Next Article in Journal
Application of Biomarkers in Spinal Muscular Atrophy
Previous Article in Journal
Innovative In Situ Interfacial Co-Assembled Lignin/Chitosan Nanoparticles—Green Synthesis, Physicochemical Characterization, In Vitro Release, and Intermolecular Interactions
Previous Article in Special Issue
TSPO Ligand 2-Cl-MGV-1 Mitigates Traumatic Brain Injury (TBI) in a Mouse Model
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Modeling Virus-Associated Central Nervous System Disease in Non-Human Primates

Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA 70433, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(14), 6886; https://doi.org/10.3390/ijms26146886
Submission received: 5 June 2025 / Revised: 4 July 2025 / Accepted: 14 July 2025 / Published: 17 July 2025
(This article belongs to the Special Issue Animal Research Model for Neurological Diseases, 2nd Edition)

Abstract

While viral pathogens are often subdivided into neurotropic and non-neurotropic categories, systemic inflammation caused by non-neurotropic viruses still possesses the ability to alter the central nervous system (CNS). Studies of CNS disease induced by viral infection, whether neurotropic or not, are presented with a unique set of challenges. First, because brain biopsies are rarely necessary to diagnose viral-associated neurological disorders, antemortem tissue samples are not readily available for study and human pathological studies must rely on end-stage, postmortem evaluations. Second, in vitro models fail to fully capture the nuances of an intact immune system, necessitating the use of animal models to fully characterize pathogenesis and identify potential therapeutic approaches. Non-human primates (NHP) represent a particularly attractive animal model in that they overcome many of the limits posed by more distant species and most closely mirror human disease pathogenesis and susceptibility. Here, we review NHP infection models of viruses known to infect and/or replicate within cells of the CNS, including West Nile virus, the equine encephalitis viruses, Zika virus, and herpesviruses, as well as those known to alter the immune status of the brain in the absence of significant CNS penetrance, including human immunodeficiency virus (HIV) in the current era of combination antiretroviral therapy (cART) and the coronavirus of severe acute respiratory syndrome (SARS)-CoV−2. This review focuses on viruses with an established role in causing CNS disease, including encephalitis, meningitis, and myelitis and NHP models of viral infection that are directly translatable to the human condition through relevant routes of infection, comparable disease pathogenesis, and responses to therapeutic intervention.

1. Introduction

Both neurotropic and non-neurotropic viral pathogens are capable of inducing changes in the central nervous system (CNS). Neurotropic viruses, defined here as viruses that directly infect CNS cells, such as neurons or microglia, often incite local cytokine or chemokine production and recruitment of inflammatory cells. However, even in the absence of true neurotropism, many viral pathogens influence the CNS immune milieu through interactions with viral proteins and/or factors associated with the systemic host immune response that influences microglial, astrocyte, and neuron activity, as well as cells comprising the blood–brain barrier (BBB), such as endothelial cells. For example, systemic inflammation is associated with impaired BBB integrity and microglial activation even in the absence of viral infection [1].
There are several challenges to interrogating viral pathogen-induced CNS disease. First, because noninvasive diagnostic methods are effective, invasive brain biopsies in living patients are uncommon. As a result, human brain tissue is not generally available until after death and human studies are largely limited to clinical observations and postmortem studies. In vitro models are increasingly sophisticated and enable cell-specific studies, but do not recapitulate a complete immune system, thus requiring the use of animal models to more completely model viral pathogenesis. Mouse models are the most widely used in vivo model given their small size, economical cost to maintain, genetic manipulability, and short breeding and life span, and are instrumental in studies identifying and screening potential therapeutic candidates. Nonetheless, the translation of mouse models to human disease is limited by vast differences in genetics, life span, anatomy, physiology, and metabolic biology, such that many discoveries made in mice that are often engineered or inherently identical fail to fully recapitulate human disease. Rodents also lack important human immune receptors, have significant anatomic differences including brain structure, and fail to reproduce some clinical symptoms observed in human disease. Non-human primates (NHP), particularly Old-World species such as macaques, anatomically and physiologically closely resemble human brain structure, immune system maturity, and BBB composition. Thus, NHPs bridge a key gap in studies modeling human disease pathogenesis and susceptibility, particularly CNS disease and provide a crucial platform for evaluating how viruses penetrate the BBB and trigger neuroinflammation.
Here, we review NHP models that recapitulate relevant aspects of human disease with an emphasis on routes of infection, comparable disease pathogenesis, and response to therapeutic intervention. We focus on viruses known to cause CNS disease and neuroinflammation, including encephalitis, meningitis, myelitis, and CNS proinflammatory cytokine production (Table 1). These include directly neuropathic viruses known to infect and/or replicate within cells of the CNS, such as West Nile virus, the equine encephalitis viruses, Zika virus, and herpesviruses, as well as non-neurotrophic viral pathogens known to perturb the immune milieu of the brain in the absence of significant viral penetrance into the CNS compartment, including human immunodeficiency virus (HIV) in the context of effective pharmacological intervention and the coronavirus of severe acute respiratory syndrome (SARS)-CoV-2.

2. Flaviviruses

The genus Flavivirus of family Flaviviridae includes Zika virus (ZIKV), Dengue virus, West Nile virus (WNV), yellow fever virus, and Japanese encephalitis virus. Here, we focus on ZIKV and WNV.

2.1. Zika Virus

ZIKV is an enveloped, RNA arbovirus initially isolated from a rhesus macaque (RM, Macaca mulatta) in the 1950s [2]. Disease due to ZIKV infection in humans is most often associated with mild rash, fever, conjunctivitis, and arthritis or joint pain [3] (Supplementary Table S1). Aedes mosquitos, especially Aedes aegypti are the primary vector, but ZIKV is also transmissible sexually and vertically during pregnancy from mother to fetus [4,5]. Indeed, ZIKV’s rise in public health significance is chiefly due to links between maternal infection and fetal loss and microcephaly, as well as the development of Guillain-Barre syndrome [6]. The virus has a particular tropism for neuroprogenitor cells (Supplementary Table S1), but also infects glia, epithelial cells, and leukocytes; cellular tropism has been reviewed in [7,8]. Furthermore, ZIKV nonstructural proteins NS4A and NS4B disrupt protein kinase B (Akt)- mammalian target of rapamycin (mTOR) and Notch signaling pathways critical for proper fetal brain development, resulting in impaired glial maturation, fetal myelination, and neurogenesis [9,10].

2.2. NHP Models of Zika Virus

To date, NHP models of ZIKV have centralized around two common themes: disease pathogenesis in adult and fetal infections, and vaccine development. There is currently no effective vaccine licensed to prevent ZIKV infection, although numerous vaccine candidates have been tested (reviewed in [11]). Viremia peaks 2–5 days post ZIKV-infection and macaques develop neutralizing antibodies [12,13]. Adult macaques develop rash and transient fever, with neurotropic strains such as the ZIKV strain associated with the 2013 epidemic in Brazil (ZIKVBR; Brazil/ZKV2015) resulting in neuroinflammation and disruption of the BBB [14] (Supplementary Table S1).
Congenital Zika syndrome, due to vertical transfer from dam to fetus, results in a range of placental and fetal neurodevelopmental abnormalities. Disease severity is primarily influenced by fetal age at infection with the most severe outcomes occurring in early gestation, and neurotropism of ZIKV sub lineage [15,16,17,18]. However, further studies are needed to fully elucidate how the relationship between sublineage neurotropism and gestational stage of infection influences Congenital Zika syndrome outcomes. Experimental gestational ZIKV (PRVABC59) exposure at the time of implantation resulted in impaired blastocyst attachment, embryonic degeneration, and reduced embryo secretion of pregnancy hormones [19]. ZIKV (PRVABC59) infection within the first and second trimester caused premature rupture of fetal membranes, premature cervical ripening, and placental abnormalities and inflammation [20,21]. Fetal pigtail macaques (Macaca nemestrina) infected in the second or early third trimester showed disruption of fetal myelin attributed to impaired oligodendrocyte maturation [22]. Infants surviving to birth showed seizure activity and motor delay, and a single rhesus infant developed features of dilated cardiomyopathy [21]. RM and pigtail macaque fetal brain lesions included mineralization, gliosis, ependymal and neuroprogenitor cell degeneration and loss, and ventricular enlargement (ZIKV strains F2213025-2010, PRVABC59, and Brazil/ZKV2015; Supplementary Table S1) [16,21,23,24,25]. In several cases, fetal brain lesions were observed in the absence of maternal clinical disease [24]. Importantly, one limitation of the NHP model is that while neuropathic strains can induce microscopic CNS lesions and reductions in fetal brain size, ZIKV-induced microcephaly is not observed in macaques [16,21,26].

2.3. West Nile Virus

WNV is a mosquito-borne flavivirus of humans, birds, horses and related animals that causes fever, flu-like symptoms, and rarely, fatal meningoencephalitis (Supplementary Table S2). WNV initially replicates in dendritic cells in the skin near the bite site, then travels hematogenously to the CNS where it primarily infects brain and spinal cord neurons, and to a lesser extent astrocytes; microglia and macrophages are less permissive to infection [27,28]. Neuroinvasive disease occurs via hematogenous spread, or viral peripheral nerve retrograde and anterograde axonal transport [29].

2.4. NHP Models of West Nile Virus

Old-World NHP species are naturally susceptible to WNV. Seroconversion occurs within 2 weeks of infection and regional seroprevalence in outdoor colonies coincides with that of humans [30,31,32,33,34]. However, as in humans, NHP WNV infection is usually subclinical by adding a layer of complexity to the utility of NHPs as a model of WNV neurological disease [30,31,32,35,36]. Clinical disease in humans is most commonly seen in WNV infections of elderly individuals, which to a limited extent, is mirrored in NHPs [37]. While an aged Barbary macaque (Macaca sylvanus) naturally infected with WNV developed meningoencephalitis and neurological signs, attempts to experimentally induce clinical disease in aged rhesus or cynomolgus macaques (CM, Macaca fascicularis) have been unsuccessful [36,38]. In studies comparing WNV immune outcomes across different age cohorts in NHPs, T cell proliferation was delayed in aged macaques compared with younger adults, but infection remained subclinical across age groups [36]. Instead, neuroinvasive WNV disease has been achieved through intracerebral inoculation, resulting in fever and neurological signs variably progressing to death [39,40,41,42]. This model has great potential for therapeutic development.

3. Alphaviruses

The genus Alphavirus consists of 29 viral species, including three encephalitis-causing arboviruses: Western, Eastern, and Venezuelan equine encephalitis viruses [43]. The equine encephalitis viruses (EEV) produce a self-limiting fever in most humans and horses, but may progress to serious neurological manifestations, including seizures and tremors, in up to 4% of cases [44,45]. Histological lesions of human EEV consist of mononuclear inflammation, demyelination, intracerebral hemorrhage, and necrosis of the brain and occasionally spinal cord, retina, or optic nerve (Supplementary Table S3) [46,47,48,49,50,51,52]. The viruses infect neurons, astrocytes, microglia, and oligodendroglia (Supplementary Table S3) [46,53]. Historically, diagnosis has relied on brain biopsy, but thankfully, modern next-generation sequencing of serum and cerebrospinal fluid (CSF) is diagnostic [54].
Importantly, aerosolized EEV are highly infectious and can cause fatal encephalitis; this potential for use as a biological weapon renders them a major threat to public health and safety [43,55]. Of note, aerosol transmission is not a natural route, necessitating the use of animal models to evaluate the pathogenesis of and countermeasures against aerosol transmission.

3.1. Eastern Equine Encephalitis Virus

Eastern equine encephalitis virus (EEEV) is the most pathogenic of the EEV in humans and NHPs [44]. Aerosol exposure to EEEV resulted in fever, seizures, and ultimately death in CMs and common marmosets due to encephalitis (Supplementary Table S3) [43,56,57,58,59]. Of note, the neuropathogenicity of EEEV does not appear to be restricted by route of infection, as subcutaneous inoculation also resulted in clinical signs and death similar to aerosol exposure [44].

3.2. Venezuelan Equine Encephalitis Virus

Venezuelan equine encephalitis virus (VEEV) consists of 6 subtypes, I–IV, divided into epizootic or enzootic strains that naturally cycle between Culex mosquitos and rodents; humans are susceptible to both subtypes. VEEV does not cause mortality in macaques except through intracranial inoculation [45,55,60]. CMs challenged with aerosolized VEEV developed fever, viremia, and ataxia [60,61,62,63,64] (Supplementary Table S3). RM challenged subcutaneously with epizootic subtypes developed fever, anorexia, diarrhea, and depression while CMs remained subclinical; rhesus macaques developed subclinical infections with enzootic strains [44,65]. The discrepancy in aerosol and subcutaneous neuropathogenicity is thought to be due to viral entry into the olfactory system and migration to the cerebral cortex where it infects neurons and astrocytes; although, the immunohistochemistry to support this has not been demonstrated in human or NHP autopsy specimens [45,53,66,67].

3.3. Western Equine Encephalitis Virus

In contrast to EEEV and VEEV, NHP studies of Western equine encephalitis virus (WEEV) are limited. Challenge with aerosolized WEEV resulted in fever, anorexia, and tremors, with fatality in 3 of 12 CMs [68] (Supplementary Table S3). Microscopically, inflammation extended into the brain, meninges, and to a lesser degree in the spinal cord [68]. Subcutaneous exposure resulted in a subclinical infection [44].

4. Herpesviruses

Herpesviruses are double-stranded DNA viruses capable of lifelong persistence as latent infections [69]. Herpesviridae is further subclassified into the subfamilies Alphaherpesvirinae, which includes the human herpesviruses varicella zoster virus (VZV), human herpes simplex virus 1 (HSV-1), and HSV-2; Betaherpesvirinae, which includes human cytomegalovirus (HCMV), human herpesvirus (HHV)-6, and HHV-7; and Gammaherpesvirinae, which includes Epstein–Barr virus, and Kaposi’s sarcoma herpesvirus (also known as HHV8) [69,70]. Here, we will focus on NHP models for VZV, HSV-1, HSV-2, and HCMV.

4.1. Varicella-Zoster Virus

VZV is a neurotropic alphaherpesvirus that initially causes varicella (chickenpox), establishes latency, and may later reactivate to cause herpes zoster (shingles). Infection occurs by direct contact with varicella lesions or inhalation of virus-laden droplets followed by viral replication in the upper respiratory tract and dissemination to the skin resulting in the characteristic vesicular rash (Supplementary Table S4) [71]. Following resolution of varicella, VZV establishes latency in the neurons of the dorsal root sensory ganglia and later reactivates in one-third of cases as herpes zoster, a painful, debilitating disease most common in adults over 50 years of age [72]. Reactivation is associated with aging, stress, organ transplantation, or immunosuppression, but may also occur in immunocompetent individuals [73]. Importantly, while herpes zoster is primarily a disease affecting the peripheral nervous system, reactivation can lead to life-threatening complications, including pneumonia and neurological involvement [74,75]. VZV encephalitis or meningitis manifests clinically as confusion, headache, nausea, and ataxia [73,75], and detection of VZV DNA in the CSF is diagnostic [76,77,78,79]. Like other herpesviruses, intranuclear viral inclusion bodies are observed microscopically but may be more prevalent in skin lesions [80]. Neurological illness is often associated with cerebrovascular disease, vasculitis, and increased risk of stroke, although encephalitis without vasculopathy has been reported [80,81].

4.2. NHP Models of Varicella-Zoster Virus

VZV is relatively species-specific and attempts to inoculate animals with VZV have failed to consistently induce varicella rash or clinical disease [82]. Simian varicella virus (SVV, Cercopithecine Herpesvirus 9) is an alphaherpesvirus of Old-World macaques that shares 70–75% DNA homology with VZV and causes clinical disease analogous to human VZV including vesicular rash, pneumonia, and less commonly, ganglioneuritis [83] (Supplementary Table S4). A key feature of SVV as a model for VZV is its ability to cause latent infection and reactivate upon immunosuppression or stress. Experimental reactivation is induced by T lymphocyte depletion via neutralizing antibodies or whole body radiation, alone or in combination with the stress induced by transport or relocation; these methods are effective at inducing SVV reactivation and development of zoster rash [83,84].
Latency in both VZV and SVV is characterized by restricted viral transcription with some differences between the two species. Viral transcription in human VZV latency is reduced to two latency-associated transcripts: VLT, which is antisense to open reading frame (ORF) 61, and ORF63 [85]. Both ORF 61 and 63 are expressed in SVV latency; however, 10 additional transcripts were identified in latently infected sensory ganglia, albeit at significantly lower levels [86]. SVV ORF 61 appears to be dispensable for infection as RM infection with a recombinant SVV lacking ORF61 had similar disease severity and viral latency in sensory ganglia wild-type SVV [87]. However, SVV ORF61 appears to interfere with host antiviral interferon response and NF-kB signaling [87,88]. Both VZV and SVV ORF 63 are expressed during latent and acute infection and impair antiviral interferon response through IRF9 degradation [89]. Additionally, macaques have also been used in vaccine development and testing [90,91].
CNS complications of human VZV infection include meningitis, myelitis, encephalitis, or vasculitis, with the primary risk factors being compromised immune status and age. SVV CNS involvement is also rare and studies of CNS involvement in NHP models are limited, despite the immunosuppressive nature of experimental reactivation. One study found a proinflammatory cytokine response in the CSF during primary SVV infection and reactivation, with variable correlation with serum levels [92]. VZV reactivation is also associated with vascular lesions, including cerebral vasculitis and stroke [81]. Vasculitis involving the lung and skin has been reported in SVV [93]. Serum substance P was elevated in SVV, which may be associated with increased risk of stroke; however, the studies of SVV vascular and CNS outcomes are incomplete [94].

4.3. Herpes Simplex Viruses

The alpha herpesviruses of herpes simplex viruses are divided into two serotypes: HSV-1 and HSV-2, both of which are widely prevalent; two-thirds of the global population are infected with HSV-1. Clinically, HSVs cause orofacial lesions, encephalitis, and genital lesions, due to their ability to infect most cell types, including immune cells, neurons, and epithelial cells (Supplementary Table S4). Both serotypes are capable of causing similar lesions; however, HSV-1 is more prevalent and more commonly associated with orofacial lesions due to preferentially establishing latency in A5-positive neurons and reactivating in the trigeminal ganglia [95]. Conversely, HSV-2 preferentially establishes latency in KH10 positive neurons and reactivation in the lumbar-sacral ganglia, and is most frequently found in genital herpes [96]. Upon entry through the skin or mucosa, HSVs undergo local lytic replication in the inoculation site, followed by retrograde transport along sensory nerve axons to peripheral sensory ganglia where they establish latency [97]. During stress-induced reactivation, HSV exits the neuron through anterograde transport and infects local cells or neurons, resulting in superficial mucocutaneous or CNS lesions [97]. Of further public health significance, HSV-2 infection is associated with a 3-fold increased risk of acquiring HIV, lending critical value to the NHP coinfection model in treatment and prevention strategies [98].

4.4. NHP Models of Herpes Simplex Virus

While many NHP viral infection models center on Old-World NHP species, HSV research spans both Old and New World models. Macaques (Old World monkeys) are susceptible to HSV infection, but less so than New World species such as marmosets (Callithrix) and owl monkeys (Aotus). This reduced susceptibility is thought to be due to simian Tripartite motif 5α (TRIM5α), which reduced HSV-1 and -2 replication in fibroblasts from RM and African green monkeys (AGM, Chlorocebus sabaeus) [99].
RM HSV-1 studies typically focus on oral and genital infection pathogenesis and development and testing of multivalent vaccines, microbicides, therapeutics, and prevention technologies [100,101,102,103,104,105,106,107,108]. RMs inoculated with HSV-1 via lip scratches developed oral vesicular lesions within 3–5 days, which resolved within 2 weeks but recurred months later in some cases [109]. CNS lesions included inflammatory cell infiltrates in the spinal cord and brain, and HSV-1 antigen was detected in these sites [109]. HSV-induced mucosal breaches facilitate HIV infection; macaque co-infection studies have also demonstrated that HSV-1 inoculation in SHIV- or SIV-infected animals resulted in vaginal inflammatory cytokine production, intermittent shedding, oral lesions, and detection of virus in trigeminal ganglia [110,111].
The utility of RMs as a model for HSV-2 is somewhat controversial; findings from at least one study suggested RMs are refractory to HSV-2 [112]. Alternatively, RMs challenged with a cocktail of HSV-2 strains demonstrated limited viral shedding, latency and reactivation, and histological lesions; however, anti-HSV-2 antibodies were only detected in 30% of animals [113]. HSV-2 failed to replicate in primary rhesus fibroblasts [114].
In contrast with macaques, New World monkeys, particularly marmosets and owl monkeys, are exquisitely sensitive to HSV infection, and are key models of HSV-1 encephalitis [115,116,117]. Marmosets naturally or experimentally infected with HSV-1 developed anorexia, dyspnea, ataxia, vesicular stomatitis, and meningoencephalitis [115,118,119,120]. In contrast, capuchins (Cebus apella), another New World primate species, exhibited HSV-2 vulvovaginal vesicular lesions in the absence of neurological symptoms [114].

4.5. Cytomegalovirus

The betaherpesvirus human cytomegalovirus (HCMV), also known as human herpesvirus 5, establishes latency after primary infection resulting in lifelong infection with the potential for reactivation. While infection in immunocompetent individuals is usually subclinical, HCMV is a common opportunistic pathogen of the immunocompromised resulting in viral dissemination and disease [121]. Furthermore, congenital HCMV infection causes fetal and neonatal disorders in 10% of cases, including neurodevelopmental delays, sensorineural hearing loss, retinitis, or microcephaly [122,123,124] (Supplementary Table S4). Diagnosis often relies on viral isolation from urine or saliva, and microscopic evaluation often reveals the characteristic “owl’s eye” inclusion bodies [121,123]. Because CMV infection in immunocompetent individuals typically results in nonfatal outcomes, animal models are key for understanding pathogenesis.

4.6. NHP Models of Cytomegalovirus

One major challenge in HCMV research is that cytomegaloviruses are highly species-specific [125,126]. Fortuitously, human and rhesus (Rh)CMV are genetically and structurally similar, and RhCMV recapitulates many features of human HCMV infection rendering it a useful experimental model for HCMV [127,128]. Still, a barrier remains, as the vast majority of macaques in primate centers and wild colonies are RhCMV seropositive by one year of age, and, as in HCMV, RhCMV generally results in subclinical infections [129,130]. Historically, seropositive rhesus only developed clinical disease with SIV-induced immunodeficiency, resulting in multisystemic lesions [131]. Neuroinflammation may be observed with initial infection or reactivation in immunosuppressed or stressed animals.
The similarities between human and NHP reproductive biology lend relevance to the congenital macaque model, as rhesus pregnancies are also divided into trimesters and demonstrate similar placental and fetal development [132]. However, natural intrauterine transmission in rhesus is thought to be uncommon given the high seroprevalence and natural infection in endemic colonies and the fact that congenital transmission occurs most commonly when primary CMV infection occurs during pregnancy [129]. Experimental models using seronegative RMs have been modestly successful [133]. In one study, fetal loss was only observed in an acutely infected (at 8 weeks gestation) rhesus in which CD4+ T cells had been depleted; an immunocompetent dam not treated with CD4+ T cell depleting antibodies did not transmit CMV to her fetus [134].

5. Human Immunodeficiency Virus

The human immunodeficiency virus (HIV) belongs to the enveloped lentivirus subgroup within the retrovirus family. Each viral particle, or virion, encapsulates two copies of positive-sense, single-stranded viral RNA. This genetic material undergoes reverse transcription within the host cell, producing double-stranded DNA that is integrated into the host genome, establishing a covert life-long presence that defines the insidious nature of HIV infection. The virus predominantly targets CD4+ cells of the immune system, including macrophages, dendritic cells, and most notably, CD4+ T cells. Infection may culminate in the development of acquired immunodeficiency syndrome (AIDS), characterized by the progressive reduction in CD4+ T cells that results in loss of cell-mediated immunity and renders infected individuals susceptible to typically rare opportunistic infections, malignant neoplasms, and neurological injury.
Immune impairment and progression to AIDS have been markedly curbed in recent times through the implementation of highly effective combination antiretroviral therapy (cART). This therapeutic approach efficiently suppresses viral replication, facilitating immune system restoration, and consequently leading to substantial enhancements in both the quality and duration of life for individuals living with HIV. Yet, this does not cure individuals of infection and long-lived latently infected cells persist and establish stable tissue reservoirs of virus that recrudesce with cART cessation. Additionally, individuals undergoing successful cART experience significant health complications, such as cardiovascular disease, metabolic syndrome, and HIV-associated neurocognitive disorders (HAND). As such, HIV research that employs models that faithfully reflect the complexities of HIV infection and cART aids in dissecting the underlying mechanisms that foster comorbidities. This research not only enhances clinical management but also uncovers opportunities for preventive measures and tailored interventions to mitigate the risk of these comorbidities among individuals living with HIV.

NHP Models of HIV: Simian Immunodeficiency Virus

Asian-origin macaques, including RMs infected with simian immunodeficiency virus (SIV) are widely recognized as the “gold standard” in HIV research, owing to their high translatability to various aspects of human disease. Several critical factors contribute to this status. Firstly, RMs are naturally vulnerable to SIV infection, which shares noteworthy similarities with HIV in critical aspects such as cell tropism, molecular structure, and the viral life cycle. Both viruses predominantly target CD4+ T cells, thereby causing immune dysfunction and eventually progressing to AIDS. This allows for in-depth exploration of viral-immune system interactions that closely emulate the human condition.
Another point of convergence between SIV and HIV is the molecular homology between the two viruses, which holds significant implications for the effectiveness of cART in SIV-infected RMs. HIV-targeting antiretroviral drugs operate by inhibiting key viral proteins essential for various life cycle stages, including viral attachment, fusion, reverse transcription, integration, assembly, and budding. Due to their structural similarity, SIV proteins are also susceptible to these antiretroviral agents, enabling effective therapeutic interventions. Of equal importance is the alignment of drug resistance mechanisms between SIV and HIV. Investigations into the development of drug resistance in SIV can thereby yield actionable insights into corresponding strategies for managing or preventing resistance in HIV therapies. Furthermore, the compatibility of cART in SIV-infected RMs facilitates research into reservoir-targeting strategies, a central area of focus in current efforts to achieve a functional cure for HIV. Although several of these therapeutic strategies show promise, it is crucial to note that the majority remain in the experimental phase, with many being tested primarily in animal models like SIV-infected RMs.
Despite these many parallels, several notable differences exist between SIV infection in RMs and HIV infection in humans, particularly related to the anatomical distribution, cellular composition, and establishment of latent viral reservoirs. In HIV-infected humans, viral replication and latency are predominantly localized in secondary lymphoid tissues such as lymph nodes, spleen, and gut-associated lymphoid tissue (GALT), as well as in long-lived memory CD4+ T cell subsets. Tissue tropism in cART-treated SIV-infected rhesus macaques reflects important nuances in reservoir localization and persistence that are critical for modeling HIV pathogenesis and cure strategies. Following infection, SIV disseminates rapidly to multiple tissue compartments, including gut-associated lymphoid tissue (GALT), lymph nodes, spleen, and CNS. Even under effective cART, viral DNA and replication-competent virus persist in these tissues, forming stable reservoirs that are refractory to immune clearance and therapeutic suppression [135,136,137]. Among these, the gastrointestinal mucosa and lymphoid tissues remain major sites of latent infection, harboring infected CD4+ T cells, particularly within transitional and effector memory subsets, despite prolonged viral suppression.
Within the CNS, perivascular and meningeal macrophages represent the principal cellular reservoirs of SIV during suppressive cART [138]. These cells can harbor integrated viral DNA, even in the absence of detectable plasma viremia. Microglia, while susceptible to SIV infection, appear to be less consistently infected during cART, and their contribution to the persistent CNS reservoir may depend on factors such as timing of therapy initiation and the neurotropism of the viral strain used. Importantly, low-level viral RNA and chronic neuroinflammation can be detected in the CNS of cART-treated macaques, implicating this compartment as both a sanctuary site and a contributor to HAND-like pathology [139].
The choice of viral strain significantly influences tissue tropism and reservoir dynamics. SIVmac239, a neurovirulent, molecularly cloned strain, exhibits robust seeding of peripheral and CNS reservoirs and is frequently used in cure and pathogenesis studies due to its high replicative fitness and consistent induction of systemic and CNS infection. SIVmac251, a more genetically heterogeneous quasispecies, demonstrates broader mucosal transmissibility and sustained infection across lymphoid and non-lymphoid tissues, while also supporting CNS persistence during long-term cART [140]. While both strains permit rigorous evaluation of tissue reservoirs and therapeutic interventions targeting viral latency, differences in viral dynamics must be accounted for in experimental design and data interpretation.
In addition to the significant relevance of macaques in therapeutic development and cure strategies for HIV, RMs are invaluable models for many key aspects of HIV neuropathogenesis. Due to the responsiveness of SIV-infected RMs to cART, they provide a critical platform for investigating the mechanisms and dynamics of the entry, replication, and potential persistence of SIV/HIV in the brain in the context of current antiretroviral treatment [138,141,142]. Chronic neuroinflammation remains a hallmark of HIV neuropathogenesis, even in the current era of effective cART, and likely plays a major role in HAND development and progression [143,144,145,146]. SIV-infected, cART-treated RMs also demonstrate evidence of persistent neuroinflammation [147], allowing researchers to examine the inflammatory mechanisms in the brain that underly neuronal injury and cognitive dysfunction in the context of virus suppression and immune restoration, which cannot be fully investigated in living human subjects.

6. Coronaviruses of Severe Acute Respiratory Syndrome

The Betacoronavirus genus of the Coronaviridae family comprises several viruses that primarily cause respiratory disease. Notable among these include severe acute respiratory syndrome coronavirus (SARS-CoV), Middle East respiratory syndrome coronavirus, and most recently, SARS-CoV-2. Each of these viruses has been linked to pandemics in the last two decades. Our focus here is on SARS-CoV-2, the etiologic agent of coronavirus disease 2019 (COVID-19), which emerged in 2019. Coronaviruses are enveloped, positive-sense, single-stranded RNA viruses, containing several structural proteins associated with the viral envelope, a ribonucleoprotein complex comprising the viral genome and nucleocapsid protein (N), and a replicase complex that encodes nonstructural proteins [148,149,150]. Nonstructural proteins include proteases and RNA-dependent RNA polymerase (RdRp) that enable viral replication. Structural proteins, spike (S), membrane (M), and envelope (E) are anchored within and comprise the majority of the viral envelope.
Coronavirus S proteins are critical for infection by facilitating cell binding and fusion through interaction with host cell receptors, such as the angiotensin-converting enzyme-2 (ACE2) receptor required for the entry of SARS-CoV-2 into host cells [151]. SARS-CoV-2 transmission typically occurs through the inhalation of contaminated droplets. The viral S protein binds the host cell receptors ACE2 and serine protease transmembrane 2 (TMPRSS2), facilitating viral entry into the host cell cytoplasm. This entry allows the viral RNA genome to be translated into structural proteins and polyproteins, promoting viral replication and the release of new viral particles.
In humans, SARS-CoV-2 infection presents a spectrum of clinical symptoms, ranging from asymptomatic or mild infection to severe manifestations, such as respiratory distress and multiorgan failure. A major challenge in combating the COVID-19 pandemic has been the long-term and extrapulmonary effects of infection. These include long-COVID, respiratory disease, and notable neurological manifestations, including headache and neurocognitive impairment.

NHP Models of SARS-CoV-2

NHPs, particularly the macaques, serve as ideal models for studying SARS-CoV-2 pathogenesis and vaccine development. They are naturally permissive to infection and accurately model clinical pathology and immunologic features of human disease [152]. Due to the high conservation of the ACE2 amino acid sequence between RMs, CMs, and humans, as well as similarities in immune responses, NHPs are crucial models for testing vaccine efficacy [153,154]. Like humans, SARS-CoV-2-infected NHPs exhibit transient viral replication in the airways, and often mild, self-limiting lung inflammation [152,155,156]; however, some study subjects develop more severe infection, characterized by respiratory distress, severe broncho-interstitial pneumonia, and, in extreme cases, death [157].
NHP models of SARS-CoV-2 infection also reflect the neurological complications of infection observed in humans. A study examining the CSF proteome post-infection in AGMs and RMs revealed an enrichment of CSF proteins associated with neuroinflammation, innate immunity, and hemostasis [158]. This is consistent with human studies, where CSF leukocytes exhibited an enhanced inflammatory signature [159]. Additionally, the brains of infected RMs and AGMs demonstrated marked neuroinflammation, microhemorrhages, and neuron degeneration and apoptosis [160], paralleling neuropathological changes reported in humans [161,162]. Notably, CNS injury and neuroinflammation were observed in animals at approximately 4 weeks post-infection and in animals that did not experience severe respiratory disease [160], suggesting ongoing neurological involvement after resolution of acute infection.
SARS-CoV-2 enters host cells via ACE2, which is expressed not only in the respiratory tract but also in various brain regions, including the olfactory bulb, hippocampus, hypothalamus, and brainstem. Variable ACE2 expression in these areas has been confirmed in both human and NHP brains [163,164], raising the possibility that SARS-CoV-2 may access the CNS through neural pathways such as the olfactory nerve. Indeed, viral RNA and protein have been detected in the olfactory bulb and associated cortices in both human postmortem tissue [165] and infected NHPs [166], supporting the hypothesis of transneuronal viral spread. These findings may explain common clinical manifestations such as anosmia and ageusia during acute infection [167]. Further, ACE2 expression in limbic and hypothalamic structures suggests that direct viral effects and neuroinflammation in these regions may contribute to neuropsychiatric symptoms reported during and after infection, including depression, anxiety, and cognitive impairment [168,169,170]. Importantly, infected NHPs exhibit robust transcriptional and histopathological alterations indicative of synaptic plasticity changes, neuroinflammation, and cerebrovascular injury, even in the absence of detectable virus [160,171], providing important mechanistic insight into neurocognitive disturbances and mood disorders that can persist long after viral clearance.
Together, these data underscore the utility of NHP models not only for characterizing the systemic and pulmonary manifestations of SARS-CoV-2 infection but also for elucidating potential routes for viral neuroinvasion and the impact of the virus on CNS function. The anatomical and functional parallels between NHP and human brains, including ACE2 distribution, make these models especially valuable for dissecting the neurobiological substrates of long COVID.

7. Discussion

Within the past few decades, events such as the Zika virus epidemic and the SARS-CoV-2 pandemic have highlighted the importance of appropriate research studies of viral pathogens. Animal models are critical for overcoming the challenges encountered in recapitulating human viral infections, particularly outcomes on the brain and the interaction between the central nervous system and the body. There are several limitations of NHP models and future directions of viral-associated CNS disease in NHP models we would like to highlight. The NHP model does not always fully mirror human disease, as seen with ZIKV microcephaly and HSV-2 infection. Others mirror human subclinical infection and require experimental immunosuppression to consistently achieve reactivation or clinical disease.
WNV encephalitis generally impacts elderly patients with co-morbidities, which likely exacerbate age-related immune suppression. While antiviral T-cell proliferative responses appeared to be delayed in aged macaques experimentally infected with WNV, aging alone was not sufficient to permit neuroinvasive disease. Natural WNV infection of an aged Barbary macaque resulted in encephalitis; this animal had an underlying co-morbidity (osteoarthritis), which may have contributed to a higher baseline inflammatory level, tipping the balance in favor of disease progression [38]. Further study is needed to define the interface between aging, specific co-morbidities, and progression to WNV encephalitis in NHP models.
One major caveat to the NHP model for VZV is that while neurological involvements, such as meningitis or stroke, are grave complications of VZV reactivation, CNS complications are not well described in NHP SVV infections. Additional studies are needed to characterize the pathological changes in the brain, vasculature, and CSF during acute SVV infection as well as during reactivation.
NHP models enable longitudinal studies of natural disease and probe the cellular and molecular outcomes during the progression of disease in an intact immune system, with more complete and thorough characterization of therapeutic efficacy or toxicity. For many viruses, NHP models represent a highly analogous model for human disease due to parallels in life span, metabolic and reproductive biology. Additionally, genetic similarities particularly with host-specific viruses and analogous pathogens render the NHP model invaluable.

Supplementary Materials

The supporting information can be downloaded at https://www.mdpi.com/article/10.3390/ijms26146886/s1.

Author Contributions

Conceptualization, K.J.V. and T.F.; writing—original draft preparation, K.J.V. and T.F.; writing—review and editing, K.J.V., T.F., B.N.M. and L.H.; funding acquisition, K.J.V. and T.F. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by NIH ORIP P51OD011104 and NIH ORIP K01OD036106 (KJV).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created.

Acknowledgments

This work was supported by RRID:SCR_008167. The authors gratefully thank the reviewers for their invaluable feedback and critiques. The authors have reviewed and edited the output and take full responsibility for the content of this publication.

Conflicts of Interest

The authors declare no conflicts of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

Abbreviations

The following abbreviations are used in this manuscript:
ACE-2Angiotensin-Converting Enzyme-2
AGMAfrican green monkey
AIDSAcquired immunodeficiency syndrome
cARTCombination antiretroviral therapy
CNSCentral nervous system
COVID-19Coronavirus disease 2019
CMCynomolgus macaque
EEEVEastern equine encephalitis virus
EEVEquine encephalitis virus
HANDHIV-associated neurocognitive disorders
HIVHuman immunodeficiency virus
HCMVHuman cytomegalovirus
HSVHerpes simplex virus
NHPNon-human primate
ORFOpen reading frame
RMRhesus macaque
RhCMVRhesus cytomegalovirus
SARS-CoV-2Coronavirus of severe acute respiratory syndrome
SIVSimian immunodeficiency virus
SVVSimian varicella virus
VEEVVenezuelan equine encephalitis virus
VZVVaricella zoster virus
WEEEWestern equine encephalitis virus
WNVWest Nile virus
ZIKVZika virus

References

  1. Hoogland, I.C.; Houbolt, C.; van Westerloo, D.J.; van Gool, W.A.; van de Beek, D. Systemic inflammation and microglial activation: Systematic review of animal experiments. J. Neuroinflamm. 2015, 12, 114. [Google Scholar] [CrossRef] [PubMed]
  2. Dick, G.W.A.; Kitchen, S.F.; Haddow, A.J. Zika Virus (I). Isolations and serological specificity. Trans. R. Soc. Trop. Med. Hyg. 1952, 46, 509–520. [Google Scholar] [CrossRef] [PubMed]
  3. Duffy, M.R.; Chen, T.-H.; Hancock, W.T.; Powers, A.M.; Kool, J.L.; Lanciotti, R.S.; Pretrick, M.; Marfel, M.; Holzbauer, S.; Dubray, C.; et al. Zika Virus Outbreak on Yap Island, Federated States of Micronesia. N. Engl. J. Med. 2009, 360, 2536–2543. [Google Scholar] [CrossRef] [PubMed]
  4. Diallo, D.; Sall, A.A.; Diagne, C.T.; Faye, O.; Faye, O.; Ba, Y.; Hanley, K.A.; Buenemann, M.; Weaver, S.C.; Diallo, M. Zika Virus Emergence in Mosquitoes in Southeastern Senegal, 2011. PLoS ONE 2014, 9, e109442. [Google Scholar] [CrossRef] [PubMed]
  5. Mansuy, J.M.; Dutertre, M.; Mengelle, C.; Fourcade, C.; Marchou, B.; Delobel, P.; Izopet, J.; Martin-Blondel, G. Zika virus: High infectious viral load in semen, a new sexually transmitted pathogen? Lancet Infect. Dis. 2016, 16, 405. [Google Scholar] [CrossRef] [PubMed]
  6. Christian, K.M.; Song, H.; Ming, G.-L. Pathophysiology and Mechanisms of Zika Virus Infection in the Nervous System. Annu. Rev. Neurosci. 2019, 42, 249–269. [Google Scholar] [CrossRef] [PubMed]
  7. Miner, J.J.; Diamond, M.S. Zika Virus Pathogenesis and Tissue Tropism. Cell Host Microbe 2017, 21, 134–142. [Google Scholar] [CrossRef] [PubMed]
  8. Komarasamy, T.V.; Adnan, N.A.A.; James, W.; Balasubramaniam, V. Zika Virus Neuropathogenesis: The Different Brain Cells, Host Factors and Mechanisms Involved. Front. Immunol. 2022, 13, 773191. [Google Scholar] [CrossRef] [PubMed]
  9. Liang, Q.; Luo, Z.; Zeng, J.; Chen, W.; Foo, S.S.; Lee, S.A.; Ge, J.; Wang, S.; Goldman, S.A.; Zlokovic, B.V.; et al. Zika Virus NS4A and NS4B Proteins Deregulate Akt-mTOR Signaling in Human Fetal Neural Stem Cells to Inhibit Neurogenesis and Induce Autophagy. Cell Stem Cell 2016, 19, 663–671. [Google Scholar] [CrossRef] [PubMed]
  10. Bindu; Pandey, H.S.; Seth, P. Interplay Between Zika Virus-Induced Autophagy and Neural Stem Cell Fate Determination. Mol. Neurobiol. 2023, 61, 9927–9944. [Google Scholar] [CrossRef] [PubMed]
  11. Pielnaa, P.; Al-Saadawe, M.; Saro, A.; Dama, M.F.; Zhou, M.; Huang, Y.; Huang, J.; Xia, Z. Zika virus-spread, epidemiology, genome, transmission cycle, clinical manifestation, associated challenges, vaccine and antiviral drug development. Virology 2020, 543, 34–42. [Google Scholar] [CrossRef] [PubMed]
  12. Rayner, J.O.; Kalkeri, R.; Goebel, S.; Cai, Z.; Green, B.; Lin, S.; Snyder, B.; Hagelin, K.; Walters, K.B.; Koide, F. Comparative Pathogenesis of Asian and African-Lineage Zika Virus in Indian Rhesus Macaque’s and Development of a Non-Human Primate Model Suitable for the Evaluation of New Drugs and Vaccines. Viruses 2018, 10, 229. [Google Scholar] [CrossRef] [PubMed]
  13. Moreno, G.K.; Newman, C.M.; Koenig, M.R.; Mohns, M.S.; Weiler, A.M.; Rybarczyk, S.; Weisgrau, K.L.; Vosler, L.J.; Pomplun, N.; Schultz-Darken, N.; et al. Long-Term Protection of Rhesus Macaques from Zika Virus Reinfection. J. Virol. 2020, 94, e01881-19. [Google Scholar] [CrossRef] [PubMed]
  14. Panganiban, A.T.; Blair, R.V.; Hattler, J.B.; Bohannon, D.G.; Bonaldo, M.C.; Schouest, B.; Maness, N.J.; Kim, W.-K. A Zika virus primary isolate induces neuroinflammation, compromises the blood-brain barrier and upregulates CXCL12 in adult macaques. Brain Pathol. 2020, 30, 1017–1027. [Google Scholar] [CrossRef] [PubMed]
  15. Hirsch, A.J.; Roberts, V.H.J.; Grigsby, P.L.; Haese, N.; Schabel, M.C.; Wang, X.; Lo, J.O.; Liu, Z.; Kroenke, C.D.; Smith, J.L.; et al. Zika virus infection in pregnant rhesus macaques causes placental dysfunction and immunopathology. Nat. Commun. 2018, 9, 263. [Google Scholar] [CrossRef] [PubMed]
  16. Martinot, A.J.; Abbink, P.; Afacan, O.; Prohl, A.K.; Bronson, R.; Hecht, J.L.; Borducchi, E.N.; Larocca, R.A.; Peterson, R.L.; Rinaldi, W.; et al. Fetal Neuropathology in Zika Virus-Infected Pregnant Female Rhesus Monkeys. Cell 2018, 173, 1111–1122.e1110. [Google Scholar] [CrossRef] [PubMed]
  17. Gurung, S.; Reuter, N.; Preno, A.; Dubaut, J.; Nadeau, H.; Hyatt, K.; Singleton, K.; Martin, A.; Parks, W.T.; Papin, J.F.; et al. Zika virus infection at mid-gestation results in fetal cerebral cortical injury and fetal death in the olive baboon. PLoS Pathog. 2019, 15, e1007507. [Google Scholar] [CrossRef] [PubMed]
  18. de Alwis, R.; Zellweger, R.M.; Chua, E.; Wang, L.-F.; Chawla, T.; Sessions, O.M.; Marlier, D.; Connolly, J.E.; von Messling, V.; Anderson, D.E. Systemic inflammation, innate immunity and pathogenesis after Zika virus infection in cynomolgus macaques are modulated by strain-specificity within the Asian lineage. Emerg. Microbes Infect. 2021, 10, 1457–1470. [Google Scholar] [CrossRef] [PubMed]
  19. Block, L.N.; Aliota, M.T.; Friedrich, T.C.; Schotzko, M.L.; Mean, K.D.; Wiepz, G.J.; Golos, T.G.; Schmidt, J.K. Embryotoxic impact of Zika virus in a rhesus macaque in vitro implantation model. Biol. Reprod. 2020, 102, 806–816. [Google Scholar] [CrossRef] [PubMed]
  20. Mohr, E.L.; Block, L.N.; Newman, C.M.; Stewart, L.M.; Koenig, M.; Semler, M.; Breitbach, M.E.; Teixeira, L.B.C.; Zeng, X.; Weiler, A.M.; et al. Ocular and uteroplacental pathology in a macaque pregnancy with congenital Zika virus infection. PLoS ONE 2018, 13, e0190617. [Google Scholar] [CrossRef] [PubMed]
  21. Steinbach, R.J.; Haese, N.N.; Smith, J.L.; Colgin, L.M.A.; MacAllister, R.P.; Greene, J.M.; Parkins, C.J.; Kempton, J.B.; Porsov, E.; Wang, X.; et al. A neonatal nonhuman primate model of gestational Zika virus infection with evidence of microencephaly, seizures and cardiomyopathy. PLoS ONE 2020, 15, e0227676. [Google Scholar] [CrossRef] [PubMed]
  22. Tisoncik-Go, J.; Stokes, C.; Whitmore, L.S.; Newhouse, D.J.; Voss, K.; Gustin, A.; Sung, C.J.; Smith, E.; Stencel-Baerenwald, J.; Parker, E.; et al. Disruption of myelin structure and oligodendrocyte maturation in a macaque model of congenital Zika infection. Nat. Commun. 2024, 15, 5173. [Google Scholar] [CrossRef] [PubMed]
  23. Adams Waldorf, K.M.; Nelson, B.R.; Stencel-Baerenwald, J.E.; Studholme, C.; Kapur, R.P.; Armistead, B.; Walker, C.L.; Merillat, S.; Vornhagen, J.; Tisoncik-Go, J.; et al. Congenital Zika virus infection as a silent pathology with loss of neurogenic output in the fetal brain. Nat. Med. 2018, 24, 368–374. [Google Scholar] [CrossRef] [PubMed]
  24. Adams Waldorf, K.M.; Stencel-Baerenwald, J.E.; Kapur, R.P.; Studholme, C.; Boldenow, E.; Vornhagen, J.; Baldessari, A.; Dighe, M.K.; Thiel, J.; Merillat, S.; et al. Fetal brain lesions after subcutaneous inoculation of Zika virus in a pregnant nonhuman primate. Nat. Med. 2016, 22, 1256–1259. [Google Scholar] [CrossRef] [PubMed]
  25. Coffey, L.L.; Keesler, R.I.; Pesavento, P.A.; Woolard, K.; Singapuri, A.; Watanabe, J.; Cruzen, C.; Christe, K.L.; Usachenko, J.; Yee, J.; et al. Intraamniotic Zika virus inoculation of pregnant rhesus macaques produces fetal neurologic disease. Nat. Commun. 2018, 9, 2414. [Google Scholar] [CrossRef] [PubMed]
  26. Robbiani, D.F.; Olsen, P.C.; Costa, F.; Wang, Q.; Oliveira, T.Y.; Nery, N., Jr.; Aromolaran, A.; do Rosário, M.S.; Sacramento, G.A.; Cruz, J.S.; et al. Risk of Zika microcephaly correlates with features of maternal antibodies. J. Exp. Med. 2019, 216, 2302–2315. [Google Scholar] [CrossRef] [PubMed]
  27. Shrestha, B.; Gottlieb, D.; Diamond, M.S. Infection and injury of neurons by West Nile encephalitis virus. J. Virol. 2003, 77, 13203–13213. [Google Scholar] [CrossRef] [PubMed]
  28. van Marle, G.; Antony, J.; Ostermann, H.; Dunham, C.; Hunt, T.; Halliday, W.; Maingat, F.; Urbanowski, M.D.; Hobman, T.; Peeling, J.; et al. West Nile Virus-Induced Neuroinflammation: Glial Infection and Capsid Protein-Mediated Neurovirulence. J. Virol. 2007, 81, 10933–10949. [Google Scholar] [CrossRef] [PubMed]
  29. Samuel, M.A.; Wang, H.; Siddharthan, V.; Morrey, J.D.; Diamond, M.S. Axonal transport mediates West Nile virus entry into the central nervous system and induces acute flaccid paralysis. Proc. Natl. Acad. Sci. USA 2007, 104, 17140–17145. [Google Scholar] [CrossRef] [PubMed]
  30. Cohen, J.K.; Kilpatrick, M.A.; Connor Stroud, F.; Paul, K.; Wolf, F.; Else, J.G. Seroprevalence of West Nile virus in nonhuman primates as related to mosquito abundance at two national primate research centers. Comp. Med. 2007, 57, 115–119. [Google Scholar] [PubMed]
  31. Ratterree, M.S.; da Rosa, A.P.T.; Bohm, R.P.; Cogswell, F.B.; Phillippi, K.M.; Caillouet, K.; Schwanberger, S.; Shope, R.E.; Tesh, R.B. West Nile virus infection in nonhuman primate breeding colony, concurrent with human epidemic, southern Louisiana. Emerg Infect Dis. 2003, 9, 1388–1394. [Google Scholar] [CrossRef] [PubMed]
  32. Robertson, S.N.; Cameron, A.I.; Morales, P.R.; Burnside, W.M. West Nile Virus Seroprevalence in an Outdoor Nonhuman Primate Breeding Colony in South Florida. J. Am. Assoc. Lab. Anim. Sci. 2021, 60, 168–175. [Google Scholar] [CrossRef] [PubMed]
  33. Ratterree, M.S.; Gutierrez, R.A.; Travassos da Rosa, A.P.; Dille, B.J.; Beasley, D.W.; Bohm, R.P.; Desai, S.M.; Didier, P.J.; Bikenmeyer, L.G.; Dawson, G.J. Experimental infection of rhesus macaques with West Nile virus: Level and duration of viremia and kinetics of the antibody response after infection. J. Infect. Dis. 2004, 189, 669–676. [Google Scholar] [CrossRef] [PubMed]
  34. Wolf, R.F.; Papin, J.F.; Hines-Boykin, R.; Chavez-Suarez, M.; White, G.L.; Sakalian, M.; Dittmer, D.P. Baboon model for West Nile Virus infection and vaccine evaluation. Virology 2006, 355, 44–51. [Google Scholar] [CrossRef] [PubMed]
  35. Verstrepen, B.E.; Fagrouch, Z.; van Heteren, M.; Buitendijk, H.; Haaksma, T.; Beenhakker, N.; Palù, G.; Richner, J.M.; Diamond, M.S.; Bogers, W.M.; et al. Experimental infection of rhesus macaques and common marmosets with a European strain of West Nile virus. PLoS Negl. Trop. Dis. 2014, 8, e2797. [Google Scholar] [CrossRef] [PubMed]
  36. Wertheimer, A.M.; Uhrlaub, J.L.; Hirsch, A.; Medigeshi, G.; Sprague, J.; Legasse, A.; Wilk, J.; Wiley, C.A.; Didier, P.; Tesh, R.B.; et al. Immune response to the West Nile virus in aged non-human primates. PLoS ONE 2010, 5, e15514. [Google Scholar] [CrossRef] [PubMed]
  37. Sampson, B.A.; Ambrosi, C.; Charlot, A.; Reiber, K.; Veress, J.F.; Armbrustmacher, V. The pathology of human West Nile virus infection. Hum. Pathol. 2000, 31, 527–531. [Google Scholar] [CrossRef] [PubMed]
  38. Ølberg, R.-A.; Barker, I.K.; Crawshaw, G.J.; Bertelsen, M.F.; Drebot, M.A.; Andonova, M. West Nile virus encephalitis in a Barbary macaque (Macaca sylvanus). Emerg. Infect. Dis. 2004, 10, 712. [Google Scholar] [CrossRef] [PubMed]
  39. Manuelidis, E.E. Neuropathology of Experimental West Nile Virus Infection in Monkeys. J. Neuropathol. Exp. Neurol. 1956, 15, 448–460. [Google Scholar] [CrossRef] [PubMed]
  40. Pogodina, V.V.; Frolova, M.P.; Malenko, G.V.; Fokina, G.I.; Koreshkova, G.V.; Kiseleva, L.L.; Bochkova, N.G.; Ralph, N.M. Study on West Nile virus persistence in monkeys. Arch. Virol. 1983, 75, 71–86. [Google Scholar] [CrossRef] [PubMed]
  41. Maximova, O.A.; Sturdevant, D.E.; Kash, J.C.; Kanakabandi, K.; Xiao, Y.; Minai, M.; Moore, I.N.; Taubenberger, J.; Martens, C.; Cohen, J.I.; et al. Virus infection of the CNS disrupts the immune-neural-synaptic axis via induction of pleiotropic gene regulation of host responses. eLife 2021, 10, e62273. [Google Scholar] [CrossRef] [PubMed]
  42. Maximova, O.A.; Speicher, J.M.; Skinner, J.R.; Murphy, B.R.; St Claire, M.C.; Ragland, D.R.; Herbert, R.L.; Pare, D.R.; Moore, R.M.; Pletnev, A.G. Assurance of neuroattenuation of a live vaccine against West Nile virus: A comprehensive study of neuropathogenesis after infection with chimeric WN/DEN4Δ30 vaccine in comparison to two parental viruses and a surrogate flavivirus reference vaccine. Vaccine 2014, 32, 3187–3197. [Google Scholar] [CrossRef] [PubMed]
  43. Weaver, S.C.; Winegar, R.; Manger, I.D.; Forrester, N.L. Alphaviruses: Population genetics and determinants of emergence. Antivir. Res. 2012, 94, 242–257. [Google Scholar] [CrossRef] [PubMed]
  44. Smith, D.R.; Schmaljohn, C.S.; Badger, C.; Ostrowski, K.; Zeng, X.; Grimes, S.D.; Rayner, J.O. Comparative pathology study of Venezuelan, eastern, and western equine encephalitis viruses in non-human primates. Antivir. Res. 2020, 182, 104875. [Google Scholar] [CrossRef] [PubMed]
  45. Steele, K.E.; Twenhafel, N.A. REVIEW PAPER: Pathology of Animal Models of Alphavirus Encephalitis. Vet. Pathol. 2010, 47, 790–805. [Google Scholar] [CrossRef] [PubMed]
  46. Bastian, F.O.; Wende, R.D.; Singer, D.B.; Zeller, R.S. Eastern Equine Encephalomyelitis: Histopathologic and Ultrastructural Changes with Isolation of the Virus in a Human Case. Am. J. Clin. Pathol. 1975, 64, 10–13. [Google Scholar] [CrossRef] [PubMed]
  47. Anderson, B.A. Focal neurologic signs in western equine encephalitis. Can. Med. Assoc. J. 1984, 130, 1019–1021. [Google Scholar] [PubMed]
  48. Bruyn, H.B.; Lennette, E.H. Western equine encephalitis in infants; a report on three cases with sequelae. Calif. Med. 1953, 79, 362–366. [Google Scholar] [PubMed]
  49. de la Monte, S.M.; Castro, F.; Bonilla, N.J.; de Urdaneta, A.G.; Hutchins, G.M. The Systemic Pathology of Venezuelan Equine Encephalitis Virus Infection in Humans. Am. J. Trop. Med. Hyg. 1985, 34, 194–202. [Google Scholar] [CrossRef] [PubMed]
  50. Delfraro, A.; Burgueño, A.; Morel, N.; González, G.; García, A.; Morelli, J.; Pérez, W.; Chiparelli, H.; Arbiza, J. Fatal human case of Western equine encephalitis, Uruguay. Emerg. Infect. Dis. 2011, 17, 952–954. [Google Scholar] [CrossRef] [PubMed]
  51. Englund, J.A.; Breningstall, G.N.; Heck, L.J.; Lazuick, J.S.; Karabatsos, N.; Calisher, C.H.; Tsai, T.F. Diagnosis of western equine encephalitis in an infant by brain biopsy. Pediatr. Infect. Dis. J. 1986, 5, 382–383. [Google Scholar] [CrossRef] [PubMed]
  52. Lad, E.M.; Ong, S.S.; Proia, A.D. Ocular histopathology in Eastern equine encephalitis: A case report. Am. J. Ophthalmol. Case Rep. 2016, 5, 99–102. [Google Scholar] [CrossRef] [PubMed]
  53. Cain, M.D.; Salimi, H.; Gong, Y.; Yang, L.; Hamilton, S.L.; Heffernan, J.R.; Hou, J.; Miller, M.J.; Klein, R.S. Virus entry and replication in the brain precedes blood-brain barrier disruption during intranasal alphavirus infection. J. Neuroimmunol. 2017, 308, 118–130. [Google Scholar] [CrossRef] [PubMed]
  54. Hughes, H.R.; Velez, J.O.; Davis, E.H.; Laven, J.; Gould, C.V.; Panella, A.J.; Lambert, A.J.; Staples, J.E.; Brault, A.C. Fatal Human Infection with Evidence of Intrahost Variation of Eastern Equine Encephalitis Virus, Alabama, USA, 2019. Emerg. Infect. Dis. 2021, 27, 1886–1892. [Google Scholar] [CrossRef] [PubMed]
  55. Dupuy, L.C.; Reed, D.S. Nonhuman primate models of encephalitic alphavirus infection: Historical review and future perspectives. Curr. Opin. Virol. 2012, 2, 363–367. [Google Scholar] [CrossRef] [PubMed]
  56. Albe, J.R.; Ma, H.; Gilliland, T.H.; McMillen, C.M.; Gardner, C.L.; Boyles, D.A.; Cottle, E.L.; Dunn, M.D.; Lundy, J.D.; O’Malley, K.J.; et al. Physiological and immunological changes in the brain associated with lethal eastern equine encephalitis virus in macaques. PLoS Pathog. 2021, 17, e1009308. [Google Scholar] [CrossRef] [PubMed]
  57. Trefry, J.C.; Rossi, F.D.; Accardi, M.V.; Dorsey, B.L.; Sprague, T.R.; Wollen-Roberts, S.E.; Shamblin, J.D.; Kimmel, A.E.; Glass, P.J.; Miller, L.J.; et al. The utilization of advance telemetry to investigate critical physiological parameters including electroencephalography in cynomolgus macaques following aerosol challenge with eastern equine encephalitis virus. PLoS Negl. Trop. Dis. 2021, 15, e0009424. [Google Scholar] [CrossRef] [PubMed]
  58. Reed, D.S.; Lackemeyer, M.G.; Garza, N.L.; Norris, S.; Gamble, S.; Sullivan, L.J.; Lind, C.M.; Raymond, J.L. Severe Encephalitis in Cynomolgus Macaques Exposed to Aerosolized Eastern Equine Encephalitis Virus. J. Infect. Dis. 2007, 196, 441–450. [Google Scholar] [CrossRef] [PubMed]
  59. Porter, A.I.; Erwin-Cohen, R.A.; Twenhafel, N.; Chance, T.; Yee, S.B.; Kern, S.J.; Norwood, D.; Hartman, L.J.; Parker, M.D.; Glass, P.J.; et al. Characterization and pathogenesis of aerosolized eastern equine encephalitis in the common marmoset (Callithrix jacchus). Virol. J. 2017, 14, 25. [Google Scholar] [CrossRef] [PubMed]
  60. Reed, D.S.; Lind, C.M.; Sullivan, L.J.; Pratt, W.D.; Parker, M.D. Aerosol Infection of Cynomolgus Macaques with Enzootic Strains of Venezuelan Equine Encephalitis Viruses. J. Infect. Dis. 2004, 189, 1013–1017. [Google Scholar] [CrossRef] [PubMed]
  61. Burke, C.W.; Froude, J.W.; Rossi, F.; White, C.E.; Moyer, C.L.; Ennis, J.; Pitt, M.L.; Streatfield, S.; Jones, R.M.; Musiychuk, K.; et al. Therapeutic monoclonal antibody treatment protects nonhuman primates from severe Venezuelan equine encephalitis virus disease after aerosol exposure. PLoS Pathog. 2019, 15, e1008157. [Google Scholar] [CrossRef] [PubMed]
  62. Ma, H.; Lundy, J.D.; Cottle, E.L.; O’Malley, K.J.; Trichel, A.M.; Klimstra, W.B.; Hartman, A.L.; Reed, D.S.; Teichert, T. Applications of minimally invasive multimodal telemetry for continuous monitoring of brain function and intracranial pressure in macaques with acute viral encephalitis. PLoS ONE 2020, 15, e0232381. [Google Scholar] [CrossRef] [PubMed]
  63. Ma, H.; Lundy, J.D.; O’Malley, K.J.; Klimstra, W.B.; Hartman, A.L.; Reed, D.S. Electrocardiography Abnormalities in Macaques after Infection with Encephalitic Alphaviruses. Pathogens 2019, 8, 240. [Google Scholar] [CrossRef] [PubMed]
  64. Rossi, S.L.; Russell-Lodrigue, K.E.; Killeen, S.Z.; Wang, E.; Leal, G.; Bergren, N.A.; Vinet-Oliphant, H.; Weaver, S.C.; Roy, C.J. IRES-Containing VEEV Vaccine Protects Cynomolgus Macaques from IE Venezuelan Equine Encephalitis Virus Aerosol Challenge. PLoS Negl. Trop. Dis. 2015, 9, e0003797. [Google Scholar] [CrossRef] [PubMed]
  65. Monath, T.P.; Calisher, C.H.; Davis, M.; Bowen, G.S.; White, J. Experimental Studies of Rhesus Monkeys Infected with Epizootic and Enzootic Subtypes of Venezuelan Equine Encephalitis Virus. J. Infect. Dis. 1974, 129, 194–200. [Google Scholar] [CrossRef] [PubMed]
  66. Dahal, B.; Lin, S.-C.; Carey, B.D.; Jacobs, J.L.; Dinman, J.D.; van Hoek, M.L.; Adams, A.A.; Kehn-Hall, K. EGR1 upregulation following Venezuelan equine encephalitis virus infection is regulated by ERK and PERK pathways contributing to cell death. Virology 2020, 539, 121–128. [Google Scholar] [CrossRef] [PubMed]
  67. Sharma, A.; Bhattacharya, B.; Puri, R.K.; Maheshwari, R.K. Venezuelan equine encephalitis virus infection causes modulation of inflammatory and immune response genes in mouse brain. BMC Genom. 2008, 9, 289. [Google Scholar] [CrossRef] [PubMed]
  68. Reed, D.S.; Larsen, T.; Sullivan, L.J.; Lind, C.M.; Lackemeyer, M.G.; Pratt, W.D.; Parker, M.D. Aerosol Exposure to Western Equine Encephalitis Virus Causes Fever and Encephalitis in Cynomolgus Macaques. J. Infect. Dis. 2005, 192, 1173–1182. [Google Scholar] [CrossRef] [PubMed]
  69. Connolly, S.A.; Jardetzky, T.S.; Longnecker, R. The structural basis of herpesvirus entry. Nat. Rev. Microbiol. 2020, 19, 110–121. [Google Scholar] [CrossRef] [PubMed]
  70. Davison, A.J. Herpesvirus systematics. Vet. Microbiol. 2010, 143, 52–69. [Google Scholar] [CrossRef] [PubMed]
  71. Sorel, O.; Messaoudi, I. Varicella Virus-Host Interactions During Latency and Reactivation: Lessons From Simian Varicella Virus. Front. Microbiol. 2018, 9, 3170. [Google Scholar] [CrossRef] [PubMed]
  72. Haberthur, K.; Messaoudi, I. Animal models of varicella zoster virus infection. Pathogens 2013, 2, 364–382. [Google Scholar] [CrossRef] [PubMed]
  73. Herlin, L.K.; Hansen, K.S.; Bodilsen, J.; Larsen, L.; Brandt, C.; Andersen, C.Ø.; Hansen, B.R.; Lüttichau, H.R.; Helweg-Larsen, J.; Wiese, L.; et al. Varicella Zoster Virus Encephalitis in Denmark from 2015 to 2019—A Nationwide Prospective Cohort Study. Clin. Infect. Dis. 2021, 72, 1192–1199. [Google Scholar] [CrossRef] [PubMed]
  74. Ueno, H.; Hayashi, M.; Nagumo, S.; Ichikawa, K.; Aoki, N.; Ohshima, Y.; Watanabe, S.; Koya, T.; Abé, T.; Ohashi, R.; et al. Disseminated Varicella-zoster Virus Infection Causing Fatal Pneumonia in an Immunocompromised Patient with Chronic Interstitial Pneumonia. Intern. Med. 2021, 60, 1077–1082. [Google Scholar] [CrossRef] [PubMed]
  75. Lenfant, T.; L’Honneur, A.-S.; Ranque, B.; Pilmis, B.; Charlier, C.; Zuber, M.; Pouchot, J.; Rozenberg, F.; Michon, A. Neurological complications of varicella zoster virus reactivation: Prognosis, diagnosis, and treatment of 72 patients with positive PCR in the cerebrospinal fluid. Brain Behav. 2021, 12, e2455. [Google Scholar] [CrossRef] [PubMed]
  76. Cao, D.-H.; Xie, Y.-N.; Ji, Y.; Han, J.-Z.; Zhu, J.-G. A case of varicella zoster encephalitis with glossopharyngeal and vagus nerve injury as primary manifestation combined with medulla lesion. J. Int. Med. Res. 2019, 47, 2256–2261. [Google Scholar] [CrossRef] [PubMed]
  77. Arruti, M.; Piñeiro, L.D.; Salicio, Y.; Cilla, G.; Goenaga, M.A.; López de Munain, A. Incidence of varicella zoster virus infections of the central nervous system in the elderly: A large tertiary hospital-based series (2007–2014). J. NeuroVirol. 2017, 23, 451–459. [Google Scholar] [CrossRef] [PubMed]
  78. Yan, Y.; Yuan, Y.; Wang, J.; Zhang, Y.; Liu, H.; Zhang, Z. Meningitis/meningoencephalitis caused by varicella zoster virus reactivation: A retrospective single-center case series study. Am. J. Transl. Res. 2022, 14, 491–500. [Google Scholar] [PubMed]
  79. Song, Y.-X.; Li, Y.; Jiang, Y.-M.; Liu, T. Detection of varicella-zoster virus from cerebrospinal fluid using advanced fragment analysis in a child with encephalitis: A case report. BMC Infect. Dis. 2019, 19, 342. [Google Scholar] [CrossRef] [PubMed]
  80. Suzuki, T.; Tetsuka, S.; Ogawa, T.; Hashimoto, R.; Okada, S.; Kato, H. An Autopsy Case of Varicella Zoster Virus Encephalitis with Multiple Brain Lesions. Intern. Med. 2020, 59, 1643–1647. [Google Scholar] [CrossRef] [PubMed]
  81. Bakradze, E.; Kirchoff, K.F.; Antoniello, D.; Springer, M.V.; Mabie, P.C.; Esenwa, C.C.; Labovitz, D.L.; Liberman, A.L. Varicella Zoster Virus Vasculitis and Adult Cerebrovascular Disease. Neurohospitalist 2019, 9, 203–208. [Google Scholar] [CrossRef] [PubMed]
  82. Willer, D.O.; Ambagala, A.P.N.; Pilon, R.; Chan, J.K.; Fournier, J.; Brooks, J.; Sandstrom, P.; Macdonald, K.S. Experimental infection of Cynomolgus Macaques (Macaca fascicularis) with human varicella-zoster virus. J. Virol. 2012, 86, 3626–3634. [Google Scholar] [CrossRef] [PubMed]
  83. Traina-Dorge, V.; Palmer, B.E.; Coleman, C.; Hunter, M.; Frieman, A.; Gilmore, A.; Altrock, K.; Doyle-Meyers, L.; Nagel, M.A.; Mahalingam, R. Reactivation of Simian Varicella Virus in Rhesus Macaques after CD4 T Cell Depletion. J. Virol. 2019, 93, e01375–18. [Google Scholar] [CrossRef] [PubMed]
  84. Arnold, N.; Meyer, C.; Engelmann, F.; Messaoudi, I. Robust gene expression changes in the ganglia following subclinical reactivation in rhesus macaques infected with simian varicella virus. J. Neurovirol. 2017, 23, 520–538. [Google Scholar] [CrossRef] [PubMed]
  85. Depledge, D.P.; Ouwendijk, W.J.D.; Sadaoka, T.; Braspenning, S.E.; Mori, Y.; Cohrs, R.J.; Verjans, G.; Breuer, J. A spliced latency-associated VZV transcript maps antisense to the viral transactivator gene 61. Nat. Commun. 2018, 9, 1167. [Google Scholar] [CrossRef] [PubMed]
  86. Meyer, C.; Kerns, A.; Barron, A.; Kreklywich, C.; Streblow, D.N.; Messaoudi, I. Simian varicella virus gene expression during acute and latent infection of rhesus macaques. J. Neurovirol. 2011, 17, 600–612. [Google Scholar] [CrossRef] [PubMed]
  87. Meyer, C.; Kerns, A.; Haberthur, K.; Dewane, J.; Walker, J.; Gray, W.; Messaoudi, I. Attenuation of the adaptive immune response in rhesus macaques infected with simian varicella virus lacking open reading frame 61. J. Virol. 2013, 87, 2151–2163. [Google Scholar] [CrossRef] [PubMed]
  88. Whitmer, T.; Malouli, D.; Uebelhoer, L.S.; DeFilippis, V.R.; Früh, K.; Verweij, M.C. The ORF61 Protein Encoded by Simian Varicella Virus and Varicella-Zoster Virus Inhibits NF-κB Signaling by Interfering with IκBα Degradation. J. Virol. 2015, 89, 8687–8700. [Google Scholar] [CrossRef] [PubMed]
  89. Verweij, M.C.; Wellish, M.; Whitmer, T.; Malouli, D.; Lapel, M.; Jonjić, S.; Haas, J.G.; DeFilippis, V.R.; Mahalingam, R.; Früh, K. Varicella Viruses Inhibit Interferon-Stimulated JAK-STAT Signaling through Multiple Mechanisms. PLoS Pathog. 2015, 11, e1004901. [Google Scholar] [CrossRef] [PubMed]
  90. Kim, J.-M.; Park, C.-G. Intratracheal inoculation of human varicella zoster virus (VZV.; MAV strain) vaccine successfully induced VZV IgG antibodies in rhesus monkeys. Lab. Anim. Res. 2021, 37, 14. [Google Scholar] [CrossRef] [PubMed]
  91. Meyer, C.; Engelmann, F.; Arnold, N.; Krah, D.L.; ter Meulen, J.; Haberthur, K.; Dewane, J.; Messaoudi, I. Abortive intrabronchial infection of rhesus macaques with varicella-zoster virus provides partial protection against simian varicella virus challenge. J. Virol. 2015, 89, 1781–1793. [Google Scholar] [CrossRef] [PubMed]
  92. Niemeyer, C.S.; Traina-Dorge, V.; Doyle-Meyers, L.; Das, A.; Looper, J.; Mescher, T.; Feia, B.; Medina, E.; Nagel, M.A.; Mahalingam, R.; et al. Simian varicella virus infection and reactivation in rhesus macaques trigger cytokine and Aβ40/42 alterations in serum and cerebrospinal fluid. J. NeuroVirol. 2024, 30, 86–99. [Google Scholar] [CrossRef] [PubMed]
  93. Roberts, E.D.; Baskin, G.B.; Soike, K.; Gibson, S.V. Pathologic changes of experimental simian varicella (Delta herpesvirus) infection in African green monkeys (Cercopithecus aethiops). Am. J. Vet. Res. 1984, 43, 523–530. [Google Scholar] [CrossRef]
  94. Bubak, A.N.; Traina-Dorge, V.; Como, C.N.; Feia, B.; Pearce, C.M.; Doyle-Meyers, L.; Das, A.; Looper, J.; Mahalingam, R.; Nagel, M.A. Elevated serum substance P during simian varicella virus infection in rhesus macaques: Implications for chronic inflammation and adverse cerebrovascular events. J. Neurovirol. 2020, 26, 945–951. [Google Scholar] [CrossRef] [PubMed]
  95. Margolis, T.P.; Imai, Y.; Yang, L.; Vallas, V.; Krause, P.R. Herpes simplex virus type 2 (HSV-2) establishes latent infection in a different population of ganglionic neurons than HSV-1: Role of latency-associated transcripts. J. Virol. 2007, 81, 1872–1878. [Google Scholar] [CrossRef] [PubMed]
  96. Lafferty, W.E.; Coombs, R.W.; Benedetti, J.; Critchlow, C.; Corey, L. Recurrences after Oral and Genital Herpes Simplex Virus Infection. New Engl. J. Med. 1987, 316, 1444–1449. [Google Scholar] [CrossRef] [PubMed]
  97. Tognarelli, E.I.; Palomino, T.F.; Corrales, N.; Bueno, S.M.; Kalergis, A.M.; González, P.A. Herpes Simplex Virus Evasion of Early Host Antiviral Responses. Front. Cell Infect. Microbiol. 2019, 9, 127. [Google Scholar] [CrossRef] [PubMed]
  98. Goode, D.; Truong, R.; Villegas, G.; Calenda, G.; Guerra-Perez, N.; Piatak, M.; Lifson, J.D.; Blanchard, J.; Gettie, A.; Robbiani, M.; et al. HSV-2-driven increase in the expression of α4β7 correlates with increased susceptibility to vaginal SHIV(SF162P3) infection. PLoS Pathog. 2014, 10, e1004567. [Google Scholar] [CrossRef] [PubMed]
  99. Reszka, N.; Zhou, C.; Song, B.; Sodroski, J.G.; Knipe, D.M. Simian TRIM5alpha proteins reduce replication of herpes simplex virus. Virology 2010, 398, 243–250. [Google Scholar] [CrossRef] [PubMed]
  100. Kenney, J.; Rodríguez, A.; Kizima, L.; Seidor, S.; Menon, R.; Jean-Pierre, N.; Pugach, P.; Levendosky, K.; Derby, N.; Gettie, A.; et al. A modified zinc acetate gel, a potential nonantiretroviral microbicide, is safe and effective against simian-human immunodeficiency virus and herpes simplex virus 2 infection in vivo. Antimicrob. Agents Chemother. 2013, 57, 4001–4009. [Google Scholar] [CrossRef] [PubMed]
  101. Stanfield, B.A.; Pahar, B.; Chouljenko, V.N.; Veazey, R.; Kousoulas, K.G. Vaccination of rhesus macaques with the live-attenuated HSV-1 vaccine VC2 stimulates the proliferation of mucosal T cells and germinal center responses resulting in sustained production of highly neutralizing antibodies. Vaccine 2017, 35, 536–543. [Google Scholar] [CrossRef] [PubMed]
  102. Xu, X.; Feng, X.; Wang, L.; Yi, T.; Zheng, L.; Jiang, G.; Fan, S.; Liao, Y.; Feng, M.; Zhang, Y.; et al. A HSV1 mutant leads to an attenuated phenotype and induces immunity with a protective effect. PLoS Pathog. 2020, 16, e1008703. [Google Scholar] [CrossRef] [PubMed]
  103. Hunter, W.D.; Martuza, R.L.; Feigenbaum, F.; Todo, T.; Mineta, T.; Yazaki, T.; Toda, M.; Newsome, J.T.; Platenberg, R.C.; Manz, H.J.; et al. Attenuated, replication-competent herpes simplex virus type 1 mutant G207: Safety evaluation of intracerebral injection in nonhuman primates. J. Virol. 1999, 73, 6319–6326. [Google Scholar] [CrossRef] [PubMed]
  104. Awasthi, S.; Hook, L.M.; Shaw, C.E.; Pahar, B.; Stagray, J.A.; Liu, D.; Veazey, R.S.; Friedman, H.M. An HSV-2 Trivalent Vaccine Is Immunogenic in Rhesus Macaques and Highly Efficacious in Guinea Pigs. PLoS Pathog. 2017, 13, e1006141. [Google Scholar] [CrossRef] [PubMed]
  105. Ugaonkar, S.R.; Wesenberg, A.; Wilk, J.; Seidor, S.; Mizenina, O.; Kizima, L.; Rodriguez, A.; Zhang, S.; Levendosky, K.; Kenney, J.; et al. A novel intravaginal ring to prevent HIV-1, HSV-2, HPV, and unintended pregnancy. J. Control Release 2015, 213, 57–68. [Google Scholar] [CrossRef] [PubMed]
  106. Derby, N.; Aravantinou, M.; Kenney, J.; Ugaonkar, S.R.; Wesenberg, A.; Wilk, J.; Kizima, L.; Rodriguez, A.; Zhang, S.; Mizenina, O.; et al. An intravaginal ring that releases three antiviral agents and a contraceptive blocks SHIV-RT infection, reduces HSV-2 shedding, and suppresses hormonal cycling in rhesus macaques. Drug Deliv. Transl. Res. 2017, 7, 840–858. [Google Scholar] [CrossRef] [PubMed]
  107. Hutterer, C.; Milbradt, J.; Hamilton, S.; Zaja, M.; Leban, J.; Henry, C.; Vitt, D.; Steingruber, M.; Sonntag, E.; Zeitträger, I.; et al. Inhibitors of dual-specificity tyrosine phosphorylation-regulated kinases (DYRK) exert a strong anti-herpesviral activity. Antivir. Res. 2017, 143, 113–121. [Google Scholar] [CrossRef] [PubMed]
  108. Fan, S.; Xu, X.; Liao, Y.; Wang, Y.; Wang, J.; Feng, M.; Wang, L.; Zhang, Y.; He, Z.; Yang, F.; et al. Attenuated Phenotype and Immunogenic Characteristics of a Mutated Herpes Simplex Virus 1 Strain in the Rhesus Macaque. Viruses 2018, 10, 234. [Google Scholar] [CrossRef] [PubMed]
  109. Fan, S.; Cai, H.; Xu, X.; Feng, M.; Wang, L.; Liao, Y.; Zhang, Y.; He, Z.; Yang, F.; Yu, W.; et al. The Characteristics of Herpes Simplex Virus Type 1 Infection in Rhesus Macaques and the Associated Pathological Features. Viruses 2017, 9, 26. [Google Scholar] [CrossRef] [PubMed]
  110. Aravantinou, M.; Frank, I.; Arrode-Bruses, G.; Szpara, M.; Grasperge, B.; Blanchard, J.; Gettie, A.; Derby, N.; Martinelli, E. A model of genital herpes simplex virus Type 1 infection in Rhesus Macaques. J. Med. Primatol. 2017, 46, 121–128. [Google Scholar] [CrossRef] [PubMed]
  111. Aravantinou, M.; Mizenina, O.; Calenda, G.; Kenney, J.; Frank, I.; Lifson, J.D.; Szpara, M.; Jing, L.; Koelle, D.M.; Teleshova, N.; et al. Experimental Oral Herpes Simplex Virus-1 (HSV-1) Co-infection in Simian Immunodeficiency Virus (SIV)-Infected Rhesus Macaques. Front. Microbiol. 2017, 8, 2342. [Google Scholar] [CrossRef] [PubMed]
  112. London, W.T.; Nahmias, A.J.; Naib, Z.M.; Fuccillo, D.A.; Ellenberg, J.H.; Sever, J.L. A nonhuman primate model for the study of the cervical oncogenic potential of herpes simplex virus type 2. Cancer Res. 1974, 34, 1118–1121. [Google Scholar] [PubMed]
  113. Lo, M.; Zhu, J.; Hansen, S.G.; Carroll, T.; Farr Zuend, C.; Nöel-Romas, L.; Ma, Z.-M.; Fritts, L.; Huang, M.-L.; Sun, S.; et al. Acute Infection and Subsequent Subclinical Reactivation of Herpes Simplex Virus 2 after Vaginal Inoculation of Rhesus Macaques. J. Virol. 2019, 93, e01574-18. [Google Scholar] [CrossRef] [PubMed]
  114. Wang, K.; Jordan, T.; Dowdell, K.; Herbert, R.; Moore, I.N.; Koelle, D.M.; Cohen, J.I. A nonhuman primate model for genital herpes simplex virus 2 infection that results in vaginal vesicular lesions, virus shedding, and seroconversion. PLoS Pathog. 2024, 20, e1012477. [Google Scholar] [CrossRef] [PubMed]
  115. Edwards, E.E.; Birch, S.M.; Hoppes, S.M.; Keating, M.K.; Stoica, G. Pathology in Practice. J. Am. Vet. Med. Assoc. 2018, 253, 423–426. [Google Scholar] [CrossRef] [PubMed]
  116. Todo, T.; Feigenbaum, F.; Rabkin, S.D.; Lakeman, F.; Newsome, J.T.; Johnson, P.A.; Mitchell, E.; Belliveau, D.; Ostrove, J.M.; Martuza, R.L. Viral Shedding and Biodistribution of G207, a MuItimutated, Conditionally Replicating Herpes Simplex Virus Type 1, after Intracerebral Inoculation in Aotus. Mol. Ther. 2000, 2, 588–595. [Google Scholar] [CrossRef] [PubMed]
  117. Roth, J.C.; Cassady, K.A.; Cody, J.J.; Parker, J.N.; Price, K.H.; Coleman, J.M.; Peggins, J.O.; Noker, P.E.; Powers, N.W.; Grimes, S.D.; et al. Evaluation of the safety and biodistribution of M032, an attenuated herpes simplex virus type 1 expressing hIL-12, after intracerebral administration to aotus nonhuman primates. Hum. Gene Ther. Clin. Dev. 2014, 25, 16–27. [Google Scholar] [CrossRef] [PubMed]
  118. Costa, É.A.; Luppi, M.M.; de Campos Cordeiro Malta, M.; Luiz, A.P.M.F.; de Araujo, M.R.; Coelho, F.M.; Fonseca, F.G.d.; Ecco, R.; Resende, M. Outbreak of Human Herpesvirus Type 1 Infection in Nonhuman Primates (Callithrix penincillata). J. Wildl. Dis. 2011, 47, 690–693. [Google Scholar] [CrossRef] [PubMed]
  119. Imura, K.; Chambers, J.K.; Uchida, K.; Nomura, S.; Suzuki, S.; Nakayama, H.; Miwa, Y. Herpes simplex virus type 1 infection in two pet marmosets in Japan. J. Vet. Med. Sci. 2014, 76, 1667–1670. [Google Scholar] [CrossRef] [PubMed]
  120. Longa, C.S.; Bruno, S.F.; Pires, A.R.; Romijn, P.C.; Kimura, L.S.; Costa, C.H.C. Human herpesvirus 1 in wild marmosets, Brazil, 2008. Emerg. Infect. Dis. 2011, 17, 1308–1310. [Google Scholar] [CrossRef] [PubMed]
  121. Griffiths, P.; Reeves, M. Pathogenesis of human cytomegalovirus in the immunocompromised host. Nat. Rev. Microbiol. 2021, 19, 759–773. [Google Scholar] [CrossRef] [PubMed]
  122. Maltezou, P.-G.; Kourlaba, G.; Kourkouni, Ε.; Luck, S.; Blázquez-Gamero, D.; Ville, Y.; Lilleri, D.; Dimopoulou, D.; Karalexi, M.; Papaevangelou, V. Maternal type of CMV infection and sequelae in infants with congenital CMV: Systematic review and meta-analysis. J. Clin. Virol. 2020, 129, 104518. [Google Scholar] [CrossRef] [PubMed]
  123. Fowler, K.B.; Boppana, S.B. Congenital cytomegalovirus infection. Semin. Perinatol. 2018, 42, 149–154. [Google Scholar] [CrossRef] [PubMed]
  124. Itell, H.L.; Kaur, A.; Deere, J.D.; Barry, P.A.; Permar, S.R. Rhesus monkeys for a nonhuman primate model of cytomegalovirus infections. Curr. Opin. Virol. 2017, 25, 126–133. [Google Scholar] [CrossRef] [PubMed]
  125. Burwitz, B.J.; Malouli, D.; Bimber, B.N.; Reed, J.S.; Ventura, A.B.; Hancock, M.H.; Uebelhoer, L.S.; Bhusari, A.; Hammond, K.B.; Espinosa Trethewy, R.G.; et al. Cross-Species Rhesus Cytomegalovirus Infection of Cynomolgus Macaques. PLoS Pathog. 2016, 12, e1006014. [Google Scholar] [CrossRef] [PubMed]
  126. Marsh, A.K.; Ambagala, A.P.; Perciani, C.T.; Russell, J.N.H.; Chan, J.K.; Janes, M.; Antony, J.M.; Pilon, R.; Sandstrom, P.; Willer, D.O.; et al. Examining the species-specificity of rhesus macaque cytomegalovirus (RhCMV) in cynomolgus macaques. PLoS ONE 2015, 10, e0121339. [Google Scholar] [CrossRef] [PubMed]
  127. Malouli, D.; Nakayasu, E.S.; Viswanathan, K.; Camp, D.G., 2nd; Chang, W.L.W.; Barry, P.A.; Smith, R.D.; Früh, K. Reevaluation of the coding potential and proteomic analysis of the BAC-derived rhesus cytomegalovirus strain 68-1. J. Virol. 2012, 86, 8959–8973. [Google Scholar] [CrossRef] [PubMed]
  128. Abel, K.; Martinez, J.; Yue, Y.; Lacey, S.F.; Wang, Z.; Strelow, L.; Dasgupta, A.; Li, Z.; Schmidt, K.A.; Oxford, K.L.; et al. Vaccine-induced control of viral shedding following rhesus cytomegalovirus challenge in rhesus macaques. J. Virol. 2011, 85, 2878–2890. [Google Scholar] [CrossRef] [PubMed]
  129. Powers, C.; Früh, K. Rhesus CMV: An emerging animal model for human CMV. Med. Microbiol. Immunol. 2008, 197, 109–115. [Google Scholar] [CrossRef] [PubMed]
  130. Lockridge, K.M.; Sequar, G.; Zhou, S.S.; Yue, Y.; Mandell, C.P.; Barry, P.A. Pathogenesis of experimental rhesus cytomegalovirus infection. J. Virol. 1999, 73, 9576–9583. [Google Scholar] [CrossRef] [PubMed]
  131. Baskin, G.B. Disseminated cytomegalovirus infection in immunodeficient rhesus monkeys. Am. J. Pathol. 1987, 129, 345–352. [Google Scholar] [PubMed]
  132. Roark, H.K.; Jenks, J.A.; Permar, S.R.; Schleiss, M.R. Animal Models of Congenital Cytomegalovirus Transmission: Implications for Vaccine Development. J. Infect. Dis. 2020, 221, S60–S73. [Google Scholar] [CrossRef] [PubMed]
  133. Bialas, K.M.; Tanaka, T.; Tran, D.; Varner, V.; Cisneros De La Rosa, E.; Chiuppesi, F.; Wussow, F.; Kattenhorn, L.; Macri, S.; Kunz, E.L.; et al. Maternal CD4+ T cells protect against severe congenital cytomegalovirus disease in a novel nonhuman primate model of placental cytomegalovirus transmission. Proc. Natl. Acad. Sci. USA 2015, 112, 13645–13650. [Google Scholar] [CrossRef] [PubMed]
  134. Fan, Q.; Nelson, C.S.; Bialas, K.M.; Chiuppesi, F.; Amos, J.; Gurley, T.C.; Marshall, D.J.; Eudailey, J.; Heimsath, H.; Himes, J.; et al. Plasmablast Response to Primary Rhesus Cytomegalovirus (CMV) Infection in a Monkey Model of Congenital CMV Transmission. Clin. Vaccine Immunol. 2017, 24, e00510–e00516. [Google Scholar] [CrossRef] [PubMed]
  135. Whitney, J.B.; Hill, A.L.; Sanisetty, S.; Penaloza-MacMaster, P.; Liu, J.; Shetty, M.; Parenteau, L.; Cabral, C.; Shields, J.; Blackmore, S.; et al. Rapid seeding of the viral reservoir prior to SIV viraemia in rhesus monkeys. Nature 2014, 512, 74–77. [Google Scholar] [CrossRef] [PubMed]
  136. Fukazawa, Y.; Lum, R.; Okoye, A.A.; Park, H.; Matsuda, K.; Bae, J.Y.; Hagen, S.I.; Shoemaker, R.; Deleage, C.; Lucero, C.; et al. B cell follicle sanctuary permits persistent productive simian immunodeficiency virus infection in elite controllers. Nat. Med. 2015, 21, 132–139. [Google Scholar] [CrossRef] [PubMed]
  137. Cadena, A.M.; Ventura, J.D.; Abbink, P.; Borducchi, E.N.; Tuyishime, H.; Mercado, N.B.; Walker-Sperling, V.; Siamatu, M.; Liu, P.T.; Chandrashekar, A.; et al. Persistence of viral RNA in lymph nodes in ART-suppressed SIV/SHIV-infected Rhesus Macaques. Nat. Commun. 2021, 12, 1474. [Google Scholar] [CrossRef] [PubMed]
  138. Abreu, C.; Veenhuis, R.; Avalos, C.; Graham, S.; Parrilla, D.; Ferreira, E.; Queen, S.; Shirk, E.; Bullock, B.; Li, M.; et al. Myeloid and CD4 T Cells Comprise the Latent Reservoir in Antiretroviral Therapy-Suppressed SIVmac251-Infected Macaques. mBio 2019, 10, e01659-19. [Google Scholar] [CrossRef] [PubMed]
  139. Clements, J.; Li, M.; Gama, L.; Bullock, B.; Carruth, L.; Mankowski, J.; Zink, M. The central nervous system is a viral reservoir in simian immunodeficiency virus–infected macaques on combined antiretroviral therapy: A model for human immunodeficiency virus patients on highly active antiretroviral theraby. J. NeuroVirol. 2005, 11, 180–189. [Google Scholar] [CrossRef] [PubMed]
  140. Perez, S.; Johnson, A.M.; Xiang, S.H.; Li, J.; Foley, B.T.; Doyle-Meyers, L.; Panganiban, A.; Kaur, A.; Veazey, R.S.; Wu, Y.; et al. Persistence of SIV in the brain of SIV-infected Chinese rhesus macaques with or without antiretroviral therapy. J. Neurovirol. 2018, 24, 62–74. [Google Scholar] [CrossRef] [PubMed]
  141. Avalos, C.; Abreu, C.; Queen, S.; Li, M.; Price, S.; Shirk, E.; Engle, E.; Forsyth, E.; Bullock, B.; Mac Gabhann, F.; et al. Brain Macrophages in Simian Immunodeficiency Virus-Infected, Antiretroviral-Suppressed Macaques: A Functional Latent Reservoir. mBio 2017, 8, e01186-17. [Google Scholar] [CrossRef] [PubMed]
  142. Mohammadzadeh, N.; Roda, W.; Branton, W.; Clain, J.; Rabezanahary, H.; Zghidi-Abouzid, O.; Gelman, B.; Angel, J.; Cohen, E.; Gill, M.; et al. Lentiviral Infections Persist in Brain despite Effective Antiretroviral Therapy and Neuroimmune Activation. mBio 2021, 12, e0278421. [Google Scholar] [CrossRef] [PubMed]
  143. Tavazzi, E.; Morrison, D.; Sullivan, P.; Morgello, S.; Fischer, T. Brain inflammation is a common feature of HIV-infected patients without HIV encephalitis or productive brain infection. Curr. HIV Res. 2014, 12, 97–110. [Google Scholar] [CrossRef] [PubMed]
  144. Cysique, L.; Jugé, L.; Gates, T.; Tobia, M.; Moffat, K.; Brew, B.; Rae, C. Covertly active and progressing neurochemical abnormalities in suppressed HIV infection. Neurol. R Neuroimmunol. Neuroinflamm. 2018, 5, e430. [Google Scholar] [CrossRef] [PubMed]
  145. Vera, J.H.; Guo, Q.; Cole, J.H.; Boasso, A.; Greathead, L.; Kelleher, P.; Rabiner, E.A.; Kalk, N.; Bishop, C.; Gunn, R.N.; et al. Neuroinflammation in treated HIV-positive individuals: A TSPO PET study. Neurology 2016, 86, 1425–1432. [Google Scholar] [CrossRef] [PubMed]
  146. Garvey, L.; Pavese, N.; Politis, M.; Ramlackhansingh, A.; Brooks, D.; Taylor-Robinson, S.; Winston, A. Increased microglia activation in neurologically asymptomatic HIV-infected patients receiving effective ART. AIDS 2014, 28, 67–72. [Google Scholar] [CrossRef] [PubMed]
  147. Solis-Leal, A.; Siddiqui, S.; Wu, F.; Mohan, M.; Hu, W.; Doyle-Meyers, L.; Dufour, J.; Ling, B. Neuroinflammatory Profiling in SIV-Infected Chinese-Origin Rhesus Macaques on Antiretroviral Therapy. Viruses 2022, 14, 139. [Google Scholar] [CrossRef] [PubMed]
  148. Macneughton, M.; Davies, H. Ribonucleoprotein-like structures from coronavirus particles. J. Gen. Virol. 1978, 39, 545–549. [Google Scholar] [CrossRef] [PubMed]
  149. Sturman, L.; Holmes, K.; Behnke, J. Isolation of coronavirus envelope glycoproteins and interaction with the viral nucleocapsid. J. Virol. 1980, 33, 449–462. [Google Scholar] [CrossRef] [PubMed]
  150. Lai, M.; Cavanagh, D. The molecular biology of coronaviruses. Adv. Virus Res. 1997, 48, 1–100. [Google Scholar] [CrossRef] [PubMed]
  151. Hoffmann, M.; Kleine-Weber, H.; Schroeder, S.; Krüger, N.; Herrler, T.; Erichsen, S.; Schiergens, T.; Herrler, G.; Wu, N.; Nitsche, A.; et al. SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor. Cell 2020, 181, 271–280.e278. [Google Scholar] [CrossRef] [PubMed]
  152. Chandrashekar, A.; Liu, J.; Martinot, A.J.; McMahan, K.; Mercado, N.B.; Peter, L.; Tostanoski, L.H.; Yu, J.; Maliga, Z.; Nekorchuk, M.; et al. SARS-CoV-2 infection protects against rechallenge in rhesus macaques. Science 2020, 369, 812–817. [Google Scholar] [CrossRef] [PubMed]
  153. Melin, A.D.; Janiak, M.C.; Marrone, F., 3rd; Arora, P.S.; Higham, J.P. Comparative ACE2 variation and primate COVID-19 risk. Commun. Biol. 2020, 3, 641. [Google Scholar] [CrossRef] [PubMed]
  154. Yang, J.; Wang, W.; Chen, Z.; Lu, S.; Yang, F.; Bi, Z.; Bao, L.; Mo, F.; Li, X.; Huang, Y.; et al. A vaccine targeting the RBD of the S protein of SARS-CoV-2 induces protective immunity. Nature 2020, 586, 572–577. [Google Scholar] [CrossRef] [PubMed]
  155. Baum, A.; Ajithdoss, D.; Copin, R.; Zhou, A.; Lanza, K.; Negron, N.; Ni, M.; Wei, Y.; Mohammadi, K.; Musser, B.; et al. REGN-COV2 antibodies prevent and treat SARS-CoV-2 infection in rhesus macaques and hamsters. Science 2020, 370, 1110–1115. [Google Scholar] [CrossRef] [PubMed]
  156. Gao, Q.; Bao, L.; Mao, H.; Wang, L.; Xu, K.; Yang, M.; Li, Y.; Zhu, L.; Wang, N.; Lv, Z.; et al. Development of an inactivated vaccine candidate for SARS-CoV-2. Science 2020, 369, 77–81. [Google Scholar] [CrossRef] [PubMed]
  157. Blair, R.V.; Vaccari, M.; Doyle-Meyers, L.A.; Roy, C.J.; Russell-Lodrigue, K.; Fahlberg, M.; Monjure, C.J.; Beddingfield, B.; Plante, K.S.; Plante, J.A.; et al. Acute Respiratory Distress in Aged, SARS-CoV-2-Infected African Green Monkeys but Not Rhesus Macaques. Am. J. Pathol. 2021, 191, 274–282. [Google Scholar] [CrossRef] [PubMed]
  158. Maity, S.; Mayer, M.G.; Shu, Q.; Linh, H.; Bao, D.; Blair, R.V.; He, Y.; Lyon, C.J.; Hu, T.Y.; Fischer, T.; et al. Cerebrospinal Fluid Protein Markers Indicate Neuro-Damage in SARS-CoV-2-Infected Nonhuman Primates. Mol. Cell Proteom. 2023, 22, 100523. [Google Scholar] [CrossRef] [PubMed]
  159. Heming, M.; Li, X.; Rauber, S.; Mausberg, A.K.; Borsch, A.L.; Hartlehnert, M.; Singhal, A.; Lu, I.N.; Fleischer, M.; Szepanowski, F.; et al. Neurological Manifestations of COVID-19 Feature T Cell Exhaustion and Dedifferentiated Monocytes in Cerebrospinal Fluid. Immunity 2021, 54, 164–175.e6. [Google Scholar] [CrossRef] [PubMed]
  160. Rutkai, I.; Mayer, M.G.; Hellmers, L.M.; Ning, B.; Huang, Z.; Monjure, C.J.; Coyne, C.; Silvestri, R.; Golden, N.; Hensley, K.; et al. Neuropathology and virus in brain of SARS-CoV-2 infected non-human primates. Nat. Commun. 2022, 13, 1745. [Google Scholar] [CrossRef] [PubMed]
  161. Napolitano, A.; Arrigoni, A.; Caroli, A.; Cava, M.; Remuzzi, A.; Longhi, L.G.; Barletta, A.; Zangari, R.; Lorini, F.L.; Sessa, M.; et al. Cerebral Microbleeds Assessment and Quantification in COVID-19 Patients With Neurological Manifestations. Front. Neurol. 2022, 13, 884449. [Google Scholar] [CrossRef] [PubMed]
  162. Etter, M.M.; Martins, T.A.; Kulsvehagen, L.; Possnecker, E.; Duchemin, W.; Hogan, S.; Sanabria-Diaz, G.; Muller, J.; Chiappini, A.; Rychen, J.; et al. Severe Neuro-COVID is associated with peripheral immune signatures, autoimmunity and neurodegeneration: A prospective cross-sectional study. Nat. Commun. 2022, 13, 6777. [Google Scholar] [CrossRef] [PubMed]
  163. Lindskog, C.; Mear, L.; Virhammar, J.; Fallmar, D.; Kumlien, E.; Hesselager, G.; Casar-Borota, O.; Rostami, E. Protein Expression Profile of ACE2 in the Normal and COVID-19-Affected Human Brain. J. Proteome Res. 2022, 21, 2137–2145. [Google Scholar] [CrossRef] [PubMed]
  164. Gao, C.C.; Li, M.; Deng, W.; Ma, C.H.; Chen, Y.S.; Sun, Y.Q.; Du, T.; Liu, Q.L.; Li, W.J.; Zhang, B.; et al. Differential transcriptomic landscapes of multiple organs from SARS-CoV-2 early infected rhesus macaques. Protein Cell 2022, 13, 920–939. [Google Scholar] [CrossRef] [PubMed]
  165. Meinhardt, J.; Radke, J.; Dittmayer, C.; Franz, J.; Thomas, C.; Mothes, R.; Laue, M.; Schneider, J.; Brunink, S.; Greuel, S.; et al. Olfactory transmucosal SARS-CoV-2 invasion as a port of central nervous system entry in individuals with COVID-19. Nat. Neurosci. 2020, 24, 168–175. [Google Scholar] [CrossRef] [PubMed]
  166. Beckman, D.; Bonillas, A.; Diniz, G.B.; Ott, S.; Roh, J.W.; Elizaldi, S.R.; Schmidt, B.A.; Sammak, R.L.; Van Rompay, K.K.A.; Iyer, S.S.; et al. SARS-CoV-2 infects neurons and induces neuroinflammation in a non-human primate model of COVID-19. Cell Rep. 2022, 41, 111573. [Google Scholar] [CrossRef] [PubMed]
  167. de Melo, G.D.; Lazarini, F.; Levallois, S.; Hautefort, C.; Michel, V.; Larrous, F.; Verillaud, B.; Aparicio, C.; Wagner, S.; Gheusi, G.; et al. COVID-19-related anosmia is associated with viral persistence and inflammation in human olfactory epithelium and brain infection in hamsters. Sci. Transl. Med. 2021, 13, eabf8396. [Google Scholar] [CrossRef] [PubMed]
  168. Chen, R.; Wang, K.; Yu, J.; Howard, D.; French, L.; Chen, Z.; Wen, C.; Xu, Z. The Spatial and Cell-Type Distribution of SARS-CoV-2 Receptor ACE2 in the Human and Mouse Brains. Front. Neurol. 2020, 11, 573095. [Google Scholar] [CrossRef] [PubMed]
  169. Lu, Y.; Li, X.; Geng, D.; Mei, N.; Wu, P.Y.; Huang, C.C.; Jia, T.; Zhao, Y.; Wang, D.; Xiao, A.; et al. Cerebral Micro-Structural Changes in COVID-19 Patients—An MRI-based 3-month Follow-up Study. EClinicalMedicine 2020, 25, 100484. [Google Scholar] [CrossRef] [PubMed]
  170. De Tanti, A.; Conforti, J.; Bruni, S.; De Gaetano, K.; Cappalli, A.; Basagni, B.; Bertoni, D.; Saviola, D. Cognitive and psychological outcomes and follow-up in severely affected COVID-19 survivors admitted to a rehabilitation hospital. Neurol. Sci. 2023, 44, 1481–1489. [Google Scholar] [CrossRef] [PubMed]
  171. Ma, Q.; Ma, W.; Song, T.Z.; Wu, Z.; Liu, Z.; Hu, Z.; Han, J.B.; Xu, L.; Zeng, B.; Wang, B.; et al. Single-nucleus transcriptomic profiling of multiple organs in a rhesus macaque model of SARS-CoV-2 infection. Zool. Res. 2022, 43, 1041–1062. [Google Scholar] [CrossRef] [PubMed]
Table 1. NHP models of CNS-associated viral disease.
Table 1. NHP models of CNS-associated viral disease.
VirusNeurotropismAdvantages of NHP ModelDisadvantages of NHP Model
Zika virusDirectly neurotropicRecapitulates human clinical diseaseMicrocephaly not observed in RM
West Nile virusDirectly neurotropicMacaques are naturally susceptibleMost infections are subclinical
Eastern equine encephalitis virusDirectly neurotropicAerosol infection modelMost infections are subclinical
Venezuelan equine encephalitis virusDirectly neurotropicRecapitulates human disease symptoms
Western equine encephalitis virusDirectly neurotropicAerosol infection modelLimited studies
Varicella -zoster virusDirectly neurotropicRecapitulates human disease, including latencyLimited CNS disease
Herpes simplex virus-1Directly neurotropicLatency modelingLimited CNS disease in macaques
Herpes simplex virus-2 (HSV-2)Directly neurotropicHIV co-infection modelsMacaques are refractory to HSV-2
Cytomegalovirus (CMV)Directly neurotropicStructural and genetic similarities between HCMV and RhCMVInfections often subclinical, limited seronegative colonies
HIVIndirectly neuropathogenicVirus homology with SIV, recapitulates human clinical disease progressionLimited to Asian-origin macaques
SARS-CoV-2Indirectly neuropathogenicNaturally susceptible and recapitulate human disease progression including neurological complications and long COVID-19
Abbreviations: NHP, non-human primate; RM, rhesus macaque; CNS, central nervous system; HIV, human immunodeficiency virus; SIV, simian immunodeficiency virus; SARS-CoV-2, severe acute respiratory syndrome coronavirus; COVID-19, coronavirus disease 2019.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Vail, K.J.; Macha, B.N.; Hellmers, L.; Fischer, T. Modeling Virus-Associated Central Nervous System Disease in Non-Human Primates. Int. J. Mol. Sci. 2025, 26, 6886. https://doi.org/10.3390/ijms26146886

AMA Style

Vail KJ, Macha BN, Hellmers L, Fischer T. Modeling Virus-Associated Central Nervous System Disease in Non-Human Primates. International Journal of Molecular Sciences. 2025; 26(14):6886. https://doi.org/10.3390/ijms26146886

Chicago/Turabian Style

Vail, Krystal J., Brittany N. Macha, Linh Hellmers, and Tracy Fischer. 2025. "Modeling Virus-Associated Central Nervous System Disease in Non-Human Primates" International Journal of Molecular Sciences 26, no. 14: 6886. https://doi.org/10.3390/ijms26146886

APA Style

Vail, K. J., Macha, B. N., Hellmers, L., & Fischer, T. (2025). Modeling Virus-Associated Central Nervous System Disease in Non-Human Primates. International Journal of Molecular Sciences, 26(14), 6886. https://doi.org/10.3390/ijms26146886

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop