of HIV-associated neurocognitive disorders (HAND). However, given the inherent limitations in the existing animal models of HAND, these investigations only recapitulated specific aspects of the disease but not the complex human syndrome. Despite the inability of HIV to infect rodents over the last 30 years, multiple strategies have been employed to develop several rodent models of HAND. While none of these models can accurately mimic the overall pathophysiology of HAND, they serve the purpose of modeling some unique aspects of HAND. This review provides an overview of various animal models used in the field and a careful evaluation of methodological strengths and limitations inherent in both the model systems and study designs to understand better how the various animal models complement one another.

Keywords: HIV, drug abuse, HAND, animal models, SIV, rhesus macaques

1. Introduction

Despite the ability of combination antiretroviral therapy (cART) to dramatically suppress viremia, the brain continues to be a reservoir for low-level HIV-1 replication [1-6]. It has been estimated that ~30-60% of infected individuals on the cART regimen go on to develop a varying degree of neurological dysfunction ranging from asymptomatic to mild neurocognitive impairments, collectively termed as HIV-associated neurocognitive disorders (HAND) or NeuroHIV, that significantly affects the quality of life of infected individuals [7-12]. Adding further complexity to HAND is the co-morbidity of drug abuse [13-17]. The Centers for Disease Control and Prevention define HIV-1 infection and drug abuse as intertwined epidemics, leading to compromised cART adherence and consequent exacerbation of HAND. The interplay of HIV-1 and drug abuse raises concern regarding the combinatorial effects of both on HAND progression [17-20]. Chronic low-level inflammation, mediated likely by residual viral proteins, cART, and drugs of abuse, has been implicated as a significant factor underlying the progression of HAND. More importantly, neuroinflammation has been recognized as an essential component of HAND.

Various studies conducted on the association between HIV-1 infection and drug addiction clearly demonstrate the role of various drugs of abuse impairing both the immunological and non-immunological pathways of the host, ultimately augmenting HIV-1 infection and disease progression [2132]. Based on the epidemiologic studies, several animal models were employed to validate findings in the animal models of HAND. However, due to the long incubation period for the development of NeuroHIV, these investigations used several transitional outcome measures, such as CD4+ T cell decline, functional immune markers, HIV-1 load, and constitutional symptoms of HIV-1 infection, as well as neurological manifestations of HIV-1 infection. While the nonhuman primate is the gold standard model of HIV-1 infection, it is cost-prohibitive. Furthermore, HIV-1 does not infect rodents and felines – together, and these limitations have hampered adequate recapitulation of NeuroHIV.

Although several excellent non-human primate models of disease have provided valuable information on the disease process [3337], working with macaques poses many other challenges, such as the need for a large team of investigators and infrastructure and the limitation of numbers of animals available for the study design (cost-driven). As a result, there is a need for developing alternative models of HAND. Over the last 30 years, several unique strategies have been employed for developing rodent models of HAND. While none of these models accurately mimic the human syndrome, they serve to model some aspects of HAND. These rodent models include: 1) Severe combined immunodeficient model involving the direct stereotactic injection of HIV-1-infected human monocytes into the CNS of immunodeficient mice [3843]; 2) Direct injection of HIV proteins either into the CNS or via intravenous route [4449]; 3) Transgenic mice with constitutive or inducible expression of HIV proteins [5055]; 4) HIV Transgenic rat model expressing all of the HIV proteins except gag and pol [5659]; 5) EcoHIV mouse generated by inoculation of mice with a chimeric HIV that uses species-specific cellular receptors to enter mouse cells [60, 61]; and 6) humanized mice generated by transplanting human cells or tissues into genetically modified immunodeficient mice [62]. Further, the feline model of HIV is the only spontaneously occurring model of immunodeficiency [6366], and the infected felines elicit high acute viremia, depleted CD4+ and CD8+ cells, and the establishment of chronic infection, whereby the virus is controlled naturally [67]. The target cells of FIV and HIV/SIV are distinct, and with the structure of the reservoir being different, the use of the FIV model for HIV and drug abuse research is likely limited [68]. While outcomes from a substantial number of laboratory-based findings reveal that drug abuse could exacerbate HIV-1 disease progression, epidemiological studies have shown mixed results. This review provides an overview of various animal models used in this field and a careful evaluation of methodological strengths and limitations inherent in the existing animal models to understand better how these animal models complement each other.

2. HIV and drug abuse comorbidity

Drug abuse is a prominent comorbidity and a contributor of HIV-1 infection. It is common among HIV-1-infected individuals and about 50% of people living with HIV-1 report current or past drug abuse history. HIV-1 infection and drug use intersect epidemiologically, and their combination can result in complex biological effects on the brain and alterations in behavior [6972]. The extent to which drugs affect persons’ health with HIV-1 depends on many factors, including drug characteristics, use patterns, stage of HIV disease and its treatment, comorbid factors, and age [69, 73, 74]. HIV-infected patients who inject drugs have exhibited delayed HIV diagnosis, insufficient HIV care, poor antiretroviral adherence, treatment outcomes, and accelerated disease progression [6971, 7478]. Behavioral disorders, including depression, have also been identified as common critical comorbidities in patients with substance use disorders and/or HIV disease [7982]. The literature survey on clinical studies shows that the prevalence of depression is high among HIV-1 infected people with substance abuse [69, 8387]. HIV-mediated immune response and chronic neuroinflammation have been reported as significant factors underlying neurocognitive disorders, a severe complication of HIV-1 infection [3]. Previous reports have demonstrated that drug abuse contributes to increased viral load in the brain via increasing the infiltration of HIV-infected cells and accelerated glial infection, in turn, contributing to impaired immune responses and increased release of inflammatory mediators as well as neurotoxins [25, 8893]. Several studies have also reported infiltration of HIV-infected cells to the brain and the role of infected glia as contributors of neuronal dysfunction and modulation of brain circuits, leading to accelerated drug abuse and addiction [9398]. The development of models that accurately recapitulate HIV-1 infection and disease progression, and the comorbidity of drug abuse patterns in humans, could help understand the effects of drugs of abuse on HIV-1 infection and the development of effective therapeutics and vaccines. Also, a growing body of data links genetics to the risk of addiction [99, 100]. For example, the role of Caenorhabditis elegans as a model to study the molecular and genetic mechanisms of drug addiction has been elegantly reviewed [101]. The potential role of Drosophila melanogaster for the identification of target genes for drug addiction has also been reported [102]. Rodent models are also instrumental in establishing a link between genes and addiction [102]. This review discusses the several experimental models used to study the neurological effects of drugs of abuse on HIV-1 infection and their limitations.

3. HIV-1 Tg26 transgenic mouse model

HIV-1 Tg26 Transgenic mice harboring a non-infectious HIV-1 transgene pNL4-3: d1443 encoding all the HIV-1 genes except gag and pol were developed by Dr. Abner L. Notkins group in 1991 [103]. pNL4-3 (HIV-1 NL4-3 Infectious Molecular Clone) is a 7.4-kb recombinant proviral clone consisting of DNA from HIV-1 NY5, and LAV is used for in vitro manipulations of the HIV-1 proviral sequences [104]. The HIV-1 transgene pNL4-3: d1443 was generated by deleting the 3.1 kb sequence between the Sphl site at base position 1443 and the Bal1 site at position 4551 in pNL4-3 [105]. As shown in Fig. 1, the construct lacking the gag/pol region after deleting the 3.1-kb sequence was microinjected into fertilized mouse eggs derived from Friend leukemia virus B (FVB/N) mating [103]. The Tg26 mice are normal in appearance and weight and express high levels of HIV-1 transcripts including gp120, Tat, Nef, Rev, Vpu, Vpr, and Vif in the skin, skeletal muscle, and low levels in brain, lung, intestine, kidney, spleen, and thymus [106]. The homozygous mice show diffuse scaling of the skin and psoriasis-like lesions shortly after birth and develop progressive HIV-associated nephropathy (HIVAN)-like renal disease, comparable to that observed in human AIDS patients, and these mice do not survive beyond 3-6 weeks of age [106, 107]. The heterozygous Tg26 mice, on the other hand, have a much longer lifespan and develop the renal disease between 60 and 250 days [107, 108]. Thus, this model was widely used in studying disease pathogenesis, genetic susceptibility, and therapeutic intervention of HIVAN owing to its clinical relevance to ART-treated HIV-1-infected patients and resemblance to human HIVAN [107, 109].

Figure1.

Figure1.

Schematic of generating the pNL4-3:d1443 proviral transgene

Since most of the Tg26 mice generated on the FVB/N background had short lifespans and developed kidney disease, it posed a limitation in studying other HIV-associated disorders. Thus, HIV-1 Tg26 transgenic mice backcrossed onto other genetic backgrounds were studied, including CBA, DBA/2, CAST/Ei, C3H/He, BALB/c, DBF1, and C57BL/6. Among these mice strains, Tg26 mice backcrossed with C57BL/6 showed either normal renal histology or only mild mesangial expansion without overt podocyte damage [110, 111] and became a reliable model for the study of HAND as these mice have longer life expectancies. The viral mRNAs encoding Tat, gp120, vpu, and nef were detected in the brains of Tg26 mice, especially in the SVZ and SGZ neurogenic regions [112]. Moreover, the HIV protein Tat was detected in the homogenates from brain microvessels of Tg26 mice and was found to have higher expression levels in the older than younger Tg26 mice [113].

Interestingly, the expression of viral transcripts in the brain was relatively low but was constitutively persistent, thus implicating its clinical relevance to chronic HIV-1 infection [112]. Furthermore, an earlier study that examined archival pediatric brain tissues from series of HIV-1 infected infants and children with severe NeuroAIDS found HIV-1 RNA transcripts localized to the neurogenic zones, which was in agreement with the presence of HIV-1 viral transcripts in neurogenic zones of Tg26 mice [114]. Putatunda et al. demonstrated that Tg26 mouse neural stem cells (NSCs) had reduced neuronal but increased astrocytic differentiation. There were significantly higher amounts of quiescent NSCs, as well as significantly lower levels of active NSCs, proliferating neural progenitor cells, and neuroblasts in the subgranular zones of Tg26 mice. Also, newborn mature granule neurons in the dentate gyri of Tg26 mice had deficiencies in dendritic arborization, dendritic length, and dendritic spine density [112]. It was also found that Tg26(+/−) mice exhibited no significant differences in behavioral functioning, contextual fear conditioning, or cued fear conditioning responses as in wild-type mice. Male Tg26(+/−) mice, on the other hand, revealed significantly impaired short and long-term spatial memory, while female Tg26(+/−) mice had impaired spatial learning abilities and short-term spatial memory, which related to fewer quiescent neural stem cells and neuroblasts in the hippocampi compared with wildtype mice [115]. Thus, these studies indicated that the Tg26 mice model could also help understand the mechanisms underlying HAND pathology and study sex-related differences associated with the disease.

HIV Tg26 mice have also been used to study the interaction of HIV-1 infection and drug abuse [72, 116]. For example, studies using HIV-1 Tg26 mice demonstrated that morphine inhibited the growth of intestinal organoids derived from Tg26 mice by inhibiting the Notch signaling pathway [117]. Although several studies reported using the Tg26 mice [62, 63, 31], this model is not without limitation since the extent of translation of the viral transcripts is unknown.

4. HIV Nef mouse model

The accessory Nef protein is expressed by the HIV genome and is one of the six accessory proteins involved in T-cell activation and the establishment of persisting infection. The persistence of HIV infection is primarily based on the ability of Nef to downmodulate the surface levels of essential molecules at the immune synapse, i.e., major histocompatibility complex-I (MHC I) and (MHC II) on antigen-presenting cells, CD4 and CD28 on present on T helper cells [118]. Several models of HIV Nef have been established to date. Briefly, transgenic mice expressing Nef in the T-cells and other regulatory elements - CD38, CD2, and TCR β have been developed [119, 120]. While these models exhibit loss of T-cells and alterations in their activation accompanied with pathological lesions in the heart and kidney, none of these models exhibit multiorgan syndrome or a disease phenotype similar to AIDS. Expression of Nef in the double Tg mice (TRE/HIV nef × CD4C/rtTA) was also induced by doxycycline administration and resulted in the aging phenotype [121, 122]. Another type of Nef transgenic mice model with the expression of Nef in the microglia was developed. These mice demonstrated hyperactive behavior in male mice, with the production of CCL-2 in the striatum, and exhibited a dysfunctional dopaminergic system, thereby underscoring their utility as a model for HAND pathogenesis [123]. There are, however, several limitations in this model - since this mouse model only expresses Nef, it precludes the contribution of various other viral proteins [124].

Additionally, humanized mouse models have also been used for the study of HIV Nef. In this model, wild type or the Nef-defective HIV-1JR-CSF/HIV-1NL4-3 were injected into the thymic organoids grown from human fetal thymus and liver implanted under the kidney capsule of SCID mice. Both the Nef defective viruses showed attenuated growth properties relative to wild type. Nef defective HIV-1NL4-3 lost the ability to prevent thymocyte loss, while active viral infection in wild-type virus caused loss of thymocytes within six weeks of infection [125]. These results demonstrated that HIV-1 Nef is required for in vivo viral replication in the thymus. Another humanized mouse model for HIV-1 Nef (LAI, CXCR4 tropic) showed significant depletion of CD4+ T cells. These results collectively demonstrate that viral Nef contributes to T-cell loss during HIV-1 infection. Infection of this mice model with HIV-1 JRCSF (CCR5 tropic) strain resulted in the loss of CD4+T cells in various organs but not in the peripheral blood [124]. Results from these studies implicated the role of Nef in HIV viral replication and CD4+ T cells. Studies also showed the role of Nef in HAND pathogenesis-transgenic mice in neuropathology, e.g., vacuolar myelopathy and changes in the oligodendrocyte phenotype in the brain [126, 127]. In another study, it was shown that rats implanted unilaterally with astrocytes expressing Nef, exhibited impairment of novel location and novel object recognition compared with controls that were implanted with astrocytes expressing the green fluorescent protein. Additionally, this impairment correlated with an increased expression of the chemokine ligand 2 (CCL2) in the implanted Nef transfected astrocytes followed by the infiltration of peripheral macrophages into the hippocampus at the site of injection. These rats also showed bilateral loss of CA3 neurons. This study thus showed that continued expression of Nef by astrocytes in the absence of viral replication has the potential to contribute to HAND [128]. Interestingly, cell culture studies have demonstrated that Nef overexpressing astrocytes (via adenovirus vectors) can release Nef in the extracellular vesicles, which, in turn, can be up taken by the neurons, leading to neuronal dysfunction in HAND [129]. Although few studies indicate the role of Nef in HAND, further detailed studies are required to dissect the role of Nef in other in vivo models of HAND.

5. Doxycycline-inducible HIV-1 Tat transgenic (iTat) mice model

The doxycycline-inducible HIV-1 Tat transgenic (iTat) mice model was generated from crossbreeding two separate transgenic lines -GFAP-rtTA mice (G-tg) and TRE-Tat86 mice (T-Tg). The G-tg transgenic line was obtained using a Teton-GFAP DNA fragment released by Xhol and Pvull digestion of the pTeton-GFAP plasmid, while the T-tg transgenic line was obtained using a DNA fragment (1189 bp) released by the same restriction digestion (Xho I and Pvu II) of the pTRE-Tat86 plasmid. These two DNA fragments were separately microinjected into fertilized eggs of F1 females (obtained from a mating between C3HeB and FeJ mice), then crossed with C57BL/6 mice to generate stable transgenic lines [50]. GFAP-rtTA mice and TRE-Tat86 transgenic mice were crossbred to obtain iTat mice carrying both GFAP-rtTA and TRE-Tat86 transgenes. The Tat expression levels were regulated by both the astrocyte-specific glial fibrillary acidic protein (GFAP) promoter and a doxycycline (Dox)-inducible promoter in the iTat mice model [130]. Expression of Tat mRNA levels in various organs or tissues of iTat mice was assessed to ensure the designed constructs specifically expressed Tat within the brain [50, 131] (Figure 2). It was shown that systematic exposure to Dox-induced Tat expression only in the brain and not in other organs or tissues such as the eye, heart, kidney, liver, lung, spleen, or thymus [50]. The protein levels of Tat in iTat mice’s brains were found to range from 1-5 ng/ml, which agrees with the levels of Tat reported in the brains of HIV-infected individuals [51, 52]. The inducible Tat mice are thus a valuable tool for understating Tat neurotoxicity and its role in HIV-1 neuropathogenesis. This model significantly improves over direct injection of recombinant Tat protein into the brain since the injection causes secondary damage to the brain [52].

Figure 2.

Figure 2.

Schematic of generating the iTat mice

The utility of this model is also evident by the fact that Tat expression in the brain of iTat mice also leads to several developmental and behavioral abnormalities, including failure to thrive, hunched posture, tremor, ataxia, slow cognitive and motor movement, seizures, and premature death [50], similar to some of the cognitive changes observed in humans with HAND. Extensive neuropathological changes have also been reported in the iTat mice, including astrocytosis, neuronal death, degeneration of neuronal dendrites, morphological changes of astrocytes and microglia, and infiltration in the CNS by monocytes and activated T lymphocytes [50, 51, 132, 133]. Furthermore, increased expression of cytokines and chemokines has also been reported in the brains of Dox exposed iTat mice, including monocyte chemoattractant protein-1 and monocyte chemoattractant protein-2 (MCP-1/MCP-2), macrophage inflammatory protein-1α and protein-1β (MlP-1α/-1β), RANTES, Interferon gamma-induced protein 10 (IP-10). Thus, this model encompasses several features that recapitulate HIV-induced neurocognitive disorder findings both before and in the era of combination antiretroviral therapy (cART) [52].

Over the past two decades, there has been an escalation in the number of studies using the iTat mice to examine the direct effects of viral Tat protein on astrocytes and the molecular mechanisms of Tat-induced GFAP expression/astrocytosis [52]. Tat neurotoxicity [51, 134]. Tat-mediated impairment of neurogenesis [135]. Tat-induced loss of neuronal integrity [135], and exosome-associated Tat release and uptake [136]. For example, it has been demonstrated that Tat affected neural progenitor cells (NPCs) and neurogenesis through Notch signaling in the iTat mice, which underscores the potential of developing Notch signaling inhibitors for NeuroHIV therapeutics [135]. Interestingly, many reports have also utilized the iTat model to address Tau processing [137], comorbidity of substance abuse [138144], sex differences [145], and aging [146]. For example, the effects of long-term Tat expression in the brain on behavior, pathology, and epigenetic landscape were studied by feeding the iTat animals a Dox-containing diet for 12 months, which was suggested to simulate people infected with HIV-1 for ~5 decades. These findings demonstrated that long-term Tat expression in the brain led to poor memory and impaired motor functioning, brain region- and sex-dependent dysregulation of neuropathological marker expression, DNMT3B expression genomic DNA methylation, thereby underscoring the critical role of HIV Tat in accelerated aging [146].

While this model has been used extensively in the field, providing relevant information on neurobehavioral and neuropathogenesis of HAND, it also has several limitations. First and foremost, the iTat model is not an infectious model and, while it can be argued that it mimics the persistence of Tat in the brain (as is evident in infected individuals on cART), the limitation is that it is Tat-centric, ignoring the impact of other residual proteins such as gp120, gp140, Nef, Vpr, and Rev [147149] in the CNS. Secondly, how does this model correlate to the HAND neuropathology in cART-treated individuals? Furthermore, while it has been reported that HIV-1 infects up to 19% of perivascular astrocytes in individuals with HAND [150], these cells are not the primary producers of viral Tat protein in the brain of HIV-infected patients.

6. EcoHIV mouse model

HIV-1 does not infect rodents owing to viral entry restriction. To overcome the viral entry restriction, Potash et al. designed a chimeric virus that contains most of the HIV genes, and that can infect mouse immune cells by replacing the HIV gp120 gene with the ecotropic mouse leukemia virus gp80 gene, thus enabling the entry of the virus into mouse cells expressing the cationic amino acid transporter [151]. The resulting chimeric virus constructs, EcoHIV, productively infects murine lymphocytes but not human lymphocytes in culture. Adult immunocompetent mice are readily susceptible to infection by a single inoculation of EcoHIV, as evidenced by the virus detection in splenic lymphocytes, peritoneal macrophages, and the brain [151].

To date, two chimeric viruses have been utilized in studies investigating gene expression in both the periphery and CNS. Specifically, the EcoHIV chimeric virus is built on a backbone of clade B NL4-3 and EcoNDK, built on a backbone of clade D HIV-1. Both EcoHIV and EcoNDK have been shown to establish a systemic infection in mice after one inoculation [151]. Mice infected with these viruses reproduce the characteristics of HIV-1 infection in humans, such as viral infection of immune cells, i.e., lymphocytes and macrophages, induction of antiviral immune responses, neuroinvasion, and expression of inflammatory and antiviral factors in the brain. Infection of mice with EcoNDK also increases proinflammatory factors such as interleukin-1 β and STAT-1 in the brain [151]. A recent study has shown that EcoHIV infection of mice can also establish latent viral reservoirs in T cells and concurrently show active viral replication in macrophages, leading to the induction of neurocognitive impairment [61].

The EcoHIV model has been valuable for many applications, including antiviral drug screening [60], development of vaccines [152, 153], studying the effects of opioids on early HIV pathogenesis in the gut [154], investigating the therapeutic potential of biologicals such as insulin for HAND [155], and more recently, for HIV viral latency, pathogenesis and neurocognitive impairment [61, 155157]. Valuable insights have been gleaned from the EcoHIV rodent model. Firstly, this model serves as an infectious model of HIV-1, resembling HIV infection in the era of cART [158, 159]. This model has provided utility in the preclinical evaluation of anti-retroviral drugs and has been found to provide rapid, statistically robust, and inexpensive evaluation of antiretroviral drugs in vivo [60]. Similarly, it has also proven useful to test novel vaccine candidates [152, 153]. Biologicals such as insulin were also tested in vivo via intranasal delivery for their protective effects against HAND using the EcoHIV-infected mice [155]. The EcoHIV model has also been reported to be a valuable model for studying the effects of opioids in HIV-induced pathology during the early stages of infection in the gut [154].

The EcoHIV model has also played an essential role in understanding how methamphetamine potentiates HIV infectivity in NPCs [160]. In this study, the authors infected mouse NPCs with EcoHIV in the presence or absence of methamphetamine (50 or 100 μm). They found that methamphetamine increased HIV infectivity of NPCs, involving the NFκB/SP1-dependent activation of HIV LTR, thereby suggesting that methamphetamine exacerbates neurocognitive dysfunction [160]. The EcoHIV model, similar to other models, is not without limitations. The chimeric virus infects multiple cell types, which may not adequately reflect the human scenario. Additionally, viral levels and the spread of infection in this model do not precisely mimic the humans infected with HIV-1.

7. HIV envelope glycoprotein 120 (gp120) Transgenic (Tg) mouse model:

HIV envelope glycoprotein 120 (gp120) is one of the most prominent viral antigens produced in HIV-infected cells and is shed into the extracellular space as a soluble protein, which, in turn, has the potential to interact with uninfected cells [161]. The toxicity associated with gp120 was examined as a model to explore the neuropathological and neuropsychiatric manifestations associated with AIDS [147, 162166]. The HIV gp120-transgenic (Tg) mouse model was generated by inserting the HIV env gene that encodes soluble gp120 under the control of the GFAP promoter, such that it is expressed in astrocytes [167]. The transgene is highly expressed in the brains, specifically in the neocortex, olfactory bulb, hippocampus, tectum, selected white matter tracts, and other areas associated with astrocytes. The neuro pathophysiology, including decreased synaptic and dendritic density, frank neuronal loss, increased numbers of activated microglia, and pronounced astrocytosis and behavioral phenotypes found in human HIV patients, has also been found in this Tg mouse model [166, 167]. An interesting study using this model demonstrated that gp120 expression was a useful indicator for assessing the pathophysiological effects on neuronal networks and showed divergent changes in short- and long-term potentiation in the CA1 hippocampus [168]. Although this Tg model only expresses viral protein gp120, this model has utility for the study of HAND pathology and recapitulates human HIV patient brains on cART therapy and low levels of replicating virus [166]. Many studies have shown that these mice’s brains mimic the pathological findings observed in HAND patients, including differentially regulated genes and cellular signaling pathways, activation of innate immunity, impaired neurogenesis and cognition, and learning and motor deficits [53, 158, 166]. These animals also exhibited impaired proliferation and differentiation of NPCs [169, 170]. Interestingly, gp120 Tg mice also demonstrate age-dependent impairment in open field activity and spatial reference memory, reflecting cognitive and neuromotor deficits seen in HIV-1-infected individuals [165]. Another study using the gp120 Tg mouse model demonstrated that pharmacological abrogation of CCR5 signaling exerted a novel protective effect on lipocalin-2 associated neuronal injury and behavioral impairment [53]. In another study, it was shown that gp120 Tg mice exhibited DNML1-associated downregulation of mitochondrial fission (Dynamin-1-like protein-associated mitochondrial fission) in neurons to that observed in the brains of HAND patients [171]. A recent study reported that gp120-tg mice showed NLRP3- dependent pyroptosis and IL-1β production in microglia [147]. Importantly, inhibition of NLRP3 inflammasome activation in microglia via chronic administration of MCC950, a novel selective NLRP3 inhibitor, to gp120 Tg mice attenuated neuroinflammation and neuronal death, while restoring neurocognitive functioning [147].

Drugs of abuse and HIV-1 are implicated in the progression of neurodegenerative effects such as astrogliosis [172], compromised blood-brain barrier (BBB) integrity, and excitotoxicity in the brain, as well as the progression of neurocognitive decline [173176]. An earlier study using the gp120 Tg mouse model showed an increased preference for methamphetamine and increased sensitivity to methamphetamine-induced conditioned reward [173]. While this model has provided value in the field, the limitation of this model is that constitutive expression of the Tg throughout the lifespan of the animals can lead to early developmental changes before the onset of the desired pathological phenotype. Like the iTat mice, another important caveat of this model is that these animals only express one viral protein, which excludes the pathological effects of other viral proteins, thus failing to recapitulate the human clinical scenario of the disease [163, 166]. Crossbreeding of the Tg models expressing distinct viral proteins could help to elucidate the combined effects of various viral proteins in mediating neuropathogenesis.

8. Humanized mouse model

While the Tg mice models expressing select HIV-1 proteins (such as HIV-1 envelope and Tat) mimic aspects of HIV-1-related pathologies [108, 177180], they only mirror specific pathophysiological effects of select HIV-1 proteins on the CNS [181] but do not recapitulate infectious virus. In light of this, the development of humanized mice models that can mimic the human immune system is of prime value. As stated above, all strains of mice are naturally refractory to HIV-1 infection. However, the humanized mice, which are immunodeficient reconstituted with human donor cells and tissues, offer a unique small animal model for studying HIV neuropathogenesis, prevention, and therapeutic development in vivo.

8.1. Severe combined immunodeficient (SCID)-HIVE model

The SCID-HIVE model was developed at Johns Hopkins University in the early 1990s [40] to investigate HIV-infected cells’ role in contributing to HIV-associated encephalitis (HIVE). In the original study, the SCID mice, which lack murine B and T cells, were intracerebrally injected with isolated human peripheral blood mononuclear cells (PBMC). Next, the PBMC-injected SCID mice and control mice were injected with HIV-1. As expected, the SCID mice injected with both PBMC and HIV-1 developed striking gliosis and pathology characteristic of HIVE. Subsequent studies of injecting HIV-infected human myeloid cells into SCID mice’s brains [39, 182184] also resulted in pathologic changes similar to HIVE, such as encephalitis, astrogliosis, and an increased influx of mononuclear phagocytes, within a week. This model was also tested to assess cART treatmen’s effects in mitigating HIVE-associated pathologies [185187]. The results demonstrated that cART could reverse HIV-mediated glial activation, mononuclear cell infiltration, and neuronal dysfunction, suggesting that replication of HIV-1 directly contributes to brain pathology in the SCID-HIVE model.

Recently, the SCID mouse model has been used to evaluate strategies for reversing HIV-induced neuroinflammation. Baricitinib, a JAK 1/2 inhibitor, which has been approved for rheumatoid arthritis, demonstrated promise in the inhibition of pro-inflammatory cytokines such as IL-6, D-dimer, CRP, TNF-α, IFN-α/β [188]. In the SCID mice, baricitinib treatment was shown to cross the BBB and reverse HIV-1-mediated behavioral abnormalities. Furthermore, baricitinib reduced glial activation, as evidenced by decreased expression of MHCII/CD45 (microglial activation) and GFAP (astrogliosis). Additionally, baricitinib treatment also significantly decreased p24+ human macrophages in mouse brains [188]. CEP-1347, an inhibitor of mixed lineage kinases, was also shown to reduce microglial inflammation as measured by decreases in Iba-1 expression [189] in the SCID-HIVE mouse model. Also, SCID-HIVE mice treated with B18R, a novel IFN-α inhibitor that crosses the BBB, demonstrated decreased numbers of mouse mononuclear phagocytes that were accompanied by significant restoration of neuronal arborization, as measured by decreases in the expression of mouse microtubule-associated protein-2 (MAP-2), compared to untreated SCID-HIVE mice [190]. SCID-HIVE mice were also utilized as a model to address the comorbidity of drugs [191194] and autoimmune diseases [195, 196].

One of the significant limitations of the SCID-HIVE model is that the direct injection of human cells into the brain results in a traumatic injury that manifests as a cellular activation in the brain. The pathology is more pronounced in mice injected with HIV-infected cells than in mice administered uninfected cells. Additionally, injection of the mature immune cells could also lead to graft-versus-host-disease, which results in a shorter lifespan of SCID-HIVE mice (four weeks or less). Thus, this limitation eliminates the use of the SCID-HIVE model for long-term analyses of HIV persistence [197].

8.2. CD34-transplanted NSG mouse model or hu-NSG

The NOD.Cg-PrkdcscidIl2rgtm1Wjl/SzJ (NSG, NOD SCID gamma) mouse was generated by introducing an Il2rg knockout allele into NOD SCID mice, producing a mouse strain that lacks functional B cell, T cells, and natural killer (NK) cells. In contrast to the NOD SCID mouse, NSG mice lack functional NK cells that prevent them from the development of thymic lymphoma (the primary cause of death of NOD SCID mouse), resulting in an increased life span (NSG = 16 months, NOD SCID = 8 to 10 months) [198], thereby making the NSG mouse a superior model for long-term HIV-1 study.

The hu-NSG model is derived from NGS mice engrafted with human CD34+ hematopoietic stem cells, isolated from the cord blood, bone marrow, or fetal liver either by intravenous or intrahepatic injection [62, 199, 200]. HIV-1-infected NSG mice were found to exhibit systemic HIV-1-infection and CNS viral burden, leading to the development of HIV-associated pathologies that are accompanied by depletion of CD4+ T-cells, viral replication, CNS infiltration of HIV-1 infected monocytes and macrophages, resident microglial and astrocytic activation, neuroinflammation and cognitive abnormalities [62, 181, 201]. This hu-NSG model has been successfully utilized to evaluate multiple HIV-1 therapeutic strategies in vivo. For example, Wu et al. investigated the prophylactic protection effect of an engineered broadly neutralizing antibody (bs-bnAb, BiIA-SG) in the hu-NSG model. Treatment with this antibody was found to eliminate infected cells in hu-NSG humanized mice. Additionally, the antibody treatment combined with cART exposure could delay the viral rebound in this hu-NSG humanized mouse model [202]. Kumar and colleagues successfully used a CD7-specific single-chain antibody conjugated to the oligo-9-arginine peptide (scFvCD7-9R) to achieve T cell-specific anti-CCR5 and antiviral silencing RNAs (siRNA) delivery in hu-NSG mice. This was found to control viral replication and prevent disease-associated CD4 T cell loss [203]. Recently, Sonic Hedgehog (Shh) mimetic, a Smoothened Agonist (SAG), was found to reduce CNS viral burden immediately after HIV-1 transmission, while also conferring extended neuroprotection via increasing BBB integrity, assessed by elevated levels of tight-junction protein, Claudin 5, and decreased levels of S100B in the periphery of HIV-1 infected huNSG mice [204].

8.3. Myeloid only mouse (MoM) model

Although several studies implicate that monocytes and macrophages are the critical cells infected by HIV-1, both in vitro and in vivo [205207], the question of whether myeloid lineage cells serve as HIV-1 reservoirs in the context of cART treatment remains an area of high investigation. Recently, Honeycutt et al. generated a myeloid-only mouse (MoM) by injecting CD34+ human hematopoietic stem cells, reconstituted with human myeloid and B cells in the absence of human T cells, into pre-conditioned NOD-SCID mice [208]. A major benefit of MoM is that it rules out the possibility that macrophages could become HIV-1 positive via the phagocytosis of infected CD4+ T cells [209211]. Through the use of this Mom mouse model, Honeycutt et al. demonstrated that macrophages could be infected by HIV-1, independent of T cells, to support efficient HIV-1 replication in vivo [212]. Additionally, the withdrawal of suppressive cART from MoM resulted in viral rebound after seven weeks [208], thus supporting the hypothesis that long-lived tissue macrophages could serve as HIV-1 reservoirs [208, 213].

8.4. Humanized microglial mouse

While most studies of HIV-1 latency focus on the peripheral population of T cells, the brain also contains a distinct reservoir of HIV-1 infected cells in the microglia. Human perivascular macrophages, microglia, and astrocyte are critical for simulating HIV-1 induced neuropathogenesis and the study of brain virus reservoirs. Immune deficient humanized mice are susceptible to HIV-1 infection and have been shown to serve as valuable tools for HIV-1 infection of the CNS. Low numbers of human cells and even lack of human microglia have been detected in humanized mice’s brains by histology and qPCR [212, 214, 215]. It is essential to generate the human microglial mice for targeting CNS HIV infection and for the study of HIV-1-mediated neuropathogenesis.

Recently, Mathews and colleagues developed a new humanized microglial mice model that comprises an immunodeficient strain supplemented with human interleukin-34 (IL-34) transgene to support microglial development [216]. Herein, the authors first generated the hIL-34 expressing transgenic NOG mice (NOG-hIL-34) by microinjecting fertilized eggs obtained by mating NOG (NOD.Cg-Prkdcscidil2gtmlSug/Jic) and NOD (NOD/ShiJcl) mice with a linearized DNA vector (pCMV6-XL4) containing human IL-34 cDNA under the control of a CMV promoter. Next, this NOG-hIL-34 mouse has engrafted intrahepatically with the umbilical cord blood-derived CD34+ hematopoietic stem progenitor cells (HSPCs) to generate the humanized microglial mice (CD34-NOG-hIL-34). HSPC-derived peripheral macrophages generated microglial-like cells in the CNS and expressed human microglial cell markers such CD14, CD68, CD163, CD11b, ITGB2, CX3CR1, CSFR1, TREM2, and P2RY12 in the CD34-NOG-hIL-34 humanized mouse model. Intriguingly, these mice expressed features resembling the pathobiology of HIV-1 infection in the CNS, including activation of astrocytes and microglia, a productive viral infection of microglia, synaptic alterations, and inflammatory responses, thus underscoring the role of this model in neural-glial cross talk and in studies aimed at targeting/eliminating the viral reservoirs in the brain. In another study, when iPSC-derived hematopoietic progenitor cells (HPCs)-primitive macrophage progenitors were transplanted into the brain ventricles and overlying cortices of the immune-deficient, humanized CSF1 mouse pups, it was shown that the expression of human CSF1 was required for long-term engraftment of human microglia in the murine brain [217]. Furthermore, it was also shown that the microglia that developed from the transplanted HPCs had transcriptional profiles similar to those of microglia from the human brains. These findings thus suggested that human iPSC-derived microglial chimeric models can mimic human microglia development and its impact on neuronal development, synaptic plasticity, and behavioral performance of the animals [217, 218]. There have also been reports wherein embryonic stem cells (ESCs)-derived human microglia were used to generate microglia humanized mouse model. These authors differentiated H9 ESCs into microglia by using IL-34, colony-stimulating factor 1 (CSF1), transforming growth factor-β (TGF-β) and CX3CL1, followed by transplanting cells into the brains of Rag2−/− Il2ry−/− hCSF1KI mice (hCSF1KI) at postnatal day 4. To create a permissive environment for human microglia integration, the neonates were pretreated with CSF1 receptor (CSF1R) inhibitor BLZ945 [219] that eliminates an average of 53% ± 7% of host microglia. Eight weeks later, H9-microglia represented 9 ± 5% of the total microglial population with a mosaic distribution across multiple areas of the brain. The H9-microglia demonstrated a complex ramified morphology that could be detected by the contribution of microglia in neurological diseases.

9. Rat models: HIV-1 Transgenic rats

The HIV-1 transgenic (Tg) rat model (HIV-1 Tg) developed by Reid et at. expresses a noninfectious HIV-1 provirus Tg that has both the gag and pol genes deleted from the infectious clone of an integrated proviral plasmid (pNLS-3) [56]. The approach involved deleting an overlapping fragment containing 2 of the 9 viral genes, i.e., the gag gene at the 3′ region and the pol gene at the 5′ region, resulting in a non-infectious provirus pEVd1443 [56]. This non-replicative provirus is regulated by the viral long terminal repeat, which activates the transcription of the env gene plus the regulatory (tat, rev) and the accessory (vif, vpr, vpu, and nef) genes.

Here, the non-infectious provirus was microinjected into the fertilized eggs derived from a Fisher/NHsd (F344) rat and Sprague Dawley rats to produce a female Tg founder [56]. The Tg was incorporated into one copy of the two alleles of the HIV-1 Tg rat. When these rats are mated with a wild type inbred F344 rat, the offspring are either HIV-1 Tg or wild type. The HIV-1 Tg rats have an easily distinguishable phenotype that includes opaque cataracts, wasting, skin lesions, neurological signs, respiratory problems, and nephropathy [56, 220]. The wild-type littermates without cataracts or standard F344 rats are usually used as control animals.

The HIV-1 Tg rat model is suitable for studying the effects of HIV-1 viral proteins in both the periphery and the CNS. Consequently, the HIV-1 Tg rat model has been utilized in studies of the neurological effects of HIV-1 [221, 222], investigations focused on the interaction of HIV proteins with drugs of abuse [223], and behavioral studies [57]. Several impressive results have been obtained using this HIV-1 Tg rat model. The HIV-1 Tg rat mimics HIV-infected patients on cART, owing to low levels of viral proteins. This model has been used to study the long-term effects of these persistent viral proteins on disease pathogenesis. For example, one study using this model demonstrated astrocytic dysfunction/damage and dopaminergic neuronal loss/dysfunction compared with wild-type controls. Interestingly, both these parameters worsened in the striatum with the animals’ increasing age [222]. Another study on immune activation, viral gene expression, and neurotoxicity in HIV-1 Tg rats demonstrated that activated mononuclear phagocytes and astrocytes expressing the HIV gene products in the brain were associated with increased neurodegeneration [221].

The HIV-1 Tg rat model also provides an opportunity to study mechanisms underlying HIV-related neurological and cognitive dysfunction, a limited research area due to the lack of an adequate animal model [57]. In a study, Vigorito et al. evaluated the HIV-1 Tg rat model, using the Morris water maze for testing learning and memory deficits and found that these rats showed a deficit in learning how to swim to the location of the hidden platform, thus suggesting the validity of HIV-1 Tg rats as a model for behavioral processes underlying HIV-related neurological dysfunction [57]. In the context of drug abuse, this model was used to demonstrate that methamphetamine self-administration increased compulsivity, inflammation, and neural injury in these animals compared to the wild-type controls [223, 224]. The HIV-1 Tg rat model was also used in behavioral studies with drug abuse [225227]. This model also is fraught with limitations. As described earlier, this is a non-infectious model and, thus, is not suitable for studies investigating viral progression or replication or for studying the impact of novel antiviral drugs on viral replication [228].

10. Rodents with direct injection of HIV proteins

Several research groups have used the direct injection of HIV proteins into the brain to study the effects of individual proteins such as Tat or gp120 in modulating cellular processes involved in neurotoxicity and neurodegeneration. Among some of the earliest studies, Hayman et al. injected synthetic peptide analogs derived from the basic region of HIV-1 Tat protein into the rat brain and found Tat to be a potent neurotoxin [45]. This was confirmed by another study that also demonstrated that blocking TNF-alpha with pentoxifylline following Tat injection led to decreased IL-1β and iNOS expression that was accompanied by a reduction in the volume of pathological lesions, indicating that the Tat-induced lesions could be likely mediated by TNF-α production [229]. To study the neurotoxicity of HIV-1 proteins gp120 and Tat in the rat striatum, Bansal et al. stereotactically injected recombinant proteins (gp120, Tat, or saline as a negative control) in the striatum of adult male rats. They also evaluated the area of tissue loss and the number of GFAP immunoreactive cells 7 days post injections [230]. These authors found that doses of gp120 (250 ng/μL or higher) and Tat (5 microg/μL or higher) produced a significant area of tissue loss and significantly increased numbers of GFAP reactive cells thus suggesting the involvement of gp120 and/or Tat in striatal toxicity. In another study, the injection of HIV-1 Tat protein into the rat striatum revealed that Tat caused an increase in protein carbonyl formation. This type of oxidative modification of proteins occurred early after Tat injection preceded Tat-mediated astrogliosis [44]. These results were part of the early evidence suggesting that increased protein oxidation could be an essential mechanism underlying Tat neurotoxicity [44]. It was also shown that intraventricular injection of HIV-1 Tat protein in rats caused a cascade of events leading to the influx of inflammatory cells, glial cell activation, and neurotoxicity [46].

The interaction of Tat with methamphetamine was studied following injection of Tat into the striatum and methamphetamine by i.p. injection [231]. These studies showed that Tat injection, with concurrent administration of methamphetamine, profoundly increased chemokine and cytokine levels (MCP-1 and interleukin-1 alpha) in the striatum resulting in enhanced dopaminergic system damage [231]. The same group also utilized the Tat injection model to demonstrate that methamphetamine and Tat synergize to destroy dopaminergic terminals in the rat striatum [232].

Direct injection of HIV proteins into the CNS, as described above, demonstrates the utility of this model in elucidating the mechanisms involved in viral protein neurotoxicity and neuroinflammation. Results from studies using this model suggest specific pathways that could be blocked to attenuate viral protein-induced neurotoxicity and neuroinflammation. However, the limitation of this model is the inherent disadvantage of short-lived neuroinflammation in the CNS due to the rapid clearance of the injected proteins. Another caveat of this model is that stereotaxic injection of the viral proteins can result in a traumatic brain injury that can confound interpretation of results because it is challenging to differentiate injection-induced injury from infection-induced injury [158].

11. Simian Immunodeficiency Virus (SIV)

11.1. Natural SIV hosts

Around 40 different African primates species are natural hosts for SIV and can be infected with their own specific strains of SIV. However, these naturally occurring SIV infections do not result in disease due to thousands of years of virus-host co-evolution and are thus not useful as pathogenic infection models [233, 234]. Among the different natural hosts, Sooty mangabey (Cercocebus atys) carrying SIVsmm virus and African green monkey (Chlorocebus aethiops) carrying SIVagm infection have been studied due to their availability at primate centers in the United States and Europe [234]. The Sooty mangabey model is of much interest as the transmission of SIVsmm to macaques resulted in the development of SIVmac, while transmission of this virus to humans gave rise to HIV-2 [234]. The non-human primate models of SIV have many advantages, including vaccine research, and as SIV is genetically and morphologically close to HIV, this model also useful to study the effects of drug abuse [235239].

11.2. Non-natural SIV hosts

In contrast to the fact that the natural hosts of SIV do not exhibit chronic infection, Asian macaques are more vulnerable to infection with certain SIV strains and have thus been the most widely accepted models of HIV-1 infection. These models have been used extensively in studies to characterize disease pathogenesis and the development of vaccine development. The most commonly used non-human primate models of AIDS comprise the Indian rhesus macaques (Macaca mulatta), pig-tailed macaques (Macaca nemestrina), and the cynomolgus macaques (Macaca fascicularis) [240]. These macaque models have a unique evolutionary history that significantly impacts the outcome of SIV infection [240]. Indian origin Rhesus macaques are the best characterized and likely most utilized non-human primate model for the study of AIDS. The most commonly used virus strains for these macaques are SIVmac251 and SIVmac239, which cause productive infection with high viral loads and progressive loss of CD4+ T cells, particularly in the GALT [241]. Both of the viruses are neurotropic and have been well studied in models of NeuroAIDS [242244], as discussed later. Tripartite-motif-containing protein 5α (TRIM5α) was previously known to inhibit the post-entry block to HIV-1 infection of rhesus macaque cells [245]. Rhesus macaques encode highly polymorphic TRIM5 proteins [246]. Reports have shown that viral loads in SIVsmE543-3-infected macaques are 100- to 1,000-fold lower in animals expressing specific TRIM5α variants (with 339-TFP-341 in the SPRY (SPla and the Ryanodine Receptor) domain) than in animals expressing variants (with Q339 in the SPRY domain) [247]. The susceptibility of SIVsmE543-3 to certain rhesus macaque TRIM5α proteins likely reflects the incomplete adaptation of this virus to this host, mainly because SIVsmE543-3 was cloned after sequential passage in only two rhesus macaques [247]. These observations are essential, as SIVsmE543-3 and the closely related SIVsmE660 strain are more often used as a heterologous-challenge virus for rhesus macaques immunized with SIVmac239-derived antigens.

In addition to the Indian, Chinese, and Burmese rhesus macaques have also been used to study AIDS, albeit less frequently than the Indian rhesus macaques due to limited availability. SIVmac239, SIVmac251, and SHIV89.6P have been less pathogenic in Chinese and Burmese macaques than their effects in Indian rhesus macaques [248251]. The difference in pathogenicity is reflected in viral loads, higher CD4+ T cell counts, and prolonged survivability of Chinese macaques. The underlying reasoning for these differences is unclear but can likely be attributed to immunogenetic differences [252, 253]. Some studies have shown the relation between inflammation and viral infectivity in these Rhesus macaques. In a study, Sandler et al. [254] utilized SIV-infected Indian rhesus macaques to investigate the role of inflammation in HIV infection, particularly the impact of type-I interferons (IFN-I). These authors found that the timing of IFN-induced innate responses in acute SIV infection significantly affected the overall disease course [254]. Another group of investigators showed that increased proliferation and infection of CD4(+) T(SCM) could contribute to SIV infection’s pathogenesis in Indian rhesus macaques. The dynamics of germinal center (GC) formation in lymphoid tissues following acute SIV infection was a significant predictor of disease in rhesus macaques. Hong et al. showed that local production of IL-21 in lymphoid tissue GCs was an indicator of better control of viral replication and slower disease progression [255]. Thus, limiting dysregulated lymphoid function may be a critical target for new therapeutics. In one study, it was observed that there were polymorphisms of the TRIM5α B30.2/SPRY domain in SIVsmm, which regulated the viremia in rhesus macaques. Viremia in macaques homozygous for the non-restrictive TRIM5α allele TRIM5Q was significantly higher than in macaques expressing two restrictive TRIM5alpha alleles TRIM5TFP/TFP or TRIM5Cyp/TFP. This study further showed that mutation of TRIM5α in SIVsmm (substitution of the amino acid (P37S and R98S) in the capsid region) resulted in the cross-species transmission of SIV in primates [256]. In fact, it has been observed that SIVsm804E is capable of establishing infection in the CNS at an early stage of infection and causes neuropathology in infected rhesus macaques at a high frequency (83%) using a single inoculum when animals with restrictive MHC-I or TRIM5α genotypes are excluded, which depicts that macaques with restrictive TRIM5α, are resistant to developing NeuroAIDS [257].

In addition to the Indian rhesus macaques, the pig-tailed macaques are the most used nonhuman primates for AIDS research. Pig-tailed macaques infected with SIVmac239 develop AIDS within 42 weeks of infection, compared to 70 weeks for rhesus macaques [258, 259]. In these macaques’ strains, the MHC genes are not as well characterized as in the rhesus macaques. However, several SIV epitopes that are recognized by CD8+ T cells, and the MHC class I molecules that present these epitopes, have been defined, such as Mane-A1*084, which inhibits an immunodominant epitope of the SIV Gag protein and is associated with decreased viral load following SIV infection [260, 261]. A unique feature of pig-tailed macaques is that they do not express TRIM5α; they encode the TRIMCyp isoform, which does not restrict HIV-1 [262266]. The absence of TRIM5α in these macaques could explain the reports showing that SHIV (HIV-1 vif gene was replaced by the vif genes from SIVmac239 and HIV-2ROD) cause a low-level transient infection in pig-tailed macaques. Several studies on SIV infection in macaques have shown virus replication in macrophages in the brain leading to encephalopathy, which was correlated with the ability of the virus (SIV/17E-Fr [267]. SIVsmmFGb [268] to replicate productively in cultured macrophages derived from peripheral blood of animals.

In AIDS research, Cynomolgus macaques have been utilized to a lesser extent than have rhesus and pig-tailed macaques. This is primarily due to the low level of immunogenic characterization of this species. In terms of viral loads and CD4+ T cell turnover, investigations have revealed that SIVmac251 and SHIV89.6P are much less pathogenic in cynomolgus macaques than in Indian or Chinese rhesus macaques [250]. Interestingly, it was found that the TRIM5 genes of cynomolgus macaques are also quite diverse, encoding polymorphisms in both TRIM5α and TRIMCyp isoforms, including TRIMCyp proteins, that could differentially restrict HIV-1 [269]. The limited MHC diversity of these macaques has dramatically increased interest in using the cynomolgus macaques in AIDS research [270, 271]. These animals originated from a small population brought to the Mauritius island approximately 400–500 years ago by European sailors. To date, only seven MHC haplotypes have been identified in Mauritian cynomolgus macaques [272]. Thus, unlike other primate models, for which it is only possible to select MHC-defined animals, studies can be done in Mauritian cynomolgus macaques using haplotype-matched or completely MHC-identical animals. This unique feature was used to demonstrate an MHC heterozygote advantage for the ability to control SIV infection [273].

11.3. Non-human primate models of NeuroAIDS

Not all SIV strains have the same pathogenic effects on the CNS. Several SIV models have been studied to understand the pathogenesis of disease in the brain. The early models of NeuroAIDS utilized Indian rhesus or pigtailed macaques infected with different viral clones that resulted in immune suppression, macrophage tropism for CNS infection leading to AIDS, and SIV encephalitis (SIVE), in approximately 30 percent of rhesus macaques, and higher frequencies in pigtailed macaques [274]. Strategies involving depletion of CD4+ and CD8+ T lymphocytes and/or serial passage of virus through monkeys for enhanced macrophage replication and neurotropism have been used in different studies [257, 275277].

A widespread and widely used rhesus model includes monoclonal antibody (mAb) depletion of CD8+ lymphocytes and infection with the viral swarm SIVmac251. This resulted in persistent and high viremia leading to the rapid onset of AIDS (3–4 months versus 2–3 years in non-lymphocyte depleted animals) and a high incidence of SIVE (greater than 80% versus approximately 30% in non-depleted animals) [278]. This model yielded several significant findings, including high monocyte turnover from the bone marrow [279], accumulation of macrophages in the CNS [280], recruitment of inflammatory macrophages into the CNS [281], elevated levels of soluble CD163 in plasma that correlated with neurocognitive impairment [282], accumulation of cardiac macrophages [283], and identified the role of macrophages in SIV neuropathogenesis [282].

Infection of rhesus macaques with both SIVmac251 and SIVmac239 leads to the generation of virus-infected cells in the CSF throughout the acute phase, which can last for 2 to 10 weeks [284286]. During this acute phase of SIV infection, neuropathogenesis is characterized by meningitis and accumulations of mononuclear cells in the Virchow-Robin spaces in the brain [287]. Studies have shown that lesions caused by SIVmac239 are less severe; instead, meningitis disappears at the end of the acute phase of infection [284, 287]. Encephalitic changes leading to dense infiltrations of mononuclear cells lead to neuroinflammatory milieu [288]. Unlike human syndrome, inflammatory lesions in the meninges and neuropil occur even when the CD4+ T lymphocytes are not depleted [288]. Furthermore, SIV lesions are much more severe in the CNS compared with those seen in HIV-infected brains. Interesting studies have shown that SIVmac251-infected macaques have altered evoked potentials in the brain, a drastic increase in the number of activated CD8+ T cells and neuroinflammatory milieu in the brains during early SIV infection [289292]. SIVmac182 infected macaques have also been shown to cause CNS neuropathogenesis. SIV-specific CD8+ CTLs were detected in the CSF of infected macaques early after infection [290]. This model has been used to study the role of immune responses in mediating CNS injury during the relatively early stage of SIV-induced disease. The primary advantage of these models is that they exhibit a very fast rate of AIDS progression post-SIV infection compared to decades it takes in humans infected with HIV-1. However, since SIVmac251 comprises of both macrophage-tropic and non-macrophage-tropic strains, it does not faithfully model native HIV transmission, which is essentially universally via non-macrophage-tropic transmitter/founder viruses. The molecular clone of SIVmac239, a non-macrophage-tropic transmitter/founder virus, is ideal for studying the mucosal transmission of HIV-1 but is not a good model for studying the CNS infection of HIV, where the virus predominantly targets the microglia and perivascular macrophages.

The other common model for SIV neuropathogenesis involves mAb depletion of CD4+ T lymphocytes before SIV infection, resulting in increased viral replication, progressively leading to AIDS [275]. Earlier depletion of CD4+T cells is associated with a higher percentage and incidence of infected macrophages in the periphery and the CNS. Additionally, this model also showed higher levels of SIV infection in the parenchymal microglia [275]. These data underscore the role of macrophages/microglia in AIDS pathogenesis. In addition to SIV, chimeric simian-human immune deficiency virus (SHIV) models have also been shown to exhibit rapid depletion of CD4+ T cells in Indian rhesus macaques, leading to the rapid development of AIDS and CNS pathology [293, 294]. These models demonstrate the significance of CD8+ T and CD4+ T lymphocytes in the development and progression of AIDS and NeuroAIDS. A third model of rapid and predictable progression to AIDS with a high incidence of SIVE involved pigtailed macaques infected with a combination of a neurovirulent molecular clone that replicates efficiently in macrophages, SIVmac/17E-Fr, and an immune-suppressive viral swarm SIVsm/Delta B670 that rapidly depletes CD4+ T lymphocytes within 2–4 weeks post intravenous inoculation [295, 296]. This model has been comprehensively used to study the mechanisms involved in CNS, peripheral neuropathy, and cardiac pathogenesis and understand innate immunity in the CNS and CNS-based therapeutics for humans [297299]. The SIVmacR71/17E virus has been shown to be neurotropic and causes neuropathogenesis in Indian Rhesus macaques. For example, a study by Bokhari et al. has shown viral replication in the brain and histolytic nodules in the basal ganglia of this virus infected for 52 weeks [300]. Several studies have shown that there was neuroinflammation in the brains of the SIV infected macaques, and this neuroinflammation was due to activation of inflammasome [301303] as well to deposition of neurotoxic amyloids in the brains [304]. SIV R71/17E infected monkeys also exhibit deficits in motor performance [305].

Several published studies have shown that serially passaged SIVSmE543 and SIVsm804E result in early viral entry into the CNS with a high incidence of SIVE. Gabuzda and collaborators similarly identified a variant of macrophage-tropic SIV env glycoprotein in the blood of SIV251-infected animals and developed a molecular clone that enters the CNS early after inoculation, resulting in a higher incidence of SIVE [257, 277]. Overall, all the models discussed above, with or without cART, provide fundamental data underscoring the role of viral evolution and macrophage tropism that facilitates rapid and consistent development of AIDS with neuropathogenesis, that takes into account the role of acquired and innate immunity in CNS disease. Studies with macaques have shown that monocyte trafficking in and out of the CNS establishes and maintains a CNS reservoir of productive SIV infection [282]. Earlier studies have shown that cART treatment demonstrated a biphasic activation and increased numbers of CD14+CD16+ monocytes first at the time of infection and later with the emergence of AIDS [282, 306]. Although the SIV-infected macaque model of neuroAIDS best recapitulates HIV neuropathogenesis, there are limitations associated with it. Not all of the SIV infected macaques predictably develop CNS disease – it is suggested that only 25% of these animals develop CNS infection. Secondly, the prolonged progression (1 to 3 years) towards AIDS development limits its usefulness. This has led to other macaque models that take a prolonged time for disease development and progression towards AIDS [267, 307]. The pigtailed macaques co-inoculated intravenously with a combination of two viruses: SIV/17E-Fr (neurovirulent) and SIV/DeltaB670 (immunosuppressive and neurovirulent virus swarm) resulted in depletion of CD4+ T-lymphocytes and efficient virus replication in the CNS macrophages within three months post-inoculation in almost 90% of inoculated macaques [295, 308]. In these models, virus load in the CSF during terminal infection and the CSF:plasma ratio of monocyte chemoattractant protein (MCP)-1 determines the severity of encephalitis.

11.4. SIV and recombinant SHIV challenge strains

It is evident that not all macaque models of SIV infection accurately replicate HIV-1 infection due to several factors, including TRIM5α-mediated restriction. Additionally, the SIV strains also differ from HIV-1 in several ways. For instance, SIVsmm and SIVmac have an accessory gene, vpx (replaces vpu gene), that is not present in HIV-1. HIV-1 and SIVsmm/SIVmac, however, have 53% nucleotide identity. As a result, there has been significant interest in engineering minimally modified HIV-1 strains capable of replicating and recapitulating disease characteristics in macaques.

11.5. SIVs

SIV was first isolated from rhesus macaques with a virulent form of immunodeficiency characterized by opportunistic infections and tumors at the New England Primate Research Center, Southborough, Massachusetts, USA [309]. However, the source of the virus in macaques was later traced to an outbreak of lymphoma in the 1970s at the California National Primate Research Center in Davis, California, USA [310]. Both SIVmac251 and smE660 are independent viral isolates that are commonly used as uncloned viruses. Infectious molecular clones of SIV were derived to obtain genetically defined challenge viruses and proviral DNAs that were suitable for genetic manipulation [311]. SIVmac239 and SIVsmE543-3 are pathogenic molecular clones related in their passage history to SIVmac251 and SIVsmE660, respectively [312, 313]. These viruses’ unique characteristics are that they use CCR5 as a co-receptor, replicate predominantly in memory CD4+ T cells, and express Env glycoproteins that are resistant to neutralizing antibodies. The susceptibility of SIVsmE543-3, and SIVsmE660, to certain rhesus macaque TRIM5 proteins, is an essential factor to be considered in using these challenge viruses. Several investigators are now working to adapt SIVsmE543-3 and SIVsmE660 to restrictive rhesus macaque TRIM5 variants to generate challenge viruses with more uniform resistance in this model [314].

12. Drugs of abuse and SIV

Different drugs of abuse, including opiates, have been found to potentiate HIV replication by diminishing the host-immune responses and accelerating disease progression. For example, SIV-infected Indian Rhesus macaques receiving morphine showed diminished antibody responses, which resulted in severe disease progression, decreased viral clearance rate, increased viral load, and marked depletion of CD4+ T cells [315]. In another study, it was observed that morphine-dependent SIV-infected Indian Rhesus macaques showed enhanced mortality rates, reduced weight gain, higher numbers of circulating CD4+ and CD8+T, higher levels of CSF and plasma viral load, and increased influx of infected monocyte/macrophages into the brains compared to the SIV alone group not dependent on morphine [300]. Indian Rhesus macaques chronically infected with SIVmac239 with/without opioid dependence demonstrated higher viral titer in the presence of morphine compared to the SIV group not administered morphine. An interesting finding from this study was that PBMCs isolated from non-infected animals exposed to morphine resulted in the increased expression of β-chemokine receptor 5 (CCR5). Since both SIV and HIV require CCR5 for cell entry, the unique role of morphine in accelerating viral infection was attributed to morphine-mediated induction of CCR5 expression [316]. Another interesting study demonstrated that morphine-exposed SIVmac239-infected cells (hybrid human B and T cell-line) accumulated in the G1 phase, thus indicating cell cycle arrest. Additionally, morphine was shown to elevate the PKC activity levels and p-ERK1/2, thus implicating that the calcium-PKC-MAPK cascade was involved in morphine-mediated survival of SIV-infected cells [317]. A fascinating study has shown that morphine-dependent (26 weeks), SIV-infected (33 weeks) Indian Rhesus macaques showed a decrease in motor skill tasks in comparison to the morphine or SIV groups alone [318], indicating thereby that morphine could potentiate behavioral impairments in SIV infection. SIV-infected, morphine-dependent macaques also demonstrated significant time-dependent changes in their gut microbial communities (16S) and metabolic profiles. Altered metabolites such as bile acids, sphingolipids, and serotonin could negatively impact the host [319]. It was also shown that there was increased expression of miR-29b in the basal ganglia of morphine-dependent SIV-infected Indian Rhesus macaques compared with the SIV-infected controls and that this involved downregulation of the miR-29b target gene- Platelet-derived growth factor (PDGF)-B, which is a growth factor for neuronal survival [320].

It must be noted that studies on the role of opioids in SIV-mediated immune responses and neuropathogenesis have yielded conflicting results. Some studies have shown that morphine administration prevented developing enzyme-linked immunosorbent spot-forming cell-mediated immune responses in R17/71E-infected rhesus macaques [289]. In contrast, SIV R17/77E infected animals developed cell-mediated immune responses [289]. Additionally, in these animals, morphine treatment did not affect viremia, cerebrospinal fluid viral titers, or survival throughout the study, and the drug was shown to potentiate a more significant build-up of the virus in the brains of infected animals. Furthermore, histopathological changes in the brains showed increased demyelination in morphine-dependent, SIV-infected animals compared to SIV alone group [289]. In another study, morphine dependence exerted a protective effect in a SIVsmm9/rhesus monkey model; however, this study lacked control animals, and virus replication was compared with historical controls [321]. Another study from the same group showed that the mixture of 2 SHIVs (SHIVKU and SHIV89.6P) and a neuropathogenic SIV (R71/17E) proved to be an excellent model to develop the rapid and reproducible disease in rhesus macaques [322]. Morphine administration in this model showed no alterations in peak viral loads in rhesus macaques; however, there was higher viral replication at six weeks post-infection in the brain and blood [323]. In light of these findings, controlled studies in non-human primates are warranted to study the effects of drugs of abuse on viral pathogenesis.

Morphine and psychostimulants such as cocaine have also been shown to reduce CD8+ T cells during acute and late stages of SIV infection. However, cocaine administration failed to impact CSF or plasma viral load or CSF CCL2 and IL-6 levels. These findings also suggested that there was no effect of cocaine on SIV encephalitis’s severity compared to vehicle-treated macaques. Additionally, cocaine administration failed to impact neuroinflammation or neurodegeneration, as determined by IFN-β, MxA, CCL2, IL-6, TNFα, IFNγ, and indolamine 2,3-deoxygenase mRNA levels. There was also no change in the APP levels in the presence of cocaine. The executive function of inhibitory control was not impaired in cocaine-treated or control animals following SIV infection. However, animals receiving 3.2 mg/kg/day cocaine performed slower in a bimanual motor test. Chronic administration of cocaine in macaques thus inflicted only minor changes in behavior, encephalitis severity, CNS inflammation/neurodegeneration, and virus replication in SIV-infected pigtailed macaques. These findings thus implicate that cocaine likely only exerts modest effects on the progression of NeuroAIDS in HIV-infected individuals [324].

Interesting studies with another drug of abuse- methamphetamine showed that methamphetamine administration increased viral loads in the brains of SIV- infected macaques but did not alter levels of virus in the plasma. Additionally, methamphetamine treatment also resulted in the activation of natural killer cells. Given the prevalence of methamphetamine use in HIV-infected and HIV at-risk populations, these findings reveal the likely deleterious effects of methamphetamine abuse in such individuals [325]. In another study, microglia were isolated from the brains of macaques infected with SIV, followed by treatment with methamphetamine. Results showed that methamphetamine alone altered the expression of different genes associated with immune pathways, inflammation, and chemotaxis. Most interestingly, it was shown that methamphetamine exposure enhanced the expression of viral entry coreceptors - CXCR4 and CCR5. The increase in CCR5 expression was confirmed in the brain and correlated with increased brain viral load. From this study, it was concluded that methamphetamine increased CCR5-expressing cells, which, in turn, was associated with a concomitant increase in the viral load [326]. Another critical study has shown that cytokines such as IL-15 have been upregulated in astrocytes by methamphetamine, resulting in T cells’ proliferation and an inflammatory phenotype in the brain. Therefore, methamphetamine and IL15 could play critical roles in developing and aggravating CNS immune-mediated inflammatory pathology in HIV-infected drug abusers [327]. Most of the studies have been performed using Indian Rhesus macaques, and the results demonstrate that these drugs of abuse facilitate increased viral replication, which exacerbates viral pathogenesis in the CNS and the periphery.

13. Feline immunodeficiency virus (FIV) model

The feline immunodeficiency virus (FIV) model is an animal model of HIV-1 infection that has been useful in understanding the pathogenesis of AIDS [328]. FIV is a naturally occurring retrovirus that infects domestic and non-domestic feline species and causes an immunodeficiency syndrome, along with clinical symptoms that resemble those observed in humans infected with HIV-1, including symptoms such as periodontitis, neurologic dysfunction, encephalitis, and lymphoma [329333]. Since the model was developed in 1987, it has proven helpful in several aspects. The primary mode of natural FIV transmission is similar to that of HIV-1 in humans, such as blood-borne infection facilitated by fighting and biting and transmission through transmucosal routes (rectal and vaginal mucosa and perinatal). Furthermore, oral opportunistic infections and changes in the salivary/oral microbiota are also prevalent in FIV infection, and these are associated with the development of oral inflammatory lesions [334337]. Several early investigations demonstrated that FIV infection results in progressive CD4 T-cell depletion with a primary immune deficiency that causes opportunistic infections, the hallmark of HIV AIDS [328, 329, 331, 338341]. According to the type and severity of the clinical signs of these infections, FIV disease progression can be divided into four or five stages, described as acute or primary, chronic asymptomatic, persistent generalized lymphadenopathy (PGL), AIDS-related complex (ARC), and feline AIDS (FAIDS), similar to that observed in HIV infection in humans. Intriguingly, FIV infection exhibits a prolonged asymptomatic phase that can last ten years or more, much like HIV-1 infection in humans [331, 342].

FIV is used as a model to study HIV pathogenesis, including immune dysfunction and neurological deficits, and is useful for exploring vaccine development. FIV infection results in acute viremia-associated immune system dysfunction such as CD4 T-cell depletion, peripheral lymphadenopathy, disruption of immune cell function, neutropenia, deterioration of major lymphoid tissues and organs of the hosts, as well as chronic persistent and opportunistic infections due to immunosuppression [330, 338, 343]. Studies on this model have shown that FIV enters the CNS during the acute stage of infection, either via the trafficking of infected monocytes and lymphocytes or by the penetration of free virus across the blood-brain or blood-CSF, barriers similar to HIV-1 [67, 343, 344]. Intriguingly, FIV can infect cells such as microglia and astrocytes in the CNS, and these infected cells serve as a reservoir for latent viral persistence, similar to HIV. The fundamental neurodegenerative and neuropathologic events, including proliferation and activation of glial cells and neuroinflammation, myelin degradation, and neuronal injury/loss observed in HIV, are replicated in the FIV infectious model [344348]. FIV-infected animals also exhibit the clinical manifestations of the neuropathology of dementia and cognitive-motor processing deficits often observed in HIV infection. This makes FIV a useful animal model to investigate the behavioral pathogenesis seen with retrovirus infection of the CNS [347, 349]. This model has also proven useful in developing and testing neuroprotective therapeutics such as neurotrophin ligands, which prevent the delayed accumulation of intracellular calcium and the decreased cytoskeletal damage of neuronal dendrites [345, 350]. The FIV model is well suited for investigating the effects of drugs of abuse on HIV-associated neuropathogenesis since these models has several commonalities with HIV infection as mentioned above, including 1) the common structural and biochemical properties of FIV and HIV; 2) similar clinical syndromes; 3) a reproducible disease model, with an accurate recapitulation of general and neurological disease progression; 4) FIV infects the CNS and shows neuropathology including neurodegeneration and cognitive and motor dysfunctions [351]. This model was used in previous reports to assess morphine-mediated viral progression [352], behavioral and physical signs of opiate exposure, and evidence of withdrawal symptoms [353]. Morphine-exposed FIV-infected cats failed to exhibit enhanced viral load or severity of FIV-related disease. Morphine demonstrated a protective effect on FIV-associated changes in brain stem auditory evoked potentials. However, morphine-treated cats exhibited clear behavioral and physical signs of opiate exposure and evidence of withdrawal symptoms such as increased cortisol levels [352354]. In another study, the FIV model was used to explore the potential effects of methamphetamine on lentivirus disease progression, and it was reported that felines are susceptible to low doses of methamphetamine and drug exposure increased FIV replication, as well as the selective and chronic chemical neurotoxicity and disease progression [355]. In another study, FIV-infected cats showed alterations in brain metabolites, such as elevated gamma-aminobutyric acid (GABA) upon methamphetamine exposure [356]. The FIV model is thus suitable for studies of pathogenesis, drug addiction, withdrawal effects, and the development of therapeutics in the context of human HIV infection. However, ethical challenges in terms of animal use and cost have limited this model’s extensive use. Furthermore, this model is not suitable for vaccine development studies and is not closely related to HIV genetic makeup, unlike non-human primate models [357]. The longer incubation periods before the onset of overt disease, less genetic overlap with the HIV genome, and the relatively poor knowledge of the feline immune system are the major factors that limit the applications of the FIV animal model. Importantly, FIV has does not have certain accessory genes that are seen in HIV-1 [329]. Unlike HIV, FIV utilizes CD134 instead of CD4 as the main binding receptor, thereby infecting B cells and CD8+ T cells and CD4+ T cells and macrophages [358, 359]. Since the infection target is different in FIV than HIV/SIV, the reservoir is also different. These factors limit the use of the FIV animal model for cure research.

14. Conclusion and future perspectives

The development of animal models that address HIV and drug abuse comorbidity continues to be an ongoing effort to improve the utility of the existing models. Here we reviewed the existing animal models, discussed their advantages and disadvantages, and learned lessons that could shed light on advancing the field. Chronic low-grade neuroinflammation observed in HIV-positive, drug-abusing humans have been recapitulated in these rodent models. Interestingly, HIV-induced behavioral changes in cognitive performance are also seen in models such as the HIV-1Tg rats, suggesting that viral proteins contribute to cognitive decline and functional deficits, which could be exacerbated by drugs of abuse. Although these rodent models have provided a vast amount of information on mechanisms of HIV-1 and drug abuse interactions, validation in non-human primate models and eventually in human clinical studies is essential. The SIV and SHIV macaque models have improved our understanding of the interaction of HIV-1 and drug abuse. Being primates, they offer the advantage of similarity with humans in immune architecture and response to HIV-1 infection. The heterogeneity and variation in genotypes of macaques, however, poses a challenge, but at the same time also offers an opportunity to examine mechanisms and test therapeutics in a system that most accurately replicates the human disease. Consequently, further development and refinement of these models are required to understand better the effect of drug abuse comorbidity on HIV-1 infection.

Table 1.

List of animal models used in NeuroHIV research.

Model Mutation/changes Expression / Target organ Phenotype Applications Advantages Limitations Drug abuse
iTat mice [50, 130, 131, 360]. Tat protein expressed in the brain astrocytes under inducible promoter. Brain–astrocytes. Increased astrocytosis, loss of neuronal dendrites, and neuroinflammation. To study the effects of Tat on astrocytes including the astrocyte-mediated Tat neurotoxicity, Tat-impaired neurogenesis, Tat-induced loss of neuronal integrity, and exosome-associated Tat release and uptake. Depletion of T cells is not necessary to create these models. Expressed only in brain astrocytes, Expression of tat protein is driven by doxycycline, which has neuroprotective property [131, 360].
Doxycycline inducible promoter is leaky, so small amounts of Tat protein is expressed in early stages of development, because of this leakiness an immune response did not seen after tat induction [158].
Methamphetamine [140], cocaine [141, 361, 362], Morphine [142144]
EcoHIV mice [61, 151] The coding region of gp120 in HIV virus was replaced with mouse leukemia virus gp80 to infect mice. Immune Cells, Spleen, and Brain. EcoHIV-infected animals showed more macrophages and monocytes. In the absence of T cells, HIV-infected macrophages are sufficient for induction of cognitive impairment in mice. Good experimental model for NeuroAIDS Therapy. EcoHIV is a good model to study some of the neuron type-specific neurotoxic functions of HIV.
EcoHIV constitutively expressed in mice for up to 16 months.
EcoHIV enters cells through CAT-1 receptor [61]. Methamphetamine [160], Morphine [154].
gp120 transgenic mice [167] A transgenic gp120 mice model was generated by inserting the portion of the HIV env gene that encodes gp120 into the mouse genome under the control of a glial fibrillary acidic protein (GFAP) promoter, resulting in gp120 protein expression by brain astrocytes. Brain-astrocytes. Abnormal dendrite morphology, decreased synaptic and dendritic density, Increased astrocytosis. Good animal model for neuroAIDS research Non-infectious model, Neurodegene ration and glial cell activation mimics that of HIV encephalitis. Protein express only in brain astrocytes. Viral protein is present throughout the lifespan might promote survival of abnormally robust neuronal populations [158]. Methamphetamine [174176], Morphine: [138, 172].
SCID [191, 363-366] Mutation in the gene for protein kinase, DNA activated, catalytic polypeptide (PRKDC). Thymus, spleen, lymph node. Absence of functional B cells and T cells. To study the human immune system and human disease in a small animal model. Useful tool for Immunology, Inflammation and Autoimmunity Research, Xenograft/transplant host AIDS research tool. In certain circumstances like unsterile conditions, can lead to activation of NK cells, leakiness of SCID mice.[195, 196] Morphine [191], Methamphetamine [192, 193], cocaine: [194, 367].
Tg 26 Mice [103, 108] Loss of neuronal dendrites, loss of synapses. Kidney disease, HIV-associated nephropathy.
Tg26 mice exhibit proteinuria.
Ubiquitous Loss of neuronal dendrites, loss of synapses. Kidney disease, HIV- associated nephropathy.
Tg26 mice exhibit proteinuria.
Animal model for human HIV-associated nephropathy (HIVAN). HIV-1-Tg26 mouse model has been used extensively to study the pathogenesis of HIVAN because these mice develop renal disease mimicking human HIV associated Nephropathy. Approximately 15% of Tg 26 mice spontaneously develop leukemia/lymphoma [368]. Cocaine [116], Morphine [117], Methamphetamine [173, 369].
HIV transgenic rats [56, 220, 370] Plasmid containing non-infectious HIV provirus and host cell flanking regions were microinjected into fertilized rat eggs. lymph nodes, thymus, liver, kidney, and spleen. Cataracts, Kidney and heart damage and loss of lymphocytes. Good model for neuroHIV study. Transgenic rat is a noninfectious small-animal model of HIV-1 pathogenesis because it efficiently express viral gene in lymph nodes, spleen, thymus, and blood. Useful to study the mechanisms underlyin HIV - associated disease progression caused by HIV proteins. This model is not useful to study the initial infection stage of HIV and there is no viral replication in the HIV-1Tg rat [228]. Methamphetami ne[224, 371, 372], Morphine, [225, 226], Cocaine: [227].
SIV and SHIV monkey models [235238, 309] Intravenous inoculation of SIV virus. Brain, Heart, and Kidney. Reduced number of CD4+ T cells. Good model system for HIV pathogenesis and vaccine research. SIV is genetically, antigenically and morphologically close to HIV.
SIV infected monkeys are immunosuppressed and display neuropathological and behavioral features are identical to HIV-infected humans.
higher maintenance costs, Not susceptible to HIV-1 [240]. Morphine [239, 315, 316], Cocaine [324], Methamphetami ne[325327].
FIV model [328] FIV is a natural lentiviral. The integrated provirus possesses gag, pol, and env genes, common elements of all retroviruses. Liver, Kidney, Brain and Lung. Progressive CD4+ T cell depletion, a deficit in the humoral immune response, and the dysregulation of cytokine expression. Valuable animal model for the development of antiretroviral drugs. Not large in size and not difficult to handle, The models can be infected with FIV, FeLV and develop immuno deficiency-like syndrome. Cat’s genetic makeup is far related to human FIV, FeLV are not closely related to HIV [330, 357]. Methamphetami ne[354, 355], Morphine[352, 353],
scid-hu-Thy/Liv mice. [373, 374] SCID mice are implanted with human liver and thymus. Thymus and Liver Decrease in the CD4/CD8 ratio This model is useful to study HIV pathogenesis and to test the efficacy of antiretroviral drugs for HIV replication. These mice have human liver and thymus cells, which are the target organ for antiretroviral drug uptake and action. Substantial degree of ‘leakiness’ in certain mouse strains (leading to the development of mouse T cells and B cells in older animals. This model cannot be used to study mucosal transmission of HIV-1 [240]. N/A
Humanized microglial mice. [216, 375, 376] Mice express a humanized form of the microglial growth factor CSF1, which is essential for microglial maintenance, and lack of two immune factors required for rejection of foreign cells. Brain N/A This model will be useful to investigate how human and mouse microglia function differently in normal and HIV conditions. Useful to study the role of human microglia in brain development and degeneration. Useful to study the pathophysiology of the human cells within an intact brain. Many human cytokines and other factors are species specific [216, 376] . N/A

Highlights:

  • HIV-1 infection and drug abuse are intertwined epidemics.

  • Animal models are necessary to decipher the impact of drugs on HAND progression.

  • Non-human primates are gold-standard model in this study but very expensive.

  • Several strategies have also been used to develop several rodent models of HAND.

  • These rodent models serve the purpose of modeling some unique aspects of HAND.

Acknowledgments

We would like to thank all members of our laboratories, past and present, for their invaluable contributions and apologize to all those collaborators whose work could not be discussed in this review owing to space limitations.

Funding

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

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Declaration of Competing of Interest

The authors state they have no conflicts of interest.

References:

  • [1].Sacktor N, McArthur J, Prospects for therapy of HIV-associated neurologic diseases, J Neurovirol 3 (1997) 89–101. [DOI] [PubMed] [Google Scholar]
  • [2].Trujillo JR, Jaramillo-Rangel G, Ortega-Martinez M, Penalva de Oliveira AC, Vidal JE, Bryant J, Gallo RC, International NeuroAIDS: prospects of HIV-1 associated neurological complications, Cell Res 15 (2005) 962–969. [DOI] [PubMed] [Google Scholar]
  • [3].Saylor D, Dickens AM, Sacktor N, Haughey N, Slusher B, Pletnikov M, Mankowski JL, Brown A, Volsky DJ, McArthur JC, HIV-associated neurocognitive disorder--pathogenesis and prospects for treatment, Nat Rev Neurol 12 (2016) 234–248. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [4].McGuire JL, Barrett JS, Vezina HE, Spitsin S, Douglas SD, Adjuvant therapies for HIV-associated neurocognitive disorders, Ann Clin Transl Neurol 1 (2014) 938–952. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [5].Omondi FH, Chandrarathna S, Mujib S, Brumme CJ, Jin SW, Sudderuddin H, Miller RL, Rahimi A, Laeyendecker O, Bonner P, Yue FY, Benko E, Kovacs CM, Brockman MA, Ostrowski M, Brumme ZL, HIV Subtype and Nef-Mediated Immune Evasion Function Correlate with Viral Reservoir Size in Early-Treated Individuals, J Virol 93 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [6].Madrid-Elena N, Garcia-Bermejo ML, Serrano-Villar S, Diaz-de Santiago A, Sastre B, Gutierrez C, Dronda F, Coronel Diaz M, Dominguez E, Lopez-Huertas MR, Hernandez-Novoa B, Moreno S, Maraviroc Is Associated with Latent HIV-1 Reactivation through NF-kappaB Activation in Resting CD4(+) T Cells from HIV-Infected Individuals on Suppressive Antiretroviral Therapy, J Virol 92 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [7].Cherner M, Masliah E, Ellis RJ, Marcotte TD, Moore DJ, Grant I, Heaton RK, Neurocognitive dysfunction predicts postmortem findings of HIV encephalitis, Neurology 59 (2002) 1563–1567. [DOI] [PubMed] [Google Scholar]
  • [8].Diesing TS, Swindells S, Gelbard H, Gendelman HE, HIV-1-associated dementia: a basic science and clinical perspective, AIDS Read 12 (2002) 358–368. [PubMed] [Google Scholar]
  • [9].Kim S, Hahn YK, Podhaizer EM, McLane VD, Zou S, Hauser KF, Knapp PE, A central role for glial CCR5 in directing the neuropathological interactions of HIV-1 Tat and opiates, J Neuroinflammation 15 (2018) 285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [10].Debalkie Animut M, Sorrie MB, Birhanu YW, Teshale MY, High prevalence of neurocognitive disorders observed among adult people living with HIV/AIDS in Southern Ethiopia: A cross-sectional study, PLoS One 14 (2019) e0204636. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [11].Tsegaw M, Andargie G, Alem G, Tareke M, Screening HIV-associated neurocognitive disorders (HAND) among HIV positive patients attending antiretroviral therapy in South Wollo, Ethiopia, J Psychiatr Res 85 (2017) 37–41. [DOI] [PubMed] [Google Scholar]
  • [12].Belete T, Medfu G, Yemiyamrew E, Prevalence of HIV Associated Neurocognitive Deficit among HIV Positive People in Ethiopia: A Cross Sectional Study at Ayder Referral Hospital, Ethiop J Health Sci 27 (2017) 67–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [13].Sanchez AB, Varano GP, de Rozieres CM, Maung R, Catalan IC, Dowling CC, Sejbuk NE, Hoefer MM, Kaul M, Antiretrovirals, Methamphetamine, and HIV-1 Envelope Protein gp120 Compromise Neuronal Energy Homeostasis in Association with Various Degrees of Synaptic and Neuritic Damage, Antimicrob Agents Chemother 60 (2016) 168–179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [14].Fogel GB, Lamers SL, Levine AJ, Valdes-Sueiras M, McGrath MS, Shapshak P, Singer EJ, Factors related to HIV-associated neurocognitive impairment differ with age, J Neurovirol 21 (2015) 56–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [15].Gaskill PJ, Calderon TM, Coley JS, Berman JW, Drug induced increases in CNS dopamine alter monocyte, macrophage and T cell functions: implications for HAND, J Neuroimmune Pharmacol 8 (2013) 621–642. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [16].Byrd DA, Fellows RP, Morgello S, Franklin D, Heaton RK, Deutsch R, Atkinson JH, Clifford DB, Collier AC, Marra CM, Gelman B, McCutchan JA, Duarte NA, Simpson DM, McArthur J, Grant I, Group C, Neurocognitive impact of substance use in HIV infection, J Acquir Immune Defic Syndr 58 (2011) 154–162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [17].Arce Renteria M, Byrd D, Coulehan K, Miranda C, Fuentes A, Rosario AK, Morris EP, Rivera Mindt M, Neurocognitive intra-individual variability within HIV+ adults with and without current substance use, Neuropsychology 34 (2020) 321–330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [18].Noel RJ Jr., Rivera-Amill V, Buch S, Kumar A, Opiates, immune system, acquired immunodeficiency syndrome, and nonhuman primate model, J Neurovirol 14 (2008) 279–285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [19].Hauser KF, Fitting S, Dever SM, Podhaizer EM, Knapp PE, Opiate drug use and the pathophysiology of neuroAIDS, Curr HIV Res 10 (2012) 435–452. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [20].Atluri VS, Editorial: HIV and Illicit Drugs of Abuse, Front Microbiol 7 (2016) 221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [21].Bloomquist A, Vaidya NK, Modelling the risk of HIV infection for drug abusers, J Biol Dyn (2020) 1–24. [DOI] [PubMed] [Google Scholar]
  • [22].Doke M, Jeganathan V, McLaughlin JP, Samikkannu T, HIV-1 Tat and cocaine impact mitochondrial epigenetics: effects on DNA methylation, Epigenetics (2020) 1–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [23].Buch S, Periyasamy P, Thangaraj A, Sil S, Chivero ET, Tripathi A, Opioid-Mediated HIV-1 Immunopathogenesis, J Neuroimmune Pharmacol (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [24].Desai N, Burns L, Gong Y, Zhi K, Kumar A, Summers N, Kumar S, Cory TJ, An update on drug-drug interactions between antiretroviral therapies and drugs of abuse in HIV systems, Expert Opin Drug Metab Toxicol 16 (2020) 1005–1018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [25].Sil S, Niu F, Chivero ET, Singh S, Periyasamy P, Buch S, Role of Inflammasomes in HIV-1 and Drug Abuse Mediated Neuroinflammaging, Cells 9 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [26].Barbour AJ, Hauser KF, McQuiston AR, Knapp PE, HIV and opiates dysregulate K(+)- Cl(−) cotransporter 2 (KCC2) to cause GABAergic dysfunction in primary human neurons and Tat-transgenic mice, Neurobiol Dis 141 (2020) 104878. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [27].Murphy A, Barbaro J, Martinez-Aguado P, Chilunda V, Jaureguiberry-Bravo M, Berman JW, The Effects of Opioids on HIV Neuropathogenesis, Front Immunol 10 (2019) 2445. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [28].Adams JW, Marshall BDL, Mohd Salleh NA, Barrios R, Nolan S, Milloy MJ, Receipt of opioid agonist treatment halves the risk of HIV-1 RNA viral load rebound through improved ART adherence for HIV-infected women who use illicit drugs, Drug Alcohol Depend 206 (2020) 107670. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [29].Ladak F, Socias E, Nolan S, Dong H, Kerr T, Wood E, Montaner J, Milloy MJ, Substance use patterns and HIV-1 RNA viral load rebound among HIV-positive illicit drug users in a Canadian setting, Antivir Ther 24 (2019) 19–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [30].Milloy MJ, Kerr T, Buxton J, Rhodes T, Krusi A, Guillemi S, Hogg R, Montaner J, Wood E, Social and environmental predictors of plasma HIV RNA rebound among injection drug users treated with antiretroviral therapy, J Acquir Immune Defic Syndr 59 (2012) 393–399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [31].Lyu Y, Kaddour H, Kopcho S, Panzner TD, Shouman N, Kim EY, Martinson J, McKay H, Martinez-Maza O, Margolick JB, Stapleton JT, Okeoma CM, Human Immunodeficiency Virus (HIV) Infection and Use of Illicit Substances Promote Secretion of Semen Exosomes that Enhance Monocyte Adhesion and Induce Actin Reorganization and Chemotactic Migration, Cells 8 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [32].Rodriguez M, Lapierre J, Ojha CR, Pawitwar S, Karuppan MKM, Kashanchi F, El-Hage N, Morphine counteracts the antiviral effect of antiretroviral drugs and causes upregulation of p62/SQSTM1 and histone-modifying enzymes in HIV-infected astrocytes, J Neurovirol 25 (2019) 263–274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [33].Gardner MB, SIV infected rhesus macaques: an AIDS model for immunoprevention and immunotherapy, Adv Exp Med Biol 251 (1989) 279–293. [DOI] [PubMed] [Google Scholar]
  • [34].Zhou Y, Bao R, Haigwood NL, Persidsky Y, Ho WZ, SIV infection of rhesus macaques of Chinese origin: a suitable model for HIV infection in humans, Retrovirology 10 (2013) 89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [35].Matsuda K, Riddick NE, Lee CA, Puryear SB, Wu F, Lafont BAP, Whitted S, Hirsch VM, A SIV molecular clone that targets the CNS and induces neuroAIDS in rhesus macaques, PLoS Pathog 13 (2017) e1006538. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [36].von Herrath M, Oldstone MB, Fox HS, Simian immunodeficiency virus (SIV)-specific CTL in cerebrospinal fluid and brains of SIV-infected rhesus macaques, J Immunol 154 (1995) 5582–5589. [PubMed] [Google Scholar]
  • [37].Gardner M, A historical perspective: Simian AIDS-an accidental windfall, J Med Primatol 45 (2016) 212–214. [DOI] [PubMed] [Google Scholar]
  • [38].Persidsky Y, Gendelman HE, Murine models for human immunodeficiency virus type 1-associated dementia: the development of new treatment testing paradigms, J Neurovirol 8 Suppl 2 (2002) 49–52. [DOI] [PubMed] [Google Scholar]
  • [39].Persidsky Y, Limoges J, McComb R, Bock P, Baldwin T, Tyor W, Patil A, Nottet HS, Epstein L, Gelbard H, Flanagan E, Reinhard J, Pirruccello SJ, Gendelman HE, Human immunodeficiency virus encephalitis in SCID mice, Am J Pathol 149 (1996) 1027–1053. [PMC free article] [PubMed] [Google Scholar]
  • [40].Tyor WR, Power C, Gendelman HE, Markham RB, A model of human immunodeficiency virus encephalitis in scid mice, Proc Natl Acad Sci U S A 90 (1993) 8658–8662. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [41].Dou H, Grotepas CB, McMillan JM, Destache CJ, Chaubal M, Werling J, Kipp J, Rabinow B, Gendelman HE, Macrophage delivery of nanoformulated antiretroviral drug to the brain in a murine model of neuroAIDS, J Immunol 183 (2009) 661–669. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [42].Limoges J, Persidsky Y, Bock P, Gendelman HE, Dexamethasone therapy worsens the neuropathology of human immunodeficiency virus type 1 encephalitis in SCID mice, J Infect Dis 175 (1997) 1368–1381. [DOI] [PubMed] [Google Scholar]
  • [43].Griffin WC 3rd, Middaugh LD, Cook JE, Tyor WR, The severe combined immunodeficient (SCID) mouse model of human immunodeficiency virus encephalitis: deficits in cognitive function, J Neurovirol 10 (2004) 109–115. [DOI] [PubMed] [Google Scholar]
  • [44].Aksenov MY, Hasselrot U, Bansal AK, Wu G, Nath A, Anderson C, Mactutus CF, Booze RM, Oxidative damage induced by the injection of HIV-1 Tat protein in the rat striatum, Neurosci Lett 305 (2001) 5–8. [DOI] [PubMed] [Google Scholar]
  • [45].Hayman M, Arbuthnott G, Harkiss G, Brace H, Filippi P, Philippon V, Thomson D, Vigne R, Wright A, Neurotoxicity of peptide analogues of the transactivating protein tat from Maedi-Visna virus and human immunodeficiency virus, Neuroscience 53 (1993) 1–6. [DOI] [PubMed] [Google Scholar]
  • [46].Jones M, Olafson K, Del Bigio MR, Peeling J, Nath A, Intraventricular injection of human immunodeficiency virus type 1 (HIV-1) tat protein causes inflammation, gliosis, apoptosis, and ventricular enlargement, J Neuropathol Exp Neurol 57 (1998) 563–570. [DOI] [PubMed] [Google Scholar]
  • [47].Lu SM, Tremblay ME, King IL, Qi J, Reynolds HM, Marker DF, Varrone JJ, Majewska AK, Dewhurst S, Gelbard HA, HIV-1 Tat-induced microgliosis and synaptic damage via interactions between peripheral and central myeloid cells, PLoS One 6 (2011) e23915. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [48].Hammond JW, Qiu WQ, Marker DF, Chamberlain JM, Greaves-Tunnell W, Bellizzi MJ, Lu SM, Gelbard HA, HIV Tat causes synapse loss in a mouse model of HIV-associated neurocognitive disorder that is independent of the classical complement cascade component C1q, Glia 66 (2018) 2563–2574. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [49].Dutta R, Roy S, Chronic morphine and HIV-1 Tat promote differential central nervous system trafficking of CD3+ and Ly6C+ immune cells in a murine Streptococcus pneumoniae infection model, J Neuroinflammation 12 (2015) 120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [50].Kim BO, Liu Y, Ruan Y, Xu ZC, Schantz L, He JJ, Neuropathologies in transgenic mice expressing human immunodeficiency virus type 1 Tat protein under the regulation of the astrocyte-specific glial fibrillary acidic protein promoter and doxycycline, Am J Pathol 162 (2003) 1693–1707. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [51].Fan Y, He JJ, HIV-1 Tat Induces Unfolded Protein Response and Endoplasmic Reticulum Stress in Astrocytes and Causes Neurotoxicity through Glial Fibrillary Acidic Protein (GFAP) Activation and Aggregation, J Biol Chem 291 (2016) 22819–22829. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [52].Langford D, Oh Kim B, Zou W, Fan Y, Rahimain P, Liu Y, He JJ, Doxycycline-inducible and astrocyte-specific HIV-1 Tat transgenic mice (iTat) as an HIV/neuroAIDS model, J Neurovirol 24 (2018) 168–179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [53].Maung R, Hoefer MM, Sanchez AB, Sejbuk NE, Medders KE, Desai MK, Catalan IC, Dowling CC, de Rozieres CM, Garden GA, Russo R, Roberts AJ, Williams R, Kaul M, CCR5 knockout prevents neuronal injury and behavioral impairment induced in a transgenic mouse model by a CXCR4-using HIV-1 glycoprotein 120, J Immunol 193 (2014) 1895–1910. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [54].Asensio VC, Maier J, Milner R, Boztug K, Kincaid C, Moulard M, Phillipson C, Lindsley K, Krucker T, Fox HS, Campbell IL, Interferon-independent, human immunodeficiency virus type 1 gp120-mediated induction of CXCL10/IP-10 gene expression by astrocytes in vivo and in vitro, J Virol 75 (2001) 7067–7077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [55].Dickens AM, Yoo SW, Chin AC, Xu J, Johnson TP, Trout AL, Hauser KF, Haughey NJ, Chronic low-level expression of HIV-1 Tat promotes a neurodegenerative phenotype with aging, Sci Rep 7 (2017) 7748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [56].Reid W, Sadowska M, Denaro F, Rao S, Foulke J Jr., Hayes N, Jones O, Doodnauth D, Davis H, Sill A, O’Driscoll P, Huso D, Fouts T, Lewis G, Hill M, Kamin-Lewis R, Wei C, Ray P, Gallo RC, Reitz M, Bryant J, An HIV-1 transgenic rat that develops HIV-related pathology and immunologic dysfunction, Proc Natl Acad Sci U S A 98 (2001) 9271–9276. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [57].Vigorito M, LaShomb AL, Chang SL, Spatial learning and memory in HIV-1 transgenic rats, J Neuroimmune Pharmacol 2 (2007) 319–328. [DOI] [PubMed] [Google Scholar]
  • [58].Moran LM, Aksenov MY, Booze RM, Webb KM, Mactutus CF, Adolescent HIV-1 transgenic rats: evidence for dopaminergic alterations in behavior and neurochemistry revealed by methamphetamine challenge, Curr HIV Res 10 (2012) 415–424. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [59].Moran LM, Booze RM, Webb KM, Mactutus CF, Neurobehavioral alterations in HIV-1 transgenic rats: evidence for dopaminergic dysfunction, Exp Neurol 239 (2013) 139–147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [60].Hadas E, Borjabad A, Chao W, Saini M, Ichiyama K, Potash MJ, Volsky DJ, Testing antiretroviral drug efficacy in conventional mice infected with chimeric HIV-1, AIDS 21 (2007) 905–909. [DOI] [PubMed] [Google Scholar]
  • [61].Gu CJ, Borjabad A, Hadas E, Kelschenbach J, Kim BH, Chao W, Arancio O, Suh J, Polsky B, McMillan J, Edagwa B, Gendelman HE, Potash MJ, Volsky DJ, EcoHIV infection of mice establishes latent viral reservoirs in T cells and active viral reservoirs in macrophages that are sufficient for induction of neurocognitive impairment, PLoS Pathog 14 (2018) e1007061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [62].Gorantla S, Makarov E, Finke-Dwyer J, Castanedo A, Holguin A, Gebhart CL, Gendelman HE, Poluektova L, Links between progressive HIV-1 infection of humanized mice and viral neuropathogenesis, Am J Pathol 177 (2010) 2938–2949. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [63].Kanzaki LI, Looney DJ, Feline immunodeficiency virus: a concise review, Front Biosci 9 (2004) 370–377. [DOI] [PubMed] [Google Scholar]
  • [64].Mitchell TW, Buckmaster PS, Hoover EA, Whalen LR, Dudek FE, Neuron loss and axon reorganization in the dentate gyrus of cats infected with the feline immunodeficiency virus, J Comp Neurol 411 (1999) 563–577. [PubMed] [Google Scholar]
  • [65].Gunn-Moore DA, McVee J, Bradshaw JM, Pearson GR, Head E, Gunn-Moore FJ, Ageing changes in cat brains demonstrated by beta-amyloid and AT8-immunoreactive phosphorylated tau deposits, J Feline Med Surg 8 (2006) 234–242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [66].Asproni P, Abramo F, Millanta F, Lorenzi D, Poli A, Amyloidosis in association with spontaneous feline immunodeficiency virus infection, J Feline Med Surg 15 (2013) 300–306. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [67].Burkhard MJ, Dean GA, Transmission and immunopathogenesis of FIV in cats as a model for HIV, Curr HIV Res 1 (2003) 15–29. [DOI] [PubMed] [Google Scholar]
  • [68].Policicchio BB, Pandrea I, Apetrei C, Animal Models for HIV Cure Research, Front Immunol 7 (2016) 12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [69].Durvasula R, Miller TR, Substance abuse treatment in persons with HIV/AIDS: challenges in managing triple diagnosis, Behav Med 40 (2014) 43–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [70].Tyagi M, Bukrinsky M, Simon GL, Mechanisms of HIV Transcriptional Regulation by Drugs of Abuse, Curr HIV Res 14 (2016) 442–454. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [71].Doherty MC, Garfein RS, Monterroso E, Brown D, Vlahov D, Correlates of HIV infection among young adult short-term injection drug users, AIDS 14 (2000) 717–726. [DOI] [PubMed] [Google Scholar]
  • [72].Mimiaga MJ, Reisner SL, Grasso C, Crane HM, Safren SA, Kitahata MM, Schumacher JE, Mathews WC, Mayer KH, Substance use among HIV-infected patients engaged in primary care in the United States: findings from the Centers for AIDS Research Network of Integrated Clinical Systems cohort, Am J Public Health 103 (2013) 1457–1467. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [73].Haverkos HW, HIV/AIDS and drug abuse: epidemiology and prevention, J Addict Dis 17 (1998) 91–103. [DOI] [PubMed] [Google Scholar]
  • [74].Altice FL, Kamarulzaman A, Soriano VV, Schechter M, Friedland GH, Treatment of medical, psychiatric, and substance-use comorbidities in people infected with HIV who use drugs, Lancet 376 (2010) 367–387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [75].Rabkin JG, McElhiney MC, Ferrando SJ, Mood and substance use disorders in older adults with HIV/AIDS: methodological issues and preliminary evidence, AIDS 18 Suppl 1 (2004) S43–48. [PubMed] [Google Scholar]
  • [76].Chander G, Himelhoch S, Moore RD, Substance abuse and psychiatric disorders in HIV-positive patients: epidemiology and impact on antiretroviral therapy, Drugs 66 (2006) 769–789. [DOI] [PubMed] [Google Scholar]
  • [77].Erickson A, Becker M, Shaw S, Kasper K, Keynan Y, Substance use and its impact on care outcomes among HIV-infected individuals in Manitoba, AIDS Care 27 (2015) 1168–1173. [DOI] [PubMed] [Google Scholar]
  • [78].Lucas GM, Substance abuse, adherence with antiretroviral therapy, and clinical outcomes among HIV-infected individuals, Life Sci 88 (2011) 948–952. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [79].Meade CS, Sikkema KJ, HIV risk behavior among adults with severe mental illness: a systematic review, Clin Psychol Rev 25 (2005) 433–457. [DOI] [PubMed] [Google Scholar]
  • [80].Klinkenberg WD, Sacks S, Hiv HO /Aids Treatment Adherence, G. Cost Study, Mental disorders and drug abuse in persons living with HIV/AIDS, AIDS Care 16 Suppl 1 (2004) S22–42. [DOI] [PubMed] [Google Scholar]
  • [81].Gum AM, King-Kallimanis B, Kohn R, Prevalence of mood, anxiety, and substance-abuse disorders for older Americans in the national comorbidity survey-replication, Am J Geriatr Psychiatry 17 (2009) 769–781. [DOI] [PubMed] [Google Scholar]
  • [82].Cook JA, Burke-Miller JK, Steigman PJ, Schwartz RM, Hessol NA, Milam J, Merenstein DJ, Anastos K, Golub ET, Cohen MH, Prevalence, Comorbidity, and Correlates of Psychiatric and Substance Use Disorders and Associations with HIV Risk Behaviors in a Multisite Cohort of Women Living with HIV, AIDS Behav 22 (2018) 3141–3154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [83].Choi SKY, Boyle E, Cairney J, Grootendorst P, Gardner S, Collins EJ, Kendall C, Rourke SB, Study OC, Impact of depression and recreational drug use on emergency department encounters and hospital admissions among people living with HIV in Ontario: A secondary analysis using the OHTN cohort study, PLoS One 13 (2018) e0195185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [84].Chibanda D, Benjamin L, Weiss HA, Abas M, Mental, neurological, and substance use disorders in people living with HIV/AIDS in low- and middle-income countries, 67 Suppl 1 (2014) S54–67. [DOI] [PubMed] [Google Scholar]
  • [85].Bing EG, Burnam MA, Longshore D, Fleishman JA, Sherbourne CD, London AS, Turner BJ, Eggan F, Beckman R, Vitiello B, Morton SC, Orlando M, Bozzette SA, Ortiz-Barron L, Shapiro M, Psychiatric disorders and drug use among human immunodeficiency virus-infected adults in the United States, Arch Gen Psychiatry 58 (2001) 721–728. [DOI] [PubMed] [Google Scholar]
  • [86].Berger-Greenstein JA, Cuevas CA, Brady SM, Trezza G, Richardson MA, Keane TM, Major depression in patients with HIV/AIDS and substance abuse, AIDS Patient Care STDS 21 (2007) 942–955. [DOI] [PubMed] [Google Scholar]
  • [87].Millar BM, Starks TJ, Gurung S, Parsons JT, The Impact of Comorbidities, Depression, and Substance Use Problems on Quality of Life Among Older Adults Living With HIV, AIDS Behav 21 (2017) 1684–1690. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [88].Hauser KF, Knapp PE, Interactions of HIV and drugs of abuse: the importance of glia, neural progenitors, and host genetic factors, Int Rev Neurobiol 118 (2014) 231–313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [89].Norman LR, Basso M, Kumar A, Malow R, Neuropsychological consequences of HIV and substance abuse: a literature review and implications for treatment and future research, Curr Drug Abuse Rev 2 (2009) 143–156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [90].Buckner CM, Luers AJ, Calderon TM, Eugenin EA, Berman JW, Neuroimmunity and the blood-brain barrier: molecular regulation of leukocyte transmigration and viral entry into the nervous system with a focus on neuroAIDS, J Neuroimmune Pharmacol 1 (2006) 160–181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [91].Borgmann K, Ghorpade A, HIV-1, methamphetamine and astrocytes at neuroinflammatory Crossroads, Front Microbiol 6 (2015) 1143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [92].Roy S, Ninkovic J, Banerjee S, Charboneau RG, Das S, Dutta R, Kirchner VA, Koodie L, Ma J, Meng J, Barke RA, Opioid drug abuse and modulation of immune function: consequences in the susceptibility to opportunistic infections, J Neuroimmune Pharmacol 6 (2011) 442–465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [93].Hermes DJ, Jacobs IR, Key MC, League AF, Yadav-Samudrala BJ, Xu C, McLane VD, Nass SR, Jiang W, Meeker RB, Ignatowska-Jankowska BM, Lichtman AH, Li Z, Wu Z, Yuan H, Knapp PE, Hauser KF, Fitting S, Escalating morphine dosing in HIV-1 Tat transgenic mice with sustained Tat exposure reveals an allostatic shift in neuroinflammatory regulation accompanied by increased neuroprotective non-endocannabinoid lipid signaling molecules and amino acids, J Neuroinflammation 17 (2020) 345. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [94].Chilunda V, Calderon TM, Martinez-Aguado P, Berman JW, The impact of substance abuse on HIV-mediated neuropathogenesis in the current ART era, Brain Res 1724 (2019) 146426. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [95].Patel SH, Ismaiel OA, Mylott WR Jr., Yuan M, McClay JL, Paris JJ, Hauser KF, McRae M, Cell-type specific differences in antiretroviral penetration and the effects of HIV-1 Tat and morphine among primary human brain endothelial cells, astrocytes, pericytes, and microglia, Neurosci Lett 712 (2019) 134475. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [96].Leibrand CR, Paris JJ, Jones AM, Masuda QN, Halquist MS, Kim WK, Knapp PE, Kashuba ADM, Hauser KF, McRae M, HIV-1 Tat and opioids act independently to limit antiretroviral brain concentrations and reduce blood-brain barrier integrity, J Neurovirol 25 (2019) 560–577. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [97].Patel S, Leibrand CR, Palasuberniam P, Couraud PO, Weksler B, Jahr FM, McClay JL, Hauser KF, McRae M, Effects of HIV-1 Tat and Methamphetamine on Blood-Brain Barrier Integrity and Function In Vitro, Antimicrob Agents Chemother 61 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [98].Cornwell WD, Sriram U, Seliga A, Zuluaga-Ramirez V, Gajghate S, Rom S, Winfield M, Heldt NA, Ambrose D, Rogers TJ, Persidsky Y, Tobacco smoke and morphine alter peripheral and CNS inflammation following HIV infection in a humanized mouse model, Sci Rep 10 (2020) 13977. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [99].Goldman D, Oroszi G, Ducci F, The genetics of addictions: uncovering the genes, Nat Rev Genet 6 (2005) 521–532. [DOI] [PubMed] [Google Scholar]
  • [100].Ball D, Addiction science and its genetics, Addiction 103 (2008) 360–367. [DOI] [PubMed] [Google Scholar]
  • [101].Engleman EA, Katner SN, Neal-Beliveau BS, Caenorhabditis elegans as a Model to Study the Molecular and Genetic Mechanisms of Drug Addiction, Prog Mol Biol Transl Sci 137 (2016) 229–252. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [102].Bevilacqua L, Goldman D, Genes and addictions, Clin Pharmacol Ther 85 (2009) 359–361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [103].Dickie P, Felser J, Eckhaus M, Bryant J, Silver J, Marinos N, Notkins AL, HIV-associated nephropathy in transgenic mice expressing HIV-1 genes, Virology 185 (1991) 109–119. [DOI] [PubMed] [Google Scholar]
  • [104].Abad ML, Verdura T, Vela A, Iglesias MJ, Gutierrez D, Veiga M, Aguilera A, Regueiro BJ, Construction and characterization of a minimized version of the HIV-1 pNL4-3 plasmid and its application for pseudotyping HIV-1 vectors, Mol Biotechnol 28 (2004) 87–95. [DOI] [PubMed] [Google Scholar]
  • [105].Felser JM, Klimkait T, Silver J, A syncytia assay for human immunodeficiency virus type I (HIV-I) envelope protein and its use in studying HIV-I mutations, Virology 170 (1989) 566–570. [DOI] [PubMed] [Google Scholar]
  • [106].De SK, Wohlenberg CR, Marinos NJ, Doodnauth D, Bryant JL, Notkins AL, Human chorionic gonadotropin hormone prevents wasting syndrome and death in HIV-1 transgenic mice, J Clin Invest 99 (1997) 1484–1491. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [107].Rosenstiel P, Gharavi A, D’Agati V, Klotman P, Transgenic and infectious animal models of HIV-associated nephropathy, J Am Soc Nephrol 20 (2009) 2296–2304. [DOI] [PubMed] [Google Scholar]
  • [108].Kopp JB, Klotman ME, Adler SH, Bruggeman LA, Dickie P, Marinos NJ, Eckhaus M, Bryant JL, Notkins AL, Klotman PE, Progressive glomerulosclerosis and enhanced renal accumulation of basement membrane components in mice transgenic for human immunodeficiency virus type 1 genes, Proc Natl Acad Sci U S A 89 (1992) 1577–1581. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [109].Ray PE, Hu CA, Advances in our understanding of the pathogenesis of HIV-1 associated nephropathy in children, Future Virol 6 (2011) 883–894. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [110].Zhong J, Zuo Y, Ma J, Fogo AB, Jolicoeur P, Ichikawa I, Matsusaka T, Expression of HIV-1 genes in podocytes alone can lead to the full spectrum of HIV-1-associated nephropathy, Kidney Int 68 (2005) 1048–1060. [DOI] [PubMed] [Google Scholar]
  • [111].Gharavi AG, Ahmad T, Wong RD, Hooshyar R, Vaughn J, Oller S, Frankel RZ, Bruggeman LA, D’Agati VD, Klotman PE, Lifton RP, Mapping a locus for susceptibility to HIV-1-associated nephropathy to mouse chromosome 3, Proc Natl Acad Sci U S A 101 (2004) 2488–2493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [112].Putatunda R, Zhang Y, Li F, Yang XF, Barbe MF, Hu W, Adult neurogenic deficits in HIV-1 Tg26 transgenic mice, J Neuroinflammation 15 (2018) 287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [113].Niu F, Yao H, Zhang W, Sutliff RL, Buch S, Tat 101-mediated enhancement of brain pericyte migration involves platelet-derived growth factor subunit B homodimer: implications for human immunodeficiency virus-associated neurocognitive disorders, J Neurosci 34 (2014) 11812–11825. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [114].Schwartz L, Civitello L, Dunn-Pirio A, Ryschkewitsch S, Berry E, Cavert W, Kinzel N, Lawrence DM, Hazra R, Major EO, Evidence of human immunodeficiency virus type 1 infection of nestin-positive neural progenitors in archival pediatric brain tissue, J Neurovirol 13 (2007) 274–283. [DOI] [PubMed] [Google Scholar]
  • [115].Putatunda R, Zhang Y, Li F, Fagan PR, Zhao H, Ramirez SH, Pratico D, Barbe MF, Hu W, Sex-specific neurogenic deficits and neurocognitive disorders in middle-aged HIV-1 Tg26 transgenic mice, Brain Behav Immun 80 (2019) 488–499. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [116].Mohseni Ahooyi T, Shekarabi M, Torkzaban B, Langford TD, Burdo TH, Gordon J, Datta PK, Amini S, Khalili K, Dysregulation of Neuronal Cholesterol Homeostasis upon Exposure to HIV-1 Tat and Cocaine Revealed by RNA-Sequencing, Sci Rep 8 (2018) 16300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [117].Meng J, Banerjee S, Zhang L, Sindberg G, Moidunny S, Li B, Robbins DJ, Girotra M, Segura A, Ramakrishnan S, Roy S, Opioids Impair Intestinal Epithelial Repair in HIV-Infected Humanized Mice, Front Immunol 10 (2019) 2999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [118].Das SR, Jameel S, Biology of the HIV Nef protein, Indian J Med Res 121 (2005) 315–332. [PubMed] [Google Scholar]
  • [119].Lindemann D, Wilhelm R, Renard P, Althage A, Zinkernagel R, Mous J, Severe immunodeficiency associated with a human immunodeficiency virus 1 NEF/3’-long terminal repeat transgene, J Exp Med 179 (1994) 797–807. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [120].Benson RE, Sanfridson A, Ottinger JS, Doyle C, Cullen BR, Downregulation of cell-surface CD4 expression by simian immunodeficiency virus Nef prevents viral super infection, J Exp Med 177 (1993) 1561–1566. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [121].Jolicoeur P, The CD4C/HIV(Nef)transgenic model of AIDS, Curr HIV Res 9 (2011) 524–530. [DOI] [PubMed] [Google Scholar]
  • [122].Rahim MM, Chrobak P, Hu C, Hanna Z, Jolicoeur P, Adult AIDS-like disease in a novel inducible human immunodeficiency virus type 1 Nef transgenic mouse model: CD4+ T-cell activation is Nef dependent and can occur in the absence of lymphophenia, J Virol 83 (2009) 11830–11846. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [123].Acharjee S, Branton WG, Vivithanaporn P, Maingat F, Paul AM, Dickie P, Baker GB, Power C, HIV-1 Nef expression in microglia disrupts dopaminergic and immune functions with associated mania-like behaviors, Brain Behav Immun 40 (2014) 74–84. [DOI] [PubMed] [Google Scholar]
  • [124].Zou W, Zhang L, Mouse Models for Studies of In Vivo Functions of HIV-1 Nef, AIDS Rev 18 (2016) 158–165. [PubMed] [Google Scholar]
  • [125].Jamieson BD, Aldrovandi GM, Planelles V, Jowett JB, Gao L, Bloch LM, Chen IS, Zack JA, Requirement of human immunodeficiency virus type 1 nef for in vivo replication and pathogenicity, J Virol 68 (1994) 3478–3485. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [126].Radja F, Kay DG, Albrecht S, Jolicoeur P, Oligodendrocyte-specific expression of human immunodeficiency virus type 1 Nef in transgenic mice leads to vacuolar myelopathy and alters oligodendrocyte phenotype in vitro, J Virol 77 (2003) 11745–11753. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [127].Goudreau G, Carpenter S, Beaulieu N, Jolicoeur P, Vacuolar myelopathy in transgenic mice expressing human immunodeficiency virus type 1 proteins under the regulation of the myelin basic protein gene promoter, Nat Med 2 (1996) 655–661. [DOI] [PubMed] [Google Scholar]
  • [128].Chompre G, Cruz E, Maldonado L, Rivera-Amill V, Porter JT, Noel RJ Jr., Astrocytic expression of HIV-1 Nef impairs spatial and recognition memory, Neurobiol Dis 49 (2013) 128–136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [129].Sami Saribas A, Cicalese S, Ahooyi TM, Khalili K, Amini S, Sariyer IK, HIV-1 Nef is released in extracellular vesicles derived from astrocytes: evidence for Nef-mediated neurotoxicity, Cell Death Dis 8 (2017) e2542. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [130].Fitting S, Xu R, Bull C, Buch SK, El-Hage N, Nath A, Knapp PE, Hauser KF, Interactive comorbidity between opioid drug abuse and HIV-1 Tat: chronic exposure augments spine loss and sublethal dendritic pathology in striatal neurons, Am J Pathol 177 (2010) 1397–1410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [131].Cho W, Hagemann TL, Johnson DA, Johnson JA, Messing A, Dual transgenic reporter mice as a tool for monitoring expression of glial fibrillary acidic protein, J Neurochem 110 (2009) 343–351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [132].Zou W, Kim BO, Zhou BY, Liu Y, Messing A, He JJ, Protection against human immunodeficiency virus type 1 Tat neurotoxicity by Ginkgo biloba extract EGb 761 involving glial fibrillary acidic protein, Am J Pathol 171 (2007) 1923–1935. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [133].Rahimian P, He JJ, HIV-1 Tat-shortened neurite outgrowth through regulation of microRNA-132 and its target gene expression, J Neuroinflammation 13 (2016) 247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [134].Fields JA, Dumaop W, Crews L, Adame A, Spencer B, Metcalf J, He J, Rockenstein E, Masliah E, Mechanisms of HIV-1 Tat neurotoxicity via CDK5 translocation and hyper-activation: role in HIV-associated neurocognitive disorders, Curr HIV Res 13 (2015) 43–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [135].Fan Y, Gao X, Chen J, Liu Y, He JJ, HIV Tat Impairs Neurogenesis through Functioning As a Notch Ligand and Activation of Notch Signaling Pathway, J Neurosci 36 (2016) 11362–11373. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [136].Rahimian P, He JJ, Exosome-associated release, uptake, and neurotoxicity of HIV-1 Tat protein, J Neurovirol 22 (2016) 774–788. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [137].Kadri F, Pacifici M, Wilk A, Parker-Struckhoff A, Del Valle L, Hauser KF, Knapp PE, Parsons A, Jeansonne D, Lassak A, Peruzzi F, HIV-1-Tat Protein Inhibits SC35-mediated Tau Exon 10 Inclusion through Up-regulation of DYRK1A Kinase, J Biol Chem 290 (2015) 30931–30946. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [138].Fitting S, Scoggins KL, Xu R, Dever SM, Knapp PE, Dewey WL, Hauser KF, Morphine efficacy is altered in conditional HIV-1 Tat transgenic mice, Eur J Pharmacol 689 (2012) 96–103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [139].Hauser KF, Hahn YK, Adjan VV, Zou S, Buch SK, Nath A, Bruce-Keller AJ, Knapp PE, HIV-1 Tat and morphine have interactive effects on oligodendrocyte survival and morphology, Glia 57 (2009) 194–206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [140].Basova LV, Kesby JP, Kaul M, Semenova S, Marcondes MCG, Systems Biology Analysis of the Antagonizing Effects of HIV-1 Tat Expression in the Brain over Transcriptional Changes Caused by Methamphetamine Sensitization, Viruses 12 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [141].Paris JJ, Carey AN, Shay CF, Gomes SM, He JJ, McLaughlin JP, Effects of conditional central expression of HIV-1 tat protein to potentiate cocaine-mediated psychostimulation and reward among male mice, Neuropsychopharmacology 39 (2014) 380–388. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [142].Gonek M, McLane VD, Stevens DL, Lippold K, Akbarali HI, Knapp PE, Dewey WL, Hauser KF, Paris JJ, CCR5 mediates HIV-1 Tat-induced neuroinflammation and influences morphine tolerance, dependence, and reward, Brain Behav Immun 69 (2018) 124–138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [143].Fitting S, Stevens DL, Khan FA, Scoggins KL, Enga RM, Beardsley PM, Knapp PE, Dewey WL, Hauser KF, Morphine Tolerance and Physical Dependence Are Altered in Conditional HIV-1 Tat Transgenic Mice, J Pharmacol Exp Ther 356 (2016) 96–105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [144].Hahn YK, Paris JJ, Lichtman AH, Hauser KF, Sim-Selley LJ, Selley DE, Knapp PE, Central HIV-1 Tat exposure elevates anxiety and fear conditioned responses of male mice concurrent with altered mu-opioid receptor-mediated G-protein activation and beta-arrestin 2 activity in the forebrain, Neurobiol Dis 92 (2016) 124–136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [145].Hahn YK, Masvekar RR, Xu R, Hauser KF, Knapp PE, Chronic HIV-1 Tat and HIV reduce Rbfox3/NeuN: evidence for sex-related effects, Curr HIV Res 13 (2015) 10–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [146].Zhao X, Fan Y, Vann PH, Wong JM, Sumien N, He JJ, Long-term HIV-1 Tat Expression in the Brain Led to Neurobehavioral, Pathological, and Epigenetic Changes Reminiscent of Accelerated Aging, Aging Dis 11 (2020) 93–107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [147].He X, Yang W, Zeng Z, Wei Y, Gao J, Zhang B, Li L, Liu L, Wan Y, Zeng Q, Gong Z, Liu L, Zhang H, Li Y, Yang S, Hu T, Wu L, Masliah E, Huang S, Cao H, NLRP3-dependent pyroptosis is required for HIV-1 gp120-induced neuropathology, Cell Mol Immunol 17 (2020) 283–299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [148].Lehmann MH, Lehmann JM, Erfle V, Nef-induced CCL2 Expression Contributes to HIV/SIV Brain Invasion and Neuronal Dysfunction, Front Immunol 10 (2019) 2447. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [149].Mamik MK, Hui E, Branton WG, McKenzie BA, Chisholm J, Cohen EA, Power C, HIV-1 Viral Protein R Activates NLRP3 Inflammasome in Microglia: implications for HIV-1 Associated Neuroinflammation, J Neuroimmune Pharmacol 12 (2017) 233–248. [DOI] [PubMed] [Google Scholar]
  • [150].Churchill MJ, Wesselingh SL, Cowley D, Pardo CA, McArthur JC, Brew BJ, Gorry PR, Extensive astrocyte infection is prominent in human immunodeficiency virus-associated dementia, Ann Neurol 66 (2009) 253–258. [DOI] [PubMed] [Google Scholar]
  • [151].Potash MJ, Chao W, Bentsman G, Paris N, Saini M, Nitkiewicz J, Belem P, Sharer L, Brooks AI, Volsky DJ, A mouse model for study of systemic HIV-1 infection, antiviral immune responses, and neuroinvasiveness, Proc Natl Acad Sci U S A 102 (2005) 3760–3765. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [152].Saini M, Hadas E, Volsky DJ, Potash MJ, Vaccine-induced protection from infection of mice by chimeric human immunodeficiency virus type 1, EcoHIV/NL4-3, Vaccine 25 (2007) 8660–8663. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [153].Roshorm Y, Hong JP, Kobayashi N, McMichael AJ, Volsky DJ, Potash MJ, Takiguchi M, Hanke T, Novel HIV-1 clade B candidate vaccines designed for HLA-B*5101(+) patients protected mice against chimaeric ecotropic HIV-1 challenge, Eur J Immunol 39 (2009) 1831–1840. [DOI] [PubMed] [Google Scholar]
  • [154].Sindberg GM, Sharma U, Banerjee S, Anand V, Dutta R, Gu CJ, Volsky DJ, Roy S, An infectious murine model for studying the systemic effects of opioids on early HIV pathogenesis in the gut, J Neuroimmune Pharmacol 10 (2015) 74–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [155].Kim BH, Kelschenbach J, Borjabad A, Hadas E, He H, Potash MJ, Nedelcovych MT, Rais R, Haughey NJ, McArthur JC, Slusher BS, Volsky DJ, Intranasal insulin therapy reverses hippocampal dendritic injury and cognitive impairment in a model of HIV-associated neurocognitive disorders in EcoHIV-infected mice, AIDS 33 (2019) 973–984. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [156].Kelschenbach J, He H, Kim BH, Borjabad A, Gu CJ, Chao W, Do M, Sharer LR, Zhang H, Arancio O, Potash MJ, Volsky DJ, Efficient Expression of HIV in Immunocompetent Mouse Brain Reveals a Novel Nonneurotoxic Viral Function in Hippocampal Synaptodendritic Injury and Memory Impairment, mBio 10 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [157].Dong B, Borjabad A, Kelschenbach J, Chao W, Volsky DJ, Potash MJ, Prevention and treatment of HIV infection and cognitive disease in mice by innate immune responses, Brain Behav Immun Health 3 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [158].Jaeger LB, Nath A, Modeling HIV-associated neurocognitive disorders in mice: new approaches in the changing face of HIV neuropathogenesis, Dis Model Mech 5 (2012) 313–322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [159].Gorantla S, Poluektova L, Gendelman HE, Rodent models for HIV-associated neurocognitive disorders, Trends Neurosci 35 (2012) 197–208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [160].Skowronska M, McDonald M, Velichkovska M, Leda AR, Park M, Toborek M, Methamphetamine increases HIV infectivity in neural progenitor cells, J Biol Chem 293 (2018) 296–311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [161].Schneider J, Kaaden O, Copeland TD, Oroszlan S, Hunsmann G, Shedding and interspecies type sero-reactivity of the envelope glycopolypeptide gp120 of the human immunodeficiency virus, J Gen Virol 67 ( Pt 11) (1986) 2533–2538. [DOI] [PubMed] [Google Scholar]
  • [162].Kanmogne GD, Kennedy RC, Grammas P, HIV-1 gp120 proteins and gp160 peptides are toxic to brain endothelial cells and neurons: possible pathway for HIV entry into the brain and HIV-associated dementia, J Neuropathol Exp Neurol 61 (2002) 992–1000. [DOI] [PubMed] [Google Scholar]
  • [163].Brenneman DE, McCune SK, Mervis RF, Hill JM, gp120 as an etiologic agent for NeuroAIDS: neurotoxicity and model systems, Adv Neuroimmunol 4 (1994) 157–165. [DOI] [PubMed] [Google Scholar]
  • [164].Gajardo-Gomez R, Santibanez CA, Labra VC, Gomez GI, Eugenin EA, Orellana JA, HIV gp120 Protein Increases the Function of Connexin 43 Hemichannels and Pannexin-1 Channels in Astrocytes: Repercussions on Astroglial Function, Int J Mol Sci 21 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [165].D’Hooge R, Franck F, Mucke L, De Deyn PP, Age-related behavioural deficits in transgenic mice expressing the HIV-1 coat protein gp120, Eur J Neurosci 11 (1999) 4398–4402. [DOI] [PubMed] [Google Scholar]
  • [166].Thaney VE, Sanchez AB, Fields JA, Minassian A, Young JW, Maung R, Kaul M, Transgenic mice expressing HIV-1 envelope protein gp120 in the brain as an animal model in neuroAIDS research, J Neurovirol 24 (2018) 156–167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [167].Toggas SM, Masliah E, Rockenstein EM, Rall GF, Abraham CR, Mucke L, Central nervous system damage produced by expression of the HIV-1 coat protein gp120 in transgenic mice, Nature 367 (1994) 188–193. [DOI] [PubMed] [Google Scholar]
  • [168].Krucker T, Toggas SM, Mucke L, Siggins GR, Transgenic mice with cerebral expression of human immunodeficiency virus type-1 coat protein gp120 show divergent changes in short- and long-term potentiation in CA1 hippocampus, Neuroscience 83 (1998) 691–700. [DOI] [PubMed] [Google Scholar]
  • [169].Lee MH, Wang T, Jang MH, Steiner J, Haughey N, Ming GL, Song H, Nath A, Venkatesan A, Rescue of adult hippocampal neurogenesis in a mouse model of HIV neurologic disease, Neurobiol Dis 41 (2011) 678–687. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [170].Okamoto S, Kang YJ, Brechtel CW, Siviglia E, Russo R, Clemente A, Harrop A, McKercher S, Kaul M, Lipton SA, HIV/gp120 decreases adult neural progenitor cell proliferation via checkpoint kinase-mediated cell-cycle withdrawal and G1 arrest, Cell Stem Cell 1 (2007) 230–236. [DOI] [PubMed] [Google Scholar]
  • [171].Fields JA, Serger E, Campos S, Divakaruni AS, Kim C, Smith K, Trejo M, Adame A, Spencer A, Rockenstein E, Murphy AN, Ellis RJ, Letendre S, Grant I, Masliah E, HIV alters neuronal mitochondrial fission/fusion in the brain during HIV-associated neurocognitive disorders, Neurobiol Dis 86 (2016) 154–169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [172].Shi Y, Yuan S, Tang SJ, Morphine and HIV-1 gp120 cooperatively promote pathogenesis in the spinal pain neural circuit, Mol Pain 15 (2019) 1744806919868380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [173].Kesby JP, Hubbard DT, Markou A, Semenova S, Expression of HIV gp120 protein increases sensitivity to the rewarding properties of methamphetamine in mice, Addict Biol 19 (2014) 593–605. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [174].Hoefer MM, Sanchez AB, Maung R, de Rozieres CM, Catalan IC, Dowling CC, Thaney VE, Pina-Crespo J, Zhang D, Roberts AJ, Kaul M, Combination of methamphetamine and HIV-1 gp120 causes distinct long-term alterations of behavior, gene expression, and injury in the central nervous system, Exp Neurol 263 (2015) 221–234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [175].Henry BL, Geyer MA, Buell MR, Perry W, Young JW, Minassian A, Translational Methamphetamine ARCG, Prepulse inhibition in HIV-1 gp120 transgenic mice after withdrawal from chronic methamphetamine, Behav Pharmacol 25 (2014) 12–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [176].Kesby JP, Heaton RK, Young JW, Umlauf A, Woods SP, Letendre SL, Markou A, Grant I, Semenova S, Methamphetamine Exposure Combined with HIV-1 Disease or gp120 Expression: Comparison of Learning and Executive Functions in Humans and Mice, Neuropsychopharmacology 40 (2015) 1899–1909. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [177].Browning J, Horner JW, Pettoello-Mantovani M, Raker C, Yurasov S, DePinho RA, Goldstein H, Mice transgenic for human CD4 and CCR5 are susceptible to HIV infection, Proc Natl Acad Sci U S A 94 (1997) 14637–14641. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [178].Hanna Z, Kay DG, Rebai N, Guimond A, Jothy S, Jolicoeur P, Nef harbors a major determinant of pathogenicity for an AIDS-like disease induced by HIV-1 in transgenic mice, Cell 95 (1998) 163–175. [DOI] [PubMed] [Google Scholar]
  • [179].Leonard JM, Abramczuk JW, Pezen DS, Rutledge R, Belcher JH, Hakim F, Shearer G, Lamperth L, Travis W, Fredrickson T, et al. , Development of disease and virus recovery in transgenic mice containing HIV proviral DNA, Science 242 (1988) 1665–1670. [DOI] [PubMed] [Google Scholar]
  • [180].Seay K, Qi X, Zheng JH, Zhang C, Chen K, Dutta M, Deneroff K, Ochsenbauer C, Kappes JC, Littman DR, Goldstein H, Mice transgenic for CD4-specific human CD4, CCR5 and cyclin T1 expression: a new model for investigating HIV-1 transmission and treatment efficacy, PLoS One 8 (2013) e63537. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [181].Evering TH, Tsuji M, Human Immune System Mice for the Study of Human Immunodeficiency Virus-Type 1 Infection of the Central Nervous System, Front Immunol 9 (2018) 649. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [182].Persidsky Y, Buttini M, Limoges J, Bock P, Gendelman HE, An analysis of HIV-1-associated inflammatory products in brain tissue of humans and SCID mice with HIV-1 encephalitis, J Neurovirol 3 (1997) 401–416. [DOI] [PubMed] [Google Scholar]
  • [183].Persidsky Y, Gendelman HE, Development of laboratory and animal model systems for HIV-1 encephalitis and its associated dementia, J Leukoc Biol 62 (1997) 100–106. [DOI] [PubMed] [Google Scholar]
  • [184].Persidsky Y, Ghorpade A, Rasmussen J, Limoges J, Liu XJ, Stins M, Fiala M, Way D, Kim KS, Witte MH, Weinand M, Carhart L, Gendelman HE, Microglial and astrocyte chemokines regulate monocyte migration through the blood-brain barrier in human immunodeficiency virus-1 encephalitis, Am J Pathol 155 (1999) 1599–1611. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [185].Cook JE, Dasgupta S, Middaugh LD, Terry EC, Gorry PR, Wesselingh SL, Tyor WR, Highly active antiretroviral therapy and human immunodeficiency virus encephalitis, Ann Neurol 57 (2005) 795–803. [DOI] [PubMed] [Google Scholar]
  • [186].Cook-Easterwood J, Middaugh LD, Griffin WC 3rd, Khan I, Tyor WR, Highly active antiretroviral therapy of cognitive dysfunction and neuronal abnormalities in SCID mice with HIV encephalitis, Exp Neurol 205 (2007) 506–512. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [187].Koneru R, Olive MF, Tyor WR, Combined antiretroviral therapy reduces brain viral load and pathological features of HIV encephalitis in a mouse model, J Neurovirol 20 (2014) 9–17. [DOI] [PubMed] [Google Scholar]
  • [188].Gavegnano C, Haile WB, Hurwitz S, Tao S, Jiang Y, Schinazi RF, Tyor WR, Baricitinib reverses HIV-associated neurocognitive disorders in a SCID mouse model and reservoir seeding in vitro, J Neuroinflammation 16 (2019) 182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [189].Eggert D, Dash PK, Gorantla S, Dou H, Schifitto G, Maggirwar SB, Dewhurst S, Poluektova L, Gelbard HA, Gendelman HE, Neuroprotective activities of CEP-1347 in models of neuroAIDS, J Immunol 184 (2010) 746–756. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [190].Fritz-French C, Shawahna R, Ward JE, Maroun LE, Tyor WR, The recombinant vaccinia virus gene product, B18R, neutralizes interferon alpha and alleviates histopathological complications in an HIV encephalitis mouse model, J Interferon Cytokine Res 34 (2014) 510–517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [191].Tyor WR, Hwang HY, Fritz-French C, Morphine exposure during HIV encephalitis in SCID mice, Neurochem Res 37 (2012) 2836–2841. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [192].Ramirez SH, Potula R, Fan S, Eidem T, Papugani A, Reichenbach N, Dykstra H, Weksler BB, Romero IA, Couraud PO, Persidsky Y, Methamphetamine disrupts blood-brain barrier function by induction of oxidative stress in brain endothelial cells, J Cereb Blood Flow Metab 29 (2009) 1933–1945. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [193].Ray LA, Bujarski S, Courtney KE, Moallem NR, Lunny K, Roche D, Leventhal AM, Shoptaw S, Heinzerling K, London ED, Miotto K, The Effects of Naltrexone on Subjective Response to Methamphetamine in a Clinical Sample: a Double-Blind, Placebo-Controlled Laboratory Study, Neuropsychopharmacology 40 (2015) 2347–2356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [194].Griffin WC 3rd, Middaugh LD, Tyor WR, Chronic cocaine exposure in the SCID mouse model of HIV encephalitis, Brain Res 1134 (2007) 214–219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [195].Davies TF, Kimura H, Fong P, Kendler D, Shultz LD, Thung S, Martin A, The SCID-hu mouse and thyroid autoimmunity: characterization of human thyroid autoantibody secretion, Clin Immunol Immunopathol 60 (1991) 319–330. [DOI] [PubMed] [Google Scholar]
  • [196].Vladutiu AO, The severe combined immunodeficient (SCID) mouse as a model for the study of autoimmune diseases, Clin Exp Immunol 93 (1993) 1–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [197].Honeycutt JB, Garcia JV, Humanized mice: models for evaluating NeuroHIV and cure strategies, J Neurovirol 24 (2018) 185–191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [198].Cai S, Wang H, Bailey B, Hartwell JR, Silver JM, Juliar BE, Sinn AL, Baluyut AR, Pollok KE, Differential Secondary Reconstitution of In Vivo-Selected Human SCID-Repopulating Cells in NOD/SCID versus NOD/SCID/gamma chain Mice, Bone Marrow Res 2011 (2011) 252953. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [199].Choi B, Chun E, Kim M, Kim SY, Kim ST, Yoon K, Lee KY, Kim SJ, Human T cell development in the liver of humanized NOD/SCID/IL-2Rgamma(null)(NSG) mice generated by intrahepatic injection of CD34(+) human (h) cord blood (CB) cells, Clin Immunol 139 (2011) 321–335. [DOI] [PubMed] [Google Scholar]
  • [200].Dash PK, Gendelman HE, Roy U, Balkundi S, Alnouti Y, Mosley RL, Gelbard HA, McMillan J, Gorantla S, Poluektova LY, Long-acting nanoformulated antiretroviral therapy elicits potent antiretroviral and neuroprotective responses in HIV-1-infected humanized mice, AIDS 26 (2012) 2135–2144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [201].Boska MD, Dash PK, Knibbe J, Epstein AA, Akhter SP, Fields N, High R, Makarov E, Bonasera S, Gelbard HA, Poluektova LY, Gendelman HE, Gorantla S, Associations between brain microstructures, metabolites, and cognitive deficits during chronic HIV-1 infection of humanized mice, Mol Neurodegener 9 (2014) 58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [202].Wu X, Guo J, Niu M, An M, Liu L, Wang H, Jin X, Zhang Q, Lam KS, Wu T, Wang H, Wang Q, Du Y, Li J, Cheng L, Tang HY, Shang H, Zhang L, Zhou P, Chen Z, Tandem bispecific neutralizing antibody eliminates HIV-1 infection in humanized mice, J Clin Invest 128 (2018) 2239–2251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [203].Kumar P, Ban HS, Kim SS, Wu H, Pearson T, Greiner DL, Laouar A, Yao J, Haridas V, Habiro K, Yang YG, Jeong JH, Lee KY, Kim YH, Kim SW, Peipp M, Fey GH, Manjunath N, Shultz LD, Lee SK, Shankar P, T cell-specific siRNA delivery suppresses HIV-1 infection in humanized mice, Cell 134 (2008) 577–586. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [204].Singh VB, Singh MV, Piekna-Przybylska D, Gorantla S, Poluektova LY, Maggirwar SB, Sonic Hedgehog mimetic prevents leukocyte infiltration into the CNS during acute HIV infection, Sci Rep 7 (2017) 9578. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [205].Embretson J, Zupancic M, Ribas JL, Burke A, Racz P, Tenner-Racz K, Haase AT, Massive covert infection of helper T lymphocytes and macrophages by HIV during the incubation period of AIDS, Nature 362 (1993) 359–362. [DOI] [PubMed] [Google Scholar]
  • [206].Gendelman HE, Orenstein JM, Martin MA, Ferrua C, Mitra R, Phipps T, Wahl LA, Lane HC, Fauci AS, Burke DS, et al. , Efficient isolation and propagation of human immunodeficiency virus on recombinant colony-stimulating factor 1-treated monocytes, J Exp Med 167 (1988) 1428–1441. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [207].Ho DD, Rota TR, Hirsch MS, Infection of monocyte/macrophages by human T lymphotropic virus type III, J Clin Invest 77 (1986) 1712–1715. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [208].Honeycutt JB, Thayer WO, Baker CE, Ribeiro RM, Lada SM, Cao Y, Cleary RA, Hudgens MG, Richman DD, Garcia JV, HIV persistence in tissue macrophages of humanized myeloid-only mice during antiretroviral therapy, Nat Med 23 (2017) 638–643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [209].Baxter AE, Russell RA, Duncan CJ, Moore MD, Willberg CB, Pablos JL, Finzi A, Kaufmann DE, Ochsenbauer C, Kappes JC, Groot F, Sattentau QJ, Macrophage infection via selective capture of HIV-1-infected Cd4+ T cells, 16 (2014) 711–721. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [210].Calantone N, Wu F, Klase Z, Deleage C, Perkins M, Matsuda K, Thompson EA, Ortiz AM, Vinton CL, Ourmanov I, Lore K, Douek DC, Estes JD, Hirsch VM, Brenchley JM, Tissue myeloid cells in SIV-infected primates acquire viral DNA through phagocytosis of infected T cells, Immunity 41 (2014) 493–502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [211].Honeycutt JB, Wahl A, Archin N, Choudhary S, Margolis D, Garcia JV, HIV-1 infection, response to treatment and establishment of viral latency in a novel humanized T cell-only mouse (TOM) model, Retrovirology 10 (2013) 121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [212].Honeycutt JB, Wahl A, Baker C, Spagnuolo RA, Foster J, Zakharova O, Wietgrefe S, Caro-Vegas A, Madden V, Sharpe G, Haase AT, Eron JJ, Garcia JV, Macrophages sustain HIV replication in vivo independently of T cells, J Clin Invest 126 (2016) 1353–1366. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [213].Arainga M, Edagwa B, Mosley RL, Poluektova LY, Gorantla S, Gendelman HE, A mature macrophage is a principal HIV-1 cellular reservoir in humanized mice after treatment with long acting antiretroviral therapy, Retrovirology 14 (2017) 17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [214].Dash PK, Gorantla S, Gendelman HE, Knibbe J, Casale GP, Makarov E, Epstein AA, Gelbard HA, Boska MD, Poluektova LY, Loss of neuronal integrity during progressive HIV-1 infection of humanized mice, J Neurosci 31 (2011) 3148–3157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [215].Llewellyn GN, Alvarez-Carbonell D, Chateau M, Karn J, Cannon PM, HIV-1 infection of microglial cells in a reconstituted humanized mouse model and identification of compounds that selectively reverse HIV latency, J Neurovirol 24 (2018) 192–203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [216].Mathews S, Branch Woods A, Katano I, Makarov E, Thomas MB, Gendelman HE, Poluektova LY, Ito M, Gorantla S, Human Interleukin-34 facilitates microglia-like cell differentiation and persistent HIV-1 infection in humanized mice, Mol Neurodegener 14 (2019) 12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [217].Hasselmann J, Coburn MA, England W, Figueroa Velez DX, Kiani Shabestari S, Tu CH, McQuade A, Kolahdouzan M, Echeverria K, Claes C, Nakayama T, Azevedo R, Coufal NG, Han CZ, Cummings BJ, Davtyan H, Glass CK, Healy LM, Gandhi SP, Spitale RC, Blurton-Jones M, Development of a Chimeric Model to Study and Manipulate Human Microglia In Vivo, Neuron 103 (2019) 1016–1033 e1010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [218].Xu R, Li X, Boreland AJ, Posyton A, Kwan K, Hart RP, Jiang P, Human iPSC-derived mature microglia retain their identity and functionally integrate in the chimeric mouse brain, Nat Commun 11 (2020) 1577. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [219].Hagemeyer N, Hanft KM, Akriditou MA, Unger N, Park ES, Stanley ER, Staszewski O, Dimou L, Prinz M, Microglia contribute to normal myelinogenesis and to oligodendrocyte progenitor maintenance during adulthood, Acta Neuropathol 134 (2017) 441–458. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [220].Ray PE, Liu XH, Robinson LR, Reid W, Xu L, Owens JW, Jones OD, Denaro F, Davis HG, Bryant JL, A novel HIV-1 transgenic rat model of childhood HIV-1-associated nephropathy, Kidney Int 63 (2003) 2242–2253. [DOI] [PubMed] [Google Scholar]
  • [221].Royal W 3rd, Zhang L, Guo M, Jones O, Davis H, Bryant JL, Immune activation, viral gene product expression and neurotoxicity in the HIV-1 transgenic rat, J Neuroimmunol 247 (2012) 16–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [222].Reid WC, Ibrahim WG, Kim SJ, Denaro F, Casas R, Lee DE, Maric D, Hammoud DA, Characterization of neuropathology in the HIV-1 transgenic rat at different ages, J Neuroimmunol 292 (2016) 116–125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [223].de Guglielmo G, Fu Y, Chen J, Larrosa E, Hoang I, Kawamura T, Lorrai I, Zorman B, Bryant J, George O, Sumazin P, Lefebvre C, Repunte-Canonigo V, Sanna PP, Increases in compulsivity, inflammation, and neural injury in HIV transgenic rats with escalated methamphetamine self-administration under extended-access conditions, Brain Res 1726 (2020) 146502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [224].Liu X, Chang L, Vigorito M, Kass M, Li H, Chang SL, Methamphetamine-induced behavioral sensitization is enhanced in the HIV-1 transgenic rat, J Neuroimmune Pharmacol 4 (2009) 309–316. [DOI] [PubMed] [Google Scholar]
  • [225].Homji NF, Vigorito M, Chang SL, Morphine-induced conditioned place preference and associated behavioural plasticity in HIV-1 transgenic rats, Int J Clin Exp Med 5 (2012) 105–123. [PMC free article] [PubMed] [Google Scholar]
  • [226].Sultana S, Li H, Puche A, Jones O, Bryant JL, Royal W, Quantitation of parvalbumin+ neurons and human immunodeficiency virus type 1 (HIV-1) regulatory gene expression in the HIV-1 transgenic rat: effects of vitamin A deficiency and morphine, J Neurovirol 16 (2010) 33–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [227].Bertrand SJ, Mactutus CF, Harrod SB, Moran LM, Booze RM, HIV-1 proteins dysregulate motivational processes and dopamine circuitry, Sci Rep 8 (2018) 7869. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [228].Vigorito M, Connaghan KP, Chang SL, The HIV-1 transgenic rat model of neuroHIV, Brain Behav Immun 48 (2015) 336–349. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [229].Philippon V, Vellutini C, Gambarelli D, Harkiss G, Arbuthnott G, Metzger D, Roubin R, Filippi P, The basic domain of the lentiviral Tat protein is responsible for damages in mouse brain: involvement of cytokines, Virology 205 (1994) 519–529. [DOI] [PubMed] [Google Scholar]
  • [230].Bansal AK, Mactutus CF, Nath A, Maragos W, Hauser KF, Booze RM, Neurotoxicity of HIV-1 proteins gp120 and Tat in the rat striatum, Brain Res 879 (2000) 42–49. [DOI] [PubMed] [Google Scholar]
  • [231].Theodore S, Cass WA, Maragos WF, Involvement of cytokines in human immunodeficiency virus-1 protein Tat and methamphetamine interactions in the striatum, Exp Neurol 199 (2006) 490–498. [DOI] [PubMed] [Google Scholar]
  • [232].Theodore S, Cass WA, Maragos WF, Methamphetamine and human immunodeficiency virus protein Tat synergize to destroy dopaminergic terminals in the rat striatum, Neuroscience 137 (2006) 925–935. [DOI] [PubMed] [Google Scholar]
  • [233].Chahroudi A, Bosinger SE, Vanderford TH, Paiardini M, Silvestri G, Natural SIV hosts: showing AIDS the door, Science 335 (2012) 1188–1193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [234].Hirsch VM, Olmsted RA, Murphey-Corb M, Purcell RH, Johnson PR, An African primate lentivirus (SIVsm) closely related to HIV-2, Nature 339 (1989) 389–392. [DOI] [PubMed] [Google Scholar]
  • [235].Daniel MD, King NW, Letvin NL, Hunt RD, Sehgal PK, Desrosiers RC, A new type D retrovirus isolated from macaques with an immunodeficiency syndrome, Science 223 (1984) 602–605. [DOI] [PubMed] [Google Scholar]
  • [236].Letvin NL, Eaton KA, Aldrich WR, Sehgal PK, Blake BJ, Schlossman SF, King NW, Hunt RD, Acquired immunodeficiency syndrome in a colony of macaque monkeys, Proc Natl Acad Sci U S A 80 (1983) 2718–2722. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [237].Hunt RD, Blake BJ, Chalifoux LV, Sehgal PK, King NW, Letvin NL, Transmission of naturally occurring lymphoma in macaque monkeys, Proc Natl Acad Sci U S A 80 (1983) 5085–5089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [238].King NW, Hunt RD, Letvin NL, Histopathologic changes in macaques with an acquired immunodeficiency syndrome (AIDS), Am J Pathol 113 (1983) 382–388. [PMC free article] [PubMed] [Google Scholar]
  • [239].Vaidya NK, Ribeiro RM, Perelson AS, Kumar A, Modeling the Effects of Morphine on Simian Immunodeficiency Virus Dynamics, PLoS Comput Biol 12 (2016) e1005127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [240].Hatziioannou T, Evans DT, Animal models for HIV/AIDS research, Nat Rev Microbiol 10 (2012) 852–867. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [241].Veazey RS, DeMaria M, Chalifoux LV, Shvetz DE, Pauley DR, Knight HL, Rosenzweig M, Johnson RP, Desrosiers RC, Lackner AA, Gastrointestinal tract as a major site of CD4+ T cell depletion and viral replication in SIV infection, Science 280 (1998) 427–431. [DOI] [PubMed] [Google Scholar]
  • [242].Burudi EM, Fox HS, Simian immunodeficiency virus model of HIV-induced central nervous system dysfunction, Adv Virus Res 56 (2001) 435–468. [DOI] [PubMed] [Google Scholar]
  • [243].Lackner AA, Vogel P, Ramos RA, Kluge JD, Marthas M, Early events in tissues during infection with pathogenic (SIVmac239) and nonpathogenic (SIVmac1A11) molecular clones of simian immunodeficiency virus, Am J Pathol 145 (1994) 428–439. [PMC free article] [PubMed] [Google Scholar]
  • [244].Luciw PA, Shaw KE, Unger RE, Planelles V, Stout MW, Lackner JE, Pratt-Lowe E, Leung NJ, Banapour B, Marthas ML, Genetic and biological comparisons of pathogenic and nonpathogenic molecular clones of simian immunodeficiency virus (SIVmac), AIDS Res Hum Retroviruses 8 (1992) 395–402. [DOI] [PubMed] [Google Scholar]
  • [245].Stremlau M, Owens CM, Perron MJ, Kiessling M, Autissier P, Sodroski J, The cytoplasmic body component TRIM5alpha restricts HIV-1 infection in Old World monkeys, Nature 427 (2004) 848–853. [DOI] [PubMed] [Google Scholar]
  • [246].Newman RM, Hall L, Connole M, Chen GL, Sato S, Yuste E, Diehl W, Hunter E, Kaur A, Miller GM, Johnson WE, Balancing selection and the evolution of functional polymorphism in Old World monkey TRIM5alpha, Proc Natl Acad Sci U S A 103 (2006) 19134–19139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [247].Kirmaier A, Wu F, Newman RM, Hall LR, Morgan JS, O’Connor S, Marx PA, Meythaler M, Goldstein S, Buckler-White A, Kaur A, Hirsch VM, Johnson WE, TRIM5 suppresses cross-species transmission of a primate immunodeficiency virus and selects for emergence of resistant variants in the new species, PLoS Biol 8 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [248].Marthas ML, Lu D, Penedo MC, Hendrickx AG, Miller CJ, Titration of an SIVmac251 stock by vaginal inoculation of Indian and Chinese origin rhesus macaques: transmission efficiency, viral loads, and antibody responses, AIDS Res Hum Retroviruses 17 (2001) 1455–1466. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [249].Ling B, Veazey RS, Luckay A, Penedo C, Xu K, Lifson JD, Marx PA, SIV(mac) pathogenesis in rhesus macaques of Chinese and Indian origin compared with primary HIV infections in humans, AIDS 16 (2002) 1489–1496. [DOI] [PubMed] [Google Scholar]
  • [250].Reimann KA, Parker RA, Seaman MS, Beaudry K, Beddall M, Peterson L, Williams KC, Veazey RS, Montefiori DC, Mascola JR, Nabel GJ, Letvin NL, Pathogenicity of simian-human immunodeficiency virus SHIV-89.6P and SIVmac is attenuated in cynomolgus macaques and associated with early T-lymphocyte responses, J Virol 79 (2005) 8878–8885. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [251].Yamamoto H, Kawada M, Takeda A, Igarashi H, Matano T, Post-infection immunodeficiency virus control by neutralizing antibodies, PLoS One 2 (2007) e540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [252].Karl JA, Wiseman RW, Campbell KJ, Blasky AJ, Hughes AL, Ferguson B, Read DS, O’Connor DH, Identification of MHC class I sequences in Chinese-origin rhesus macaques, Immunogenetics 60 (2008) 37–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [253].Naruse TK, Chen Z, Yanagida R, Yamashita T, Saito Y, Mori K, Akari H, Yasutomi Y, Miyazawa M, Matano T, Kimura A, Diversity of MHC class I genes in Burmese-origin rhesus macaques, Immunogenetics 62 (2010) 601–611. [DOI] [PubMed] [Google Scholar]
  • [254].Sandler NG, Bosinger SE, Estes JD, Zhu RT, Tharp GK, Boritz E, Levin D, Wijeyesinghe S, Makamdop KN, del Prete GQ, Hill BJ, Timmer JK, Reiss E, Yarden G, Darko S, Contijoch E, Todd JP, Silvestri G, Nason M, Norgren RB Jr., Keele BF, Rao S, Langer JA, Lifson JD, Schreiber G, Douek DC, Type I interferon responses in rhesus macaques prevent SIV infection and slow disease progression, Nature 511 (2014) 601–605. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [255].Hong JJ, Amancha PK, Rogers KA, Courtney CL, Havenar-Daughton C, Crotty S, Ansari AA, Villinger F, Early lymphoid responses and germinal center formation correlate with lower viral load set points and better prognosis of simian immunodeficiency virus infection, J Immunol 193 (2014) 797–806. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [256].Wu F, Kirmaier A, Goeken R, Ourmanov I, Hall L, Morgan JS, Matsuda K, Buckler-White A, Tomioka K, Plishka R, Whitted S, Johnson W, Hirsch VM, TRIM5 alpha drives SIVsmm evolution in rhesus macaques, PLoS Pathog 9 (2013) e1003577. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [257].Matsuda K, Dang Q, Brown CR, Keele BF, Wu F, Ourmanov I, Goeken R, Whitted S, Riddick NE, Buckler-White A, Hirsch VM, Characterization of simian immunodeficiency virus (SIV) that induces SIV encephalitis in rhesus macaques with high frequency: role of TRIM5 and major histocompatibility complex genotypes and early entry to the brain, J Virol 88 (2014) 13201–13211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [258].Klatt NR, Canary LA, Vanderford TH, Vinton CL, Engram JC, Dunham RM, Cronise HE, Swerczek JM, Lafont BA, Picker LJ, Silvestri G, Brenchley JM, Dynamics of simian immunodeficiency virus SIVmac239 infection in pigtail macaques, J Virol 86 (2012) 1203–1213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [259].Batten CJ, De Rose R, Wilson KM, Agy MB, Chea S, Stratov I, Montefiori DC, Kent SJ, Comparative evaluation of simian, simian-human, and human immunodeficiency virus infections in the pigtail macaque (Macaca nemestrina) model, AIDS Res Hum Retroviruses 22 (2006) 580–588. [DOI] [PubMed] [Google Scholar]
  • [260].Fernandez CS, Reece JC, Saepuloh U, De Rose R, Ishkandriati D, O’Connor DH, Wiseman RW, Kent SJ, Screening and confirmatory testing of MHC class I alleles in pig-tailed macaques, Immunogenetics 63 (2011) 511–521. [DOI] [PubMed] [Google Scholar]
  • [261].Smith MZ, Dale CJ, De Rose R, Stratov I, Fernandez CS, Brooks AG, Weinfurter J, Krebs K, Riek C, Watkins DI, O’Connor D H, Kent SJ, Analysis of pigtail macaque major histocompatibility complex class I molecules presenting immunodominant simian immunodeficiency virus epitopes, J Virol 79 (2005) 684–695. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [262].Liao CH, Kuang YQ, Liu HL, Zheng YT, Su B, A novel fusion gene, TRIM5-Cyclophilin A in the pig-tailed macaque determines its susceptibility to HIV-1 infection, AIDS 21 Suppl 8 (2007) S19–26. [DOI] [PubMed] [Google Scholar]
  • [263].Wilson SJ, Webb BL, Ylinen LM, Verschoor E, Heeney JL, Towers GJ, Independent evolution of an antiviral TRIMCyp in rhesus macaques, Proc Natl Acad Sci U S A 105 (2008) 3557–3562. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [264].Virgen CA, Kratovac Z, Bieniasz PD, Hatziioannou T, Independent genesis of chimeric TRIM5-cyclophilin proteins in two primate species, Proc Natl Acad Sci U S A 105 (2008) 3563–3568. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [265].Newman RM, Hall L, Kirmaier A, Pozzi LA, Pery E, Farzan M, O’Neil SP, Johnson W, Evolution of a TRIM5-CypA splice isoform in old world monkeys, PLoS Pathog 4 (2008) e1000003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [266].Brennan G, Kozyrev Y, Hu SL, TRIMCyp expression in Old World primates Macaca nemestrina and Macaca fascicularis, Proc Natl Acad Sci U S A 105 (2008) 3569–3574. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [267].Zink MC, Amedee AM, Mankowski JL, Craig L, Didier P, Carter DL, Munoz A, Murphey-Corb M, Clements JE, Pathogenesis of SIV encephalitis. Selection and replication of neurovirulent SIV, Am J Pathol 151 (1997) 793–803. [PMC free article] [PubMed] [Google Scholar]
  • [268].O’Neil SP, Suwyn C, Anderson DC, Niedziela G, Bradley J, Novembre FJ, Herndon JG, McClure HM, Correlation of acute humoral response with brain virus burden and survival time in pig-tailed macaques infected with the neurovirulent simian immunodeficiency virus SIVsmmFGb, Am J Pathol 164 (2004) 1157–1172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [269].Dietrich EA, Brennan G, Ferguson B, Wiseman RW, O’Connor D, Hu SL, Variable prevalence and functional diversity of the antiretroviral restriction factor TRIMCyp in Macaca fascicularis, J Virol 85 (2011) 9956–9963. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [270].Krebs KC, Jin Z, Rudersdorf R, Hughes AL, O’Connor DH, Unusually high frequency MHC class I alleles in Mauritian origin cynomolgus macaques, J Immunol 175 (2005) 5230–5239. [DOI] [PubMed] [Google Scholar]
  • [271].Wiseman RW, Wojcechowskyj JA, Greene JM, Blasky AJ, Gopon T, Soma T, Friedrich TC, O’Connor SL, O’Connor DH, Simian immunodeficiency virus SIVmac239 infection of major histocompatibility complex-identical cynomolgus macaques from Mauritius, J Virol 81 (2007) 349–361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [272].Budde ML, Wiseman RW, Karl JA, Hanczaruk B, Simen BB, O’Connor DH, Characterization of Mauritian cynomolgus macaque major histocompatibility complex class I haplotypes by high-resolution pyrosequencing, Immunogenetics 62 (2010) 773–780. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [273].O’Connor SL, Lhost JJ, Becker EA, Detmer AM, Johnson RC, Macnair CE, Wiseman RW, Karl JA, Greene JM, Burwitz BJ, Bimber BN, Lank SM, Tuscher JJ, Mee ET, Rose NJ, Desrosiers RC, Hughes AL, Friedrich TC, Carrington M, O’Connor DH, MHC heterozygote advantage in simian immunodeficiency virus-infected Mauritian cynomolgus macaques, Sci Transl Med 2 (2010) 22ra18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [274].Beck SE, Kelly KM, Queen SE, Adams RJ, Zink MC, Tarwater PM, Mankowski JL, Macaque species susceptibility to simian immunodeficiency virus: increased incidence of SIV central nervous system disease in pigtailed macaques versus rhesus macaques, J Neurovirol 21 (2015) 148–158. [DOI] [PubMed] [Google Scholar]
  • [275].Micci L, Alvarez X, Iriele RI, Ortiz AM, Ryan ES, McGary CS, Deleage C, McAtee BB, He T, Apetrei C, Easley K, Pahwa S, Collman RG, Derdeyn CA, Davenport MP, Estes JD, Silvestri G, Lackner AA, Paiardini M, CD4 depletion in SIV-infected macaques results in macrophage and microglia infection with rapid turnover of infected cells, PLoS Pathog 10 (2014) e1004467. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [276].Schmitz JE, Kuroda MJ, Santra S, Sasseville VG, Simon MA, Lifton MA, Racz P, Tenner-Racz K, Dalesandro M, Scallon BJ, Ghrayeb J, Forman MA, Montefiori DC, Rieber EP, Letvin NL, Reimann KA, Control of viremia in simian immunodeficiency virus infection by CD8+ lymphocytes, Science 283 (1999) 857–860. [DOI] [PubMed] [Google Scholar]
  • [277].Yen PJ, Mefford ME, Hoxie JA, Williams KC, Desrosiers RC, Gabuzda D, Identification and characterization of a macrophage-tropic SIV envelope glycoprotein variant in blood from early infection in SIVmac251-infected macaques, Virology 458-459 (2014) 53–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [278].Williams K, Westmoreland S, Greco J, Ratai E, Lentz M, Kim WK, Fuller RA, Kim JP, Autissier P, Sehgal PK, Schinazi RF, Bischofberger N, Piatak M, Lifson JD, Masliah E, Gonzalez RG, Magnetic resonance spectroscopy reveals that activated monocytes contribute to neuronal injury in SIV neuroAIDS, J Clin Invest 115 (2005) 2534–2545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [279].Burdo TH, Soulas C, Orzechowski K, Button J, Krishnan A, Sugimoto C, Alvarez X, Kuroda MJ, Williams KC, Increased monocyte turnover from bone marrow correlates with severity of SIV encephalitis and CD163 levels in plasma, PLoS Pathog 6 (2010) e1000842. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [280].Nowlin BT, Burdo TH, Midkiff CC, Salemi M, Alvarez X, Williams KC, SIV encephalitis lesions are composed of CD163(+) macrophages present in the central nervous system during early SIV infection and SIV-positive macrophages recruited terminally with AIDS, Am J Pathol 185 (2015) 1649–1665. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [281].Soulas C, Conerly C, Kim WK, Burdo TH, Alvarez X, Lackner AA, Williams KC, Recently infiltrating MAC387(+) monocytes/macrophages a third macrophage population involved in SIV and HIV encephalitic lesion formation, Am J Pathol 178 (2011) 2121–2135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [282].Burdo TH, Lackner A, Williams KC, Monocyte/macrophages and their role in HIV neuropathogenesis, Immunol Rev 254 (2013) 102–113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [283].Walker JA, Sulciner ML, Nowicki KD, Miller AD, Burdo TH, Williams KC, Elevated numbers of CD163+ macrophages in hearts of simian immunodeficiency virus-infected monkeys correlate with cardiac pathology and fibrosis, AIDS Res Hum Retroviruses 30 (2014) 685–694. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [284].Joag SV, Adams RJ, Foresman L, Galbreath D, Zink MC, Pinson DM, McClure H, Narayan O, Early activation of PBMC and appearance of antiviral CD8+ cells influence the prognosis of SIV-induced disease in rhesus macaques, J Med Primatol 23 (1994) 108–116. [DOI] [PubMed] [Google Scholar]
  • [285].Sharma DP, Anderson M, Zink MC, Adams RJ, Donnenberg AD, Clements JE, Narayan O, Pathogenesis of acute infection in rhesus macaques with a lymphocyte-tropic strain of simian immunodeficiency virus, J Infect Dis 166 (1992) 738–746. [DOI] [PubMed] [Google Scholar]
  • [286].Sharma DP, Zink MC, Anderson M, Adams R, Clements JE, Joag SV, Narayan O, Derivation of neurotropic simian immunodeficiency virus from exclusively lymphocytetropic parental virus: pathogenesis of infection in macaques, J Virol 66 (1992) 3550–3556. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [287].Lackner AA, Smith MO, Munn RJ, Martfeld DJ, Gardner MB, Marx PA, Dandekar S, Localization of simian immunodeficiency virus in the central nervous system of rhesus monkeys, Am J Pathol 139 (1991) 609–621. [PMC free article] [PubMed] [Google Scholar]
  • [288].Raghavan R, Cheney PD, Raymond LA, Joag SV, Stephens EB, Adany I, Pinson DM, Li Z, Marcario JK, Jia F, Wang C, Foresman L, Berman NE, Narayan O, Morphological correlates of neurological dysfunction in macaques infected with neurovirulent simian immunodeficiency virus, Neuropathol Appl Neurobiol 25 (1999) 285–294. [DOI] [PubMed] [Google Scholar]
  • [289].Marcario JK, Riazi M, Adany I, Kenjale H, Fleming K, Marquis J, Nemon O, Mayo MS, Yankee T, Narayan O, Cheney PD, Effect of morphine on the neuropathogenesis of SIVmac infection in Indian Rhesus Macaques, J Neuroimmune Pharmacol 3 (2008) 12–25. [DOI] [PubMed] [Google Scholar]
  • [290].Marcondes MC, Burdo TH, Sopper S, Huitron-Resendiz S, Lanigan C, Watry D, Flynn C, Zandonatti M, Fox HS, Enrichment and persistence of virus-specific CTL in the brain of simian immunodeficiency virus-infected monkeys is associated with a unique cytokine environment, J Immunol 178 (2007) 5812–5819. [DOI] [PubMed] [Google Scholar]
  • [291].Marcondes MC, Burudi EM, Huitron-Resendiz S, Sanchez-Alavez M, Watry D, Zandonatti M, Henriksen SJ, Fox HS, Highly activated CD8(+) T cells in the brain correlate with early central nervous system dysfunction in simian immunodeficiency virus infection, J Immunol 167 (2001) 5429–5438. [DOI] [PubMed] [Google Scholar]
  • [292].Marcondes MC, Phillipson CA, Fox HS, Distinct clonal repertoire of brain CD8+ cells in simian immunodeficiency virus infection, AIDS 17 (2003) 1605–1611. [DOI] [PubMed] [Google Scholar]
  • [293].Igarashi T, Imamichi H, Brown CR, Hirsch VM, Martin MA, The emergence and characterization of macrophage-tropic SIV/HIV chimeric viruses (SHIVs) present in CD4+ T cell-depleted rhesus monkeys, J Leukoc Biol 74 (2003) 772–780. [DOI] [PubMed] [Google Scholar]
  • [294].Igarashi T, Brown CR, Endo Y, Buckler-White A, Plishka R, Bischofberger N, Hirsch V, Martin MA, Macrophage are the principal reservoir and sustain high virus loads in rhesus macaques after the depletion of CD4+ T cells by a highly pathogenic simian immunodeficiency virus/HIV type 1 chimera (SHIV): Implications for HIV-1 infections of humans, Proc Natl Acad Sci U S A 98 (2001) 658–663. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [295].Zink MC, Suryanarayana K, Mankowski JL, Shen A, Piatak M Jr., Spelman JP, Carter DL, Adams RJ, Lifson JD, Clements JE, High viral load in the cerebrospinal fluid and brain correlates with severity of simian immunodeficiency virus encephalitis, J Virol 73 (1999) 10480–10488. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [296].Mankowski JL, Clements JE, Zink MC, Searching for clues: tracking the pathogenesis of human immunodeficiency virus central nervous system disease by use of an accelerated, consistent simian immunodeficiency virus macaque model, J Infect Dis 186 Suppl 2 (2002) S199–208. [DOI] [PubMed] [Google Scholar]
  • [297].Kelly KM, Tocchetti CG, Lyashkov A, Tarwater PM, Bedja D, Graham DR, Beck SE, Metcalf Pate KA, Queen SE, Adams RJ, Paolocci N, Mankowski JL, CCR5 inhibition prevents cardiac dysfunction in the SIV/macaque model of HIV, J Am Heart Assoc 3 (2014) e000874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [298].Laast VA, Shim B, Johanek LM, Dorsey JL, Hauer PE, Tarwater PM, Adams RJ, Pardo CA, McArthur JC, Ringkamp M, Mankowski JL, Macrophage-mediated dorsal root ganglion damage precedes altered nerve conduction in SIV-infected macaques, Am J Pathol 179 (2011) 2337–2345. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [299].Dorsey JL, Mangus LM, Hauer P, Ebenezer GJ, Queen SE, Laast VA, Adams RJ, Mankowski JL, Persistent Peripheral Nervous System Damage in Simian Immunodeficiency Virus-Infected Macaques Receiving Antiretroviral Therapy, J Neuropathol Exp Neurol 74 (2015) 1053–1060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [300].Bokhari SM, Hegde R, Callen S, Yao H, Adany I, Li Q, Li Z, Pinson D, Yeh HW, Cheney PD, Buch S, Morphine potentiates neuropathogenesis of SIV infection in rhesus macaques, J Neuroimmune Pharmacol 6 (2011) 626–639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [301].Chivero ET, Guo ML, Periyasamy P, Liao K, Callen SE, Buch S, HIV-1 Tat Primes and Activates Microglial NLRP3 Inflammasome-Mediated Neuroinflammation, J Neurosci 37 (2017) 3599–3609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [302].Periyasamy P, Thangaraj A, Guo ML, Hu G, Callen S, Buch S, Epigenetic Promoter DNA Methylation of miR-124 Promotes HIV-1 Tat-Mediated Microglial Activation via MECP2-STAT3 Axis, J Neurosci 38 (2018) 5367–5383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [303].Periyasamy P, Thangaraj A, Bendi VS, Buch S, HIV-1 Tat-mediated microglial inflammation involves a novel miRNA-34a-NLRC5-NFkappaB signaling axis, Brain Behav Immun 80 (2019) 227–237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [304].Sil S, Hu G, Liao K, Niu F, Callen S, Periyasamy P, Fox HS, Buch S, HIV-1 Tat-mediated astrocytic amyloidosis involves the HIF-1alpha/lncRNA BACE1-AS axis, PLoS Biol 18 (2020) e3000660. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [305].Marcario JK, Raymond LA, McKiernan BJ, Foresman LL, Joag SV, Raghavan R, Narayan O, Cheney PD, Motor skill impairment in SIV-infected rhesus macaques with rapidly and slowly progressing disease, J Med Primatol 28 (1999) 105–117. [DOI] [PubMed] [Google Scholar]
  • [306].Williams K, Burdo TH, Monocyte mobilization, activation markers, and unique macrophage populations in the brain: observations from SIV infected monkeys are informative with regard to pathogenic mechanisms of HIV infection in humans, J Neuroimmune Pharmacol 7 (2012) 363–371. [DOI] [PubMed] [Google Scholar]
  • [307].Schmitz JE, Simon MA, Kuroda MJ, Lifton MA, Ollert MW, Vogel CW, Racz P, Tenner-Racz K, Scallon BJ, Dalesandro M, Ghrayeb J, Rieber EP, Sasseville VG, Reimann KA, A nonhuman primate model for the selective elimination of CD8+ lymphocytes using a mouse-human chimeric monoclonal antibody, Am J Pathol 154 (1999) 1923–1932. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [308].Clements JE, Babas T, Mankowski JL, Suryanarayana K, Piatak M Jr., Tarwater PM, Lifson JD, Zink MC, The central nervous system as a reservoir for simian immunodeficiency virus (SIV): steady-state levels of SIV DNA in brain from acute through asymptomatic infection, J Infect Dis 186 (2002) 905–913. [DOI] [PubMed] [Google Scholar]
  • [309].Daniel MD, Letvin NL, King NW, Kannagi M, Sehgal PK, Hunt RD, Kanki PJ, Essex M, Desrosiers RC, Isolation of T-cell tropic HTLV-III-like retrovirus from macaques, Science 228 (1985) 1201–1204. [DOI] [PubMed] [Google Scholar]
  • [310].Mansfield KG, Lerch NW, Gardner MB, Lackner AA, Origins of simian immunodeficiency virus infection in macaques at the New England Regional Primate Research Center, J Med Primatol 24 (1995) 116–122. [DOI] [PubMed] [Google Scholar]
  • [311].Keele BF, Li H, Learn GH, Hraber P, Giorgi EE, Grayson T, Sun C, Chen Y, Yeh WW, Letvin NL, Mascola JR, Nabel GJ, Haynes BF, Bhattacharya T, Perelson AS, Korber BT, Hahn BH, Shaw GM, Low-dose rectal inoculation of rhesus macaques by SIVsmE660 or SIVmac251 recapitulates human mucosal infection by HIV-1, J Exp Med 206 (2009) 1117–1134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [312].Kestler H, Kodama T, Ringler D, Marthas M, Pedersen N, Lackner A, Regier D, Sehgal P, Daniel M, King N, et al. , Induction of AIDS in rhesus monkeys by molecularly cloned simian immunodeficiency virus, Science 248 (1990) 1109–1112. [DOI] [PubMed] [Google Scholar]
  • [313].Hirsch V, Adger-Johnson D, Campbell B, Goldstein S, Brown C, Elkins WR, Montefiori DC, A molecularly cloned, pathogenic, neutralization-resistant simian immunodeficiency virus, SIVsmE543–3, J Virol 71 (1997) 1608–1620. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [314].Reynolds MR, Sacha JB, Weiler AM, Borchardt GJ, Glidden CE, Sheppard NC, Norante FA, Castrovinci PA, Harris JJ, Robertson HT, Friedrich TC, McDermott AB, Wilson NA, Allison DB, Koff WC, Johnson WE, Watkins DI, The TRIM5{alpha} genotype of rhesus macaques affects acquisition of simian immunodeficiency virus SIVsmE660 infection after repeated limiting-dose intrarectal challenge, J Virol 85 (2011) 9637–9640. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [315].Mutua JM, Perelson AS, Kumar A, Vaidya NK, Modeling the Effects of Morphine-Altered Virus Specific Antibody Responses on HIV/SIV Dynamics, Sci Rep 9 (2019) 5423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [316].Suzuki S, Chuang AJ, Chuang LF, Doi RH, Chuang RY, Morphine promotes simian acquired immunodeficiency syndrome virus replication in monkey peripheral mononuclear cells: induction of CC chemokine receptor 5 expression for virus entry, J Infect Dis 185 (2002) 1826–1829. [DOI] [PubMed] [Google Scholar]
  • [317].Li PF, Hao YS, Huang DA, Liu XH, Liu SL, Li G, Morphine-promoted survival of CEMx174 cells in early stages of SIV infection in vitro: involvement of the multiple molecular mechanisms, Toxicol In Vitro 18 (2004) 449–456. [DOI] [PubMed] [Google Scholar]
  • [318].Marcario JK, Pendyala G, Riazi M, Fleming K, Marquis J, Callen S, Lisco SJ, Fowler SC, Cheney PD, Buch SJ, Effects of Morphine on Behavioral Task Performance in SIV-Infected Rhesus Macaques, J Neuroimmune Pharmacol 11 (2016) 348–357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [319].Sindberg GM, Callen SE, Banerjee S, Meng J, Hale VL, Hegde R, Cheney PD, Villinger F, Roy S, Buch S, Morphine Potentiates Dysbiotic Microbial and Metabolic Shifts in Acute SIV Infection, J Neuroimmune Pharmacol 14 (2019) 200–214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [320].Hu G, Yao H, Chaudhuri AD, Duan M, Yelamanchili SV, Wen H, Cheney PD, Fox HS, Buch S, Exosome-mediated shuttling of microRNA-29 regulates HIV Tat and morphine-mediated neuronal dysfunction, Cell Death Dis 3 (2012) e381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [321].Donahoe RM, Byrd LD, McClure HM, Fultz P, Brantley M, Marsteller F, Ansari AA, Wenzel D, Aceto M, Consequences of opiate-dependency in a monkey model of AIDS, Adv Exp Med Biol 335 (1993) 21–28. [DOI] [PubMed] [Google Scholar]
  • [322].Kumar A, Mukherjee S, Shen J, Buch S, Li Z, Adany I, Liu Z, Zhuge W, Piatak M Jr., Lifson J, McClure H, Narayan O, Immunization of macaques with live simian human immunodeficiency virus (SHIV) vaccines conferred protection against AIDS induced by homologous and heterologous SHIVs and simian immunodeficiency virus, Virology 301 (2002) 189–205. [DOI] [PubMed] [Google Scholar]
  • [323].Kumar R, Torres C, Yamamura Y, Rodriguez I, Martinez M, Staprans S, Donahoe RM, Kraiselburd E, Stephens EB, Kumar A, Modulation by morphine of viral set point in rhesus macaques infected with simian immunodeficiency virus and simian-human immunodeficiency virus, J Virol 78 (2004) 11425–11428. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [324].Weed M, Adams RJ, Hienz RD, Meulendyke KA, Linde ME, Clements JE, Mankowski JL, Zink MC, SIV/macaque model of HIV infection in cocaine users: minimal effects of cocaine on behavior, virus replication, and CNS inflammation, J Neuroimmune Pharmacol 7 (2012) 401–411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [325].Marcondes MC, Flynn C, Watry DD, Zandonatti M, Fox HS, Methamphetamine increases brain viral load and activates natural killer cells in simian immunodeficiency virus-infected monkeys, Am J Pathol 177 (2010) 355–361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [326].Najera JA, Bustamante EA, Bortell N, Morsey B, Fox HS, Ravasi T, Marcondes MC, Methamphetamine abuse affects gene expression in brain-derived microglia of SIV-infected macaques to enhance inflammation and promote virus targets, BMC Immunol 17 (2016) 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [327].Bortell N, Morsey B, Basova L, Fox HS, Marcondes MC, Phenotypic changes in the brain of SIV-infected macaques exposed to methamphetamine parallel macrophage activation patterns induced by the common gamma-chain cytokine system, Front Microbiol 6 (2015) 900. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [328].Pedersen NC, Ho EW, Brown ML, Yamamoto JK, Isolation of a T-lymphotropic virus from domestic cats with an immunodeficiency-like syndrome, Science 235 (1987) 790–793. [DOI] [PubMed] [Google Scholar]
  • [329].Sparger EE, FIV as a Model for HIV: An Overview. In: Friedman H, Specter S, Bendinelli M (Eds.), In vivo Models of HIV Disease and Control, Springer US, Boston, MA, 2006, pp. 149–237. [Google Scholar]
  • [330].Miller C, Abdo Z, Ericsson A, Elder J, VandeWoude S, Applications of the FIV Model to Study HIV Pathogenesis, Viruses 10 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [331].Bendinelli M, Pistello M, Lombardi S, Poli A, Garzelli C, Matteucci D, Ceccherini-Nelli L, Malvaldi G, Tozzini F, Feline immunodeficiency virus: an interesting model for AIDS studies and an important cat pathogen, Clin Microbiol Rev 8 (1995) 87–112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [332].Ackley CD, Yamamoto JK, Levy N, Pedersen NC, Cooper MD, Immunologic abnormalities in pathogen-free cats experimentally infected with feline immunodeficiency virus, J Virol 64 (1990) 5652–5655. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [333].Siebelink KH, Chu IH, Rimmelzwaan GF, Weijer K, van Herwijnen R, Knell P, Egberink HF, Bosch ML, Osterhaus AD, Feline immunodeficiency virus (FIV) infection in the cat as a model for HIV infection in man: FIV-induced impairment of immune function, AIDS Res Hum Retroviruses 6 (1990) 1373–1378. [DOI] [PubMed] [Google Scholar]
  • [334].Bishop SA, Stokes CR, Gruffydd-Jones TJ, Whiting CV, Harbour DA, Vaginal and rectal infection of cats with feline immunodeficiency virus, Vet Microbiol 51 (1996) 217–227. [DOI] [PubMed] [Google Scholar]
  • [335].Obert LA, Hoover EA, Early pathogenesis of transmucosal feline immunodeficiency virus infection, J Virol 76 (2002) 6311–6322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [336].Obert LA, Hoover EA, Relationship of lymphoid lesions to disease course in mucosal feline immunodeficiency virus type C infection, Vet Pathol 37 (2000) 386–401. [DOI] [PubMed] [Google Scholar]
  • [337].Tenorio AP, Franti CE, Madewell BR, Pedersen NC, Chronic oral infections of cats and their relationship to persistent oral carriage of feline calici-, immunodeficiency, or leukemia viruses, Vet Immunol Immunopathol 29 (1991) 1–14. [DOI] [PubMed] [Google Scholar]
  • [338].Bienzle D, FIV in cats--a useful model of HIV in people?, Vet Immunol Immunopathol 159 (2014) 171–179. [DOI] [PubMed] [Google Scholar]
  • [339].Sparkes AH, Hopper CD, Millard WG, Gruffydd-Jones TJ, Harbour DA, Feline immunodeficiency virus infection. Clinicopathologic findings in 90 naturally occurring cases, J Vet Intern Med 7 (1993) 85–90. [DOI] [PubMed] [Google Scholar]
  • [340].Shelton GH, Linenberger ML, Persik MT, Abkowitz JL, Prospective hematologic and clinicopathologic study of asymptomatic cats with naturally acquired feline immunodeficiency virus infection, J Vet Intern Med 9 (1995) 133–140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [341].Yamamoto JK, Hansen H, Ho EW, Morishita TY, Okuda T, Sawa TR, Nakamura RM, Pedersen NC, Epidemiologic and clinical aspects of feline immunodeficiency virus infection in cats from the continental United States and Canada and possible mode of transmission, J Am Vet Med Assoc 194 (1989) 213–220. [PubMed] [Google Scholar]
  • [342].Ishida T, Taniguchi A, Matsumura S, Washizu T, Tomoda I, Long-term clinical observations on feline immunodeficiency virus infected asymptomatic carriers, Vet Immunol Immunopathol 35 (1992) 15–22. [DOI] [PubMed] [Google Scholar]
  • [343].Eckstrand CD, Hillman C, Smith AL, Sparger EE, Murphy BG, Viral Reservoirs in Lymph Nodes of FIV-Infected Progressor and Long-Term Non-Progressor Cats during the Asymptomatic Phase, PLoS One 11 (2016) e0146285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [344].Fletcher NF, Meeker RB, Hudson LC, Callanan JJ, The neuropathogenesis of feline immunodeficiency virus infection: barriers to overcome, Vet J 188 (2011) 260–269. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [345].Meeker RB, Hudson L, Feline Immunodeficiency Virus Neuropathogenesis: A Model for HIV-Induced CNS Inflammation and Neurodegeneration, Vet Sci 4 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [346].Dow SW, Poss ML, Hoover EA, Feline immunodeficiency virus: a neurotropic lentivirus, J Acquir Immune Defic Syndr (1988) 3 (1990) 658–668. [PubMed] [Google Scholar]
  • [347].Burdo TH, Miller AD, Animal models of HIV peripheral neuropathy, Future Virol 9 (2014) 465–474. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [348].Poli A, Abramo F, Di Iorio C, Cantile C, Carli MA, Pollera C, Vago L, Tosoni A, Costanzi G, Neuropathology in cats experimentally infected with feline immunodeficiency virus: a morphological, immunocytochemical and morphometric study, J Neurovirol 3 (1997) 361–368. [DOI] [PubMed] [Google Scholar]
  • [349].Podell M, March PA, Buck WR, Mathes LE, The feline model of neuroAIDS: understanding the progression towards AIDS dementia, J Psychopharmacol 14 (2000) 205–213. [DOI] [PubMed] [Google Scholar]
  • [350].Meeker RB, Poulton W, Feng WH, Hudson L, Longo FM, Suppression of immunodeficiency virus-associated neural damage by the p75 neurotrophin receptor ligand, LM11A-31, in an in vitro feline model, J Neuroimmune Pharmacol 7 (2012) 388–400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [351].Barr MC, Testa MP, Phillips TR, Drugs of Abuse, AIDS, and the FIV Model. In: Friedman H, Specter S, Bendinelli M (Eds.), In vivo Models of HIV Disease and Control, Springer US, Boston, MA, 2006, pp. 275–291. [Google Scholar]
  • [352].Barr MC, Billaud JN, Selway DR, Huitron-Resendiz S, Osborn KG, Henriksen SJ, Phillips TR, Effects of multiple acute morphine exposures on feline immunodeficiency virus disease progression, J Infect Dis 182 (2000) 725–732. [DOI] [PubMed] [Google Scholar]
  • [353].Barr MC, Huitron-Resendiz S, Sanchez-Alavez M, Henriksen SJ, Phillips TR, Escalating morphine exposures followed by withdrawal in feline immunodeficiency virus-infected cats: a model for HIV infection in chronic opiate abusers, Drug Alcohol Depend 72 (2003) 141–149. [DOI] [PubMed] [Google Scholar]
  • [354].Gavrilin MA, Mathes LE, Podell M, Methamphetamine enhances cell-associated feline immunodeficiency virus replication in astrocytes, J Neurovirol 8 (2002) 240–249. [DOI] [PubMed] [Google Scholar]
  • [355].Phillips TR, Billaud JN, Henriksen SJ, Methamphetamine and HIV-1: potential interactions and the use of the FIV/cat model, J Psychopharmacol 14 (2000) 244–250. [DOI] [PubMed] [Google Scholar]
  • [356].Cloak CC, Chang L, Ernst T, Barr MC, Huitron-Resendiz S, Sanchez-Alavez M, Phillips TR, Henriksen S, Methamphetamine and AIDS: 1HMRS studies in a feline model of human disease, J Neuroimmunol 147 (2004) 16–20. [DOI] [PubMed] [Google Scholar]
  • [357].Yamamoto JK, Sanou MP, Abbott JR, Coleman JK, Feline immunodeficiency virus model for designing HIV/AIdS vaccines, Curr HIV Res 8 (2010) 14–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [358].Shimojima M, Miyazawa T, Ikeda Y, McMonagle EL, Haining H, Akashi H, Takeuchi Y, Hosie MJ, Willett BJ, Use of CD134 as a primary receptor by the feline immunodeficiency virus, Science 303 (2004) 1192–1195. [DOI] [PubMed] [Google Scholar]
  • [359].Dean GA, Reubel GH, Moore PF, Pedersen NC, Proviral burden and infection kinetics of feline immunodeficiency virus in lymphocyte subsets of blood and lymph node, J Virol 70 (1996) 5165–5169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [360].Jantzie LL, Rauw GA, Todd KG, The effects of doxycycline administration on amino acid neurotransmitters in an animal model of neonatal hypoxia-ischemia, Neurochem Int 49 (2006) 717–728. [DOI] [PubMed] [Google Scholar]
  • [361].Mediouni S, Jablonski J, Paris JJ, Clementz MA, Thenin-Houssier S, McLaughlin JP, Valente ST, Didehydro-cortistatin A inhibits HIV-1 Tat mediated neuroinflammation and prevents potentiation of cocaine reward in Tat transgenic mice, Curr HIV Res 13 (2015) 64–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [362].Yao H, Duan M, Yang L, Buch S, Platelet-derived growth factor-BB restores human immunodeficiency virus Tat-cocaine-mediated impairment of neurogenesis: role of TRPC1 channels, J Neurosci 32 (2012) 9835–9847. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [363].Shultz LD, Brehm MA, Bavari S, Greiner DL, Humanized mice as a preclinical tool for infectious disease and biomedical research, Ann N Y Acad Sci 1245 (2011) 50–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [364].Woodbine L, Neal JA, Sasi NK, Shimada M, Deem K, Coleman H, Dobyns WB, Ogi T, Meek K, Davies EG, Jeggo PA, PRKDC mutations in a SCID patient with profound neurological abnormalities, J Clin Invest 123 (2013) 2969–2980. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [365].Beamer WG, Shultz KL, Tennent BJ, Shultz LD, Granulosa cell tumorigenesis in genetically hypogonadal-immunodeficient mice grafted with ovaries from tumor-susceptible donors, Cancer Res 53 (1993) 3741–3746. [PubMed] [Google Scholar]
  • [366].Ross J, Comments on the article “Persistent confusion of total entropy and chemical system entropy in chemical thermodynamics” [(1996) Proc. Natl. Acad. Sci. USA 93, 7452-7453], Proc Natl Acad Sci U S A 93 (1996) 14314; discussion 14315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [367].Zaparte A, Schuch JB, Viola TW, Baptista TAS, Beidacki AS, do Prado CH, Sanvicente-Vieira B, Bauer ME, Grassi-Oliveira R, Cocaine Use Disorder Is Associated With Changes in Th1/Th2/Th17 Cytokines and Lymphocytes Subsets, Front Immunol 10 (2019) 2435. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [368].Rey P, [Ergonomics, a help in occupational health. A Swiss opinion], Soins Psychiatr (1985) 15–18. [PubMed] [Google Scholar]
  • [369].Roberts AJ, Maung R, Sejbuk NE, Ake C, Kaul M, Alteration of Methamphetamine-induced stereotypic behaviour in transgenic mice expressing HIV-1 envelope protein gp120, J Neurosci Methods 186 (2010) 222–225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [370].Peng J, Vigorito M, Liu X, Zhou D, Wu X, Chang SL, The HIV-1 transgenic rat as a model for HIV-1 infected individuals on HAART, J Neuroimmunol 218 (2010) 94–101. [DOI] [PubMed] [Google Scholar]
  • [371].Sarkar S, Mao X, Liu C, Chang SL, Age- and ethanol concentration-dependent effects of acute binge drinking in the HIV-1 transgenic rat, Alcohol Clin Exp Res 37 Suppl 1 (2013) E70–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [372].Clary CR, Guidot DM, Bratina MA, Otis JS, Chronic alcohol ingestion exacerbates skeletal muscle myopathy in HIV-1 transgenic rats, AIDS Res Ther 8 (2011) 30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [373].McCune JM, Namikawa R, Kaneshima H, Shultz LD, Lieberman M, Weissman IL, The SCID-hu mouse: murine model for the analysis of human hematolymphoid differentiation and function, Science 241 (1988) 1632–1639. [DOI] [PubMed] [Google Scholar]
  • [374].Namikawa R, Weilbaecher KN, Kaneshima H, Yee EJ, McCune JM, Long-term human hematopoiesis in the SCID-hu mouse, J Exp Med 172 (1990) 1055–1063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [375].Honeycutt JB, Sheridan PA, Matsushima GK, Garcia JV, Humanized mouse models for HIV-1 infection of the CNS, J Neurovirol 21 (2015) 301–309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [376].Bennett ML, Bennett FC, Liddelow SA, Ajami B, Zamanian JL, Fernhoff NB, Mulinyawe SB, Bohlen CJ, Adil A, Tucker A, Weissman IL, Chang EF, Li G, Grant GA, Hayden Gephart MG, Barres BA, New tools for studying microglia in the mouse and human CNS, Proc Natl Acad Sci U S A 113 (2016) E1738–1746. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES