Purpose of the review
Persistent infections capable of causing central nervous system (CNS) complications months or years after the initial infection represent a major public health concern. This concern is particularly relevant considering the ongoing coronavirus disease 2019 pandemic, where the long-term neurological effects are still being recognized.
Recent findings
Viral infections are a risk factor for the development of neurodegenerative diseases. In this paper, we provide an in-depth exploration of the prevalent known and suspected persistent pathogens and their epidemiological and mechanistic links to later development of CNS disease. We examine the pathogenic mechanisms involved, including direct viral damage and indirect immune dysregulation, while also addressing the challenges associated with detecting persistent pathogens.
Summary
Viral encephalitis has been closely associated with the later development of neurodegenerative diseases and persistent viral infections of the CNS can result in severe and debilitating symptoms. Further, persistent infections may result in the development of autoreactive lymphocytes and autoimmune mediated tissue damage. Diagnosis of persistent viral infections of the CNS remains challenging and treatment options are limited. The development of additional testing modalities as well as novel antiviral agents and vaccines against these persistent infections remains a crucial research goal.
Keywords: autoimmune, encephalitis, neurological impairments, persistent infections, post-acute sequelae of SARS CoV-2 infection
INTRODUCTION
The central nervous system (CNS) is comprised of long-lived cells and is capable of modulating inflammatory processes [1] providing an ideal location in which pathogens can persist [2]. However, not all pathogens require presence in the CNS to cause neurologic disease. Emerging evidence from the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic (COVID19) has reinforced the concept that infections, irrespective of their anatomical location, can contribute to neurological disease in both acute and late phases [3].
Delayed onset of neurological issues due to infections can occur months or years after the initial infectious insult as highlighted by the recent striking finding that viral encephalitis is associated with significant risk of later development of dementia [4▪▪] and that vaccination against some viruses can decrease the risk of developing dementia [5]. However, several barriers exist in distinguishing if the neurological symptoms are due to persistent infectious agents or postinfectious inflammatory sequelae. First, persistent infections, especially those in the CNS, are often underdiagnosed as they are not readily detected by conventional tests. Clinically, these may account for a significant proportion of the approximately 40–50% of cases of encephalitis that have no identifiable cause [6–9]. Second, during persistent infections there is ongoing inflammation which can mimic autoimmune or autoinflammatory processes leading to misdiagnosis of autoimmune encephalitis [10▪]. However, both persistent infections and dysbiosis can also influence the development of autoimmune diseases, including CNS autoimmune diseases, through several different mechanisms including molecular mimicry and bystander activation [11–15]. It is important to understand the molecular mechanisms driving associations between persistent infections and neuro-immunologic diseases. Defining their overlap will allow for the development of diagnostic tools to help distinguish these processes.
Box 1.
no caption available
INFECTIONS ASSOCIATED WITH LATE CENTRAL NERVOUS SYSTEM DISEASE
Multiple pathogens can cause pathology years after acute infection via direct damage through viral-induced cell death and dysfunction and indirectly by triggering immune-mediated damage (recently reviewed in [16,17]). The clinical phenotypes resulting from these processes are often distinct from the pathologies observed during acute infection. In this section we briefly review some of the known and suspected persistent infections associated with later development of CNS disease.
Human immunodeficiency virus
Human immunodeficiency virus (HIV) penetrates the CNS early during acute infection and infects microglia, macrophages, and to a lesser extent astrocytes, which can result in a macrophage-mediated encephalitis [18,19]. Clinically this is associated with a rapidly progressing subacute subcortical dementia [20] which is treatable with antiretroviral therapies (ART). However, even with adequate ART and suppression of virus both in the periphery and the CNS, HIV can cause CNS disease years after initial infection, collectively known as HIV-associated neurocognitive disorders (HAND). One risk factor for HAND is severe immune depletion during acute infection. This may be due to the relationship between lower CD4+ T-cell nadirs and a larger viral reservoir and more residual viral activity [21]. This larger and more active viral presence can lead to CSF viral escape and T-cell mediated encephalitis, which presents clinically with headache, tremors, cognitive impairment, confusion, focal neurologic deficits, and seizures [20]. However, even in the absence of viral replication, expression of viral proteins can lead to CNS disease. Viral proteins, such as Tat, gp120, and Nef, are produced despite ART, even from replication defective virus [22–24]. These proteins are toxic and alter neuronal and glial cell functioning (reviewed in [25–27]). Patients present with varying degrees of cognitive and psychomotor impairments that slowly progress over time, a spectrum disorder collectively known as HIV-protein associated encephalopathy [20]. As ART does not impact the production of viral proteins from integrated virus, the development of new therapies to inhibit viral transcription and translation are needed.
Herpes viruses
Multiple human herpes viruses can infect the CNS and acutely cause encephalitis. These viruses also establish persistent infections from which they can reemerge and cause viral shedding with or without recrudescence of disease [28–31]. Repeat reemergence of virus is associated with CNS pathology and the development of cognitive impairment in animal models [32]. However, even during latency herpesviruses express viral genes which can contribute to immune activation [31,33]. These viral-driven processes may contribute to the development of CNS diseases not associated with acute infection including multiple sclerosis [34▪] and dementia [32,35]. Recent work suggests that these viruses also impact the development of CNS complications associated with other infections. For example, reactivation of Epstein-Barr virus (EBV) is associated with the development of post-acute sequelae of COVID-19 (PASC) [36]. Mechanistically herpesviruses are thought to initiate the formation of Aβ plaques [37–39] through stimulation of the innate immune response as well as drive the expansion of autoreactive T and B cells [40,41▪▪]. However, data from epidemiological, pathology, and animal and in vitro models both support and refute the roles of herpesviruses in late-stage CNS disease [38,42–45].
Flaviviruses
Flaviviruses are typically vector-borne although sexual, transplacental, and transfusion transmission have all been documented [46,47]. With a world-wide distribution, a high rate of infection, and the ability to cause significant morbidity and mortality, this group of viruses are of great medical importance [48]. Flaviviruses such as Zika virus [49,50], Dengue virus [51–53], West Nile virus [54], and Chikungunya virus [55] have all been associated with CNS disease years after initial infection. In animal models, T cells post-viral clearance have been demonstrated to contribute to synaptic loss, neuronal apoptosis, and cognitive impairments [56]. These brain resident T cells, mostly CD8+ cells, produce interferon-γ, which triggers microglial cell activation. This, in turn, activates astrocytes, leading to glial cell dysfunction and specific patterns of neuronal and synaptic loss. The T cell is the main driver in this process, as cognitive impairments were prevented when CD8+ T-cells or microglial interferon-γ receptor levels were reduced after viral infection [56]. This ongoing T-cell induced damage to neurons following viral infection might explain the correlation between viral infections and the later onset of neurodegenerative processes [57].
Additionally, several flaviviruses are suggested to be present months or years after acute infection, although data demonstrating viral persistence from humans are limited. Preliminary evidence from animal models and case reports suggests that flaviviruses could reside in the CNS for extended times and are associated with progressive cognitive impairments [54,58–62]. A significant proportion, up to 80%, of flavivirus infections are asymptomatic [48]. This poses a challenge as the subsequent development of cognitive impairment may not be linked to a prior infection. To fully comprehend the long-term neurological effects of these viruses, it is imperative that future research investigates the frequency and persistence of flavivirus CNS infections.
Ebola
The recent outbreaks of Ebola have revealed that the majority of survivors have significant sequelae [63–65], including neurological impairments such as vocal outbursts, cognitive deficits, hallucinations, numbness, and fatigue [64,66,67]. In some patients this may be due to persistent virus [68]. Ebola can infect the CNS and can reemerge months to years later to cause significant pathology [69,70,71▪]. However, how often this occurs or for how long the virus endures in the CNS are unknown.
Measles
Measles virus infection can lead to subacute sclerosing panencephalitis (SSPE), a fatal CNS complication which typically occurs 6 to 10 years after infection [72]. A mutated fusion protein allows for cell-to-cell spread within the CNS with significant viral damage resulting in cognitive decline, behavioral changes, motor dysfunction, and seizures [73]. Patients progress quickly to coma and death. As vaccinations have declined, a rise in SSPE has been observed [74▪,75] and is predicted to increase over the coming years. SSPE occurs at an increased rate, with a shorter incubation period and a more fulminant form in children who acquire measles prior to two years of age [75–78].
Influenza
Influenza viruses can cause encephalitis and encephalopathy and are associated with significant levels of mortality and enduring neurological deficits [79–82]. Recent work demonstrated a striking association between influenza infections and later development of Alzheimer's disease, amyotrophic lateral sclerosis, generalized dementia, vascular dementia, and Parkinson's disease [4▪▪]. As influenza is not thought to persist in immunocompetent hosts, this association may be due to damage sustained during acute infection. For example, influenza replication in neurons can lead to α-synuclein aggregation [83], which may set the stage for later Parkinson's disease development. However, an alternative hypothesis is that influenza, much like SARS-CoV-2, could remain persistent in a small fraction of cases. Although there are conflicting reports in the literature [84], influenza A was detected in macrophages in the CNS of patients with Parkinson's disease postmortem [85], although if this was a recently acquired or chronic infection remains unknown.
Severe acute respiratory syndrome coronavirus 2
Ongoing investigations have revealed that a subset of patients may have persistent SARS-CoV-2 infections. Biopsy and postmortem studies have demonstrated viral presence months after initial infection in the small intestine, heart, lung, and brain [86▪▪,87–90] and continuous affinity maturation of immune responses, which require viral proteins, have been detected 6 to 14 months after infection [88,91,92].
This viral persistence is suggested to be one of the causes of PASC [93]. PASC is part of a spectrum of disorders known as post-acute infection syndromes [94] and shares some clinical features with myalgic encephalomyelitis (ME)/chronic fatigue syndrome [94]. One of the chief complaints among people with PASC is fatigue, a complex and poorly understood symptom associated with activation of the immune response which can occur in both autoimmune and infectious processes [94]. Patients with PASC also develop several other neurological complications including cognitive impairments, anosmia, myalgia, and autonomic dysfunction [95,96].
In patients with PASC, there are increased levels of both viral RNA and the spike protein as compared to patients with past COVID infection without PASC [97▪▪,98▪]. Viral RNA and spike protein levels increased from acute infection levels in patients who developed PASC. This is in notable contrast to patients who fully recover from COVID in which both viral protein and RNA significantly decreased as compared to acute infection [97▪▪]. Collectively these data, along with case reports [99–101], suggest that in some patients with PASC persistent viral infection is occurring, and that they may benefit from combination antiviral therapies. Although the exact causes of PASC are likely complex and multifactorial [93,95], a deeper exploration of the subgroup of PASC patients with indications of persistent infection could offer crucial insights not only for those afflicted by PASC, but also for individuals experiencing other variants of post-acute infection syndromes.
PERSISTENT INFECTIONS AND AUTOIMMUNE PROCESSES
Infections are recognized as a susceptibility factor for autoimmune diseases, including autoimmune diseases of the CNS. For example, herpes simplex virus (HSV) infections are associated with the development of N-methyl-D-aspartate receptor-encephalitis [14,15]. Initially, it was believed that CNS-specific self-reactive T-cells remained ignorant, as antigens from the CNS were thought to be sequestered. However, this notion has been challenged following the rediscovery of the lymphatic system in the CNS [102] and the identification of deep cervical lymph nodes as the site for CNS antigen presentation [103]. Indeed, T-cells activated at the cervical lymph nodes can develop tropism for the CNS. These brain resident cells express less perforin and granzyme B and higher levels of inhibitory molecules programmed cell death protein 1 and cytotoxic T-lymphocyte associated protein 4 as compared to cells in the periphery [104]. Although the barrier for overcoming inhibition is quite high, upon antigen stimulation and recognition, these cells do produce effector molecules efficiently. Furthermore, brain resident T-cells can reactivate via cytokine signaling without stimulation at the lymph node [105,106▪▪] and can be both protective and detrimental in the CNS (reviewed in [107]).
Persistent infections can improve the immune response to the pathogen by driving continued affinity maturation and refinement of antibody responses. However, this process can also result in autoimmunity. A recent example of this was described in the context of MS where continued affinity maturation of antibodies against the EBV protein EBV nuclear antigen 1 (EBNA-1) generated antibodies that cross-reacted with a host protein GlialCAM, a glial cell adhesion molecule [106▪▪]. Given this finding, in combination with another recent large study which strongly supports the involvement of EBV in the pathophysiology of MS [34▪], prevention of MS could potentially be achieve through vaccination. EBV vaccine trials are currently underway.
While autoreactive GlialCAM antibodies may contribute to pathology, other autoreactive antibodies may not. During CNS infections lymphocytes entering the CNS may undergo clonal expansion, driven in part due to ample secondary signaling from the recognition of pathogens in the CNS, a process known as bystander activation [108]. This may also give rise to autoreactive lymphocytes in the CNS and the detection of autoantibodies in the CSF that do not cross-react with the pathogen, despite concurrent presence of a persistent infection. For example, in a patient with chronic dengue virus encephalitis [61] multiple lines of evidence indicated the patient had high levels of CSF autoantibodies to microtubule-associated proteins that were confirmed biochemically (Fig. 1). As no pathogen could be detected in this patient initially, a diagnosis of autoimmune encephalitis was considered. Indeed, the misdiagnosis of autoimmune encephalitis is increasing [10▪] some which may be due to immune abnormalities driven by persistent infections. It will be important to elucidate the factors that drive the development of autoantibodies and to determine the specific consequences of autoreactivity in each case.
FIGURE 1.
Autoantibodies are detectable from the sera and CSF of a patient with chronic dengue encephalitis. (A) Immunofluorescence images from human neuronal cultures were fixed with paraformaldehyde, permeabilized with acetone and then stained with mouse MAP6 antibody (red) and human CSF (1 : 25) from a patient with chronic dengue encephalitis (green). Dapi was used to visualize the nuclei (blue). Colocalization of mouse MAP6 antibodies with antibodies in the CSF was observed (bottom right, merged image). (B) Immunoblot images from reciprocal immunoprecipitations (IP). After approval by the Institutional Review Board at the NIH, adult human brain homogenate extracts (temporal cortex, 20 μg) from patients undergoing surgery for epilepsy, were incubated with three microliters of patient sera or mouse MAP6 antibodies for 1 h at 4°C. Antibodies were then captured with protein G beads and eluted from the beads by heating in LDS buffer. Proteins were resolved by gel electrophoresis and probed with either patient sera (1 : 1,000) or mouse anti-MAP6 (1 : 1,000) overnight at 4°C. Horseradish peroxidase (HRP) secondary antibodies (antihuman HRP or antimouse HRP; 1 : 5000) were incubated at room temperature for 1 h. Membranes were developed with enhanced chemiluminescence and visualized on a ProteinSimple FluorChem.
DIAGNOSTICS AND TESTING
Diagnosing CNS infections remains a challenge. Common modes of testing currently in use for the detection of CNS infections primarily utilize the CSF (Table 1). While biopsied tissue samples are often considered optimal for diagnosis, they are invasive and oftentimes risky for the patient. Serum testing has limited value as it does not typically yield CNS-specific information.
Table 1.
Sensitivity and specificity of detecting representative pathogens from the CSF.
| Test | Fluid | Sensitivity (%) | Specificity (%) | Other results/comments | References | |
| BioFire ME panel (Multiplex PCR) | CSF | S. pneumoniae | 87.5 | 98.5 | [109▪] | |
| H. influenzae | 64.9 | 99.4 | ||||
| S. agalactiae | 71.5 | 99.5 | ||||
| E. coli | 70.9 | 99.6 | ||||
| N. meningitidis | 74.5 | 99.1 | ||||
| L. monocytogenes | 70.4 | 98.9 | ||||
| Enterovirus | 93.8 | 99.3 | ||||
| HSV-1 | 75.5 | 99.9 | ||||
| HSV-2 | 94.4 | 99.9 | ||||
| VZV | 91.4 | 99.8 | ||||
| mNGS (UCSF) | CSF | N/A | N/A | 13/58 (22%) CNS infections identified by mNGS that were not identified by clinical testing; 19/58 concurrently diagnosed by mNGS; 11/58 diagnosed by serologic testing only; 7/58 diagnosed by tissue samples; 8/58 negative on mNGS due to low CSF pathogen titers | [119] | |
| VirCapSeq | CSF | N/A | N/A | VirCapSeq-VERT detected virus in the CSF of 16/38 (42%) of patients with no pathogen identified previously; In at least 50% of these cases the virus identified was possibly causative. | [123] | |
| JC virus PCR | CSF | 74–92 | 92–100 | [124,125] | ||
| JC virus PCR (ultrasensitive) | CSF | >95 | N/A | [126] | ||
| HSV PCR | CSF | 98–100 | 94–100 | [110,127] | ||
| VZV IgG | CSF | 93 | N/A | [128] | ||
| VZV PCR | CSF | 30 | N/A | |||
| SARS-CoV-2 PCR | CSF | N/A | N/A | Positive in 0/76 patients | [129] | |
| Positive in 0/21 patients | [130] | |||||
| Ebola PCR | CSF | N/A | N/A | PCR was negative in 0/7 Ebola survivors in 2017 publication | [131] | |
| VDRL | CSF | 67–72 | 80–98 | [132] | ||
CFU, colony forming units; CSF, cerebrospinal fluid; HSV, Herpes simplex virus; mNGS, Metagenomic next-generation sequencing; PCR, polymerase chain reaction; SARS-CoV-2, severe acute respiratory syndrome coronavirus 2; VDRL, venereal disease research laboratory test; VirCap Seq, virome capture sequencing; VZV, varicella-zoster virus.
Polymerase chain reaction (PCR) testing for microbial genetic material in the CSF is often considered the gold standard for diagnosis of neurological infections. However, while this is true for certain organisms, caution must be taken as this assay has suboptimal sensitivity for a number of pathogens. The BioFire FilmArray Meningitis/Encephalitis (ME) Panel is widely used in cases where infectious meningitis and/or encephalitis are suspected. It employs multiplex PCR of CSF samples to detect multiple bacterial and viral organisms. However, a recent meta-analysis showed suboptimal detection for several organisms in the panel, including <75% for five bacterial organisms and 75.5% for HSV-1 [109▪], which has a sensitivity of 98% on conventional PCR testing [110,111]. Another example that illustrates the shortcomings of PCR is varicella-zoster virus (VZV) vasculopathy, in which IgG detection in the CSF demonstrated a three-fold sensitivity over PCR [112]. A more nuanced case of PCR limitations is the diagnosis of PML. Conventional PCR methods are often used for confirmation in suspected PML cases and have a sensitivity of 74–92% [113,114]. However, ultrasensitive PCR methods (sensitivity >95%) [115] have recently been developed, enabling earlier therapeutic intervention and avoiding the need for invasive brain biopsies.
Antibody detection and quantification represent another testing modality. However, the assays used often lack the accuracy desired for clinical diagnosis. For example, the current Lyme antibody testing paradigm is labor intensive and yields both low sensitivity and specificity, and West Nile Virus immunoassays demonstrate poor specificity due to cross-reactivity with other flaviviruses [116]. However, antibody testing may be invoked more frequently for SSPE in coming years, given the expected rise in incidence of this pathology. Established prior data dictates that SSPE may be diagnosed with specific antibody titers of 1:256 or greater in serum, and 1:4 or greater in CSF [117]. Further, antibody testing by VirScan, a phage-display screening tool, has been useful in identifying immune responses to viruses in the CSF which can implicate etiologic agents in cases which have no identified cause [61,118].
Metagenomic next generation sequencing (mNGS) represents a new frontier in detection of CNS microbial infections. The clear benefit of this analysis is seen in cases where uncommon or novel pathogens are implicated in perplexing cases. However, like the BioFire ME panel, sensitivities for certain organisms via mNGS can be lower compared to standard tests for specific organisms, especially when the pathogen load is low [119,120]. Improvements in sensitivity have been demonstrated by using cell-free nucleic acids from CSF as compared to cell-associated DNA [121,122]. Making metagenomics and Virscan diagnostics more accessible could have a significant impact on the diagnosis and management of infectious diseases, especially in resource-poor settings, potentially improving health outcomes and saving lives.
CONCLUSION
In conclusion, the long-term neurological sequelae of persistent infections represent a significant public health concern, particularly in the context of emerging infections such as COVID-19. Despite advancements in our knowledge of the immune response and pathogenesis of these infections, much remains unknown about how persistent infections result in later CNS disease. However, recent findings suggests that there is a window in which intervening by targeting the persistent infections could prevent later development of autoimmunity or neurodegeneration. This is a complex and challenging area of study that requires further research in order to fully understand the mechanisms and develop effective treatments and prevention strategies to mitigate long-term neurological damage. In addition, there is a need for improved diagnostic tools and biomarkers to aid in the identification and monitoring of these infections.
Acknowledgements
This work was supported by the Intramural Research Programs of the NIH National Institute of Neurological Disorders and Stroke.
Financial support and sponsorship
None.
Conflicts of interest
There are no conflicts of interest.
REFERENCES AND RECOMMENDED READING
Papers of particular interest, published within the annual period of review, have been highlighted as:
▪ of special interest
▪▪ of outstanding interest
REFERENCES
- 1.Tian L, Rauvala H, Gahmberg CG. Neuronal regulation of immune responses in the central nervous system. Trends Immunol 2009; 30:91–99. [DOI] [PubMed] [Google Scholar]
- 2.Boldogh I, Albrecht T, Porter DD. Baron S. Persistent Viral Infections. Medical Microbiology 4th ed.Galveston (TX): 1996. [PubMed] [Google Scholar]
- 3.DeMarino C, Lee MH, Cowen M, et al. Detection of SARS-CoV-2 nucleocapsid and microvascular disease in the brain: a case report. Neurology 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4▪▪.Levine KS, Leonard HL, Blauwendraat C, et al. Virus exposure and neurodegenerative disease risk across national biobanks. Neuron 2023; S0896-6273:01147–1153. [DOI] [PMC free article] [PubMed] [Google Scholar]; By examining medical records of hundreds of thousands of people in Finland and the United Kingdom this study identified significant associations between viral infections, especially viral encephalitis, and the later development of neurodegenerative diseases.
- 5.Bukhbinder AS, Ling Y, Hasan O, et al. Risk of Alzheimer's disease following influenza vaccination: a claims-based cohort study using propensity score matching. J Alzheimers Dis 2022; 88:1061–1074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Granerod J, Tam CC, Crowcroft NS, et al. Challenge of the unknown. A systematic review of acute encephalitis in nonoutbreak situations. Neurology 2010; 75:924–932. [DOI] [PubMed] [Google Scholar]
- 7.Granerod J, Ambrose HE, Davies NW, et al. Causes of encephalitis and differences in their clinical presentations in England: a multicentre, population-based prospective study. Lancet Infect Dis 2010; 10:835–844. [DOI] [PubMed] [Google Scholar]
- 8.Vora NM, Holman RC, Mehal JM, et al. Burden of encephalitis-associated hospitalizations in the United States. Neurology 2014; 82:443–451. [DOI] [PubMed] [Google Scholar]
- 9.Glaser CA, Honarmand S, Anderson LJ, et al. Beyond viruses: clinical profiles and etiologies associated with encephalitis. Clin Infect Dis 2006; 43:1565–1577. [DOI] [PubMed] [Google Scholar]
- 10▪.Flanagan EP, Geschwind MD, Lopez-Chiriboga AS, et al. Autoimmune encephalitis misdiagnosis in adults. JAMA Neurol 2023; 80:30–39. [DOI] [PMC free article] [PubMed] [Google Scholar]; This retrospective multicenter study showed 27% of patients diagnosed with autoimmune encephalitis were misdiagnosed. Causes of syndromes included neoplasms, neurodegenerative diseases, psychiatric diseases, and infections among others. One of the primary drivers of misdiagnoses was the presence of autoantibodies in the serum.
- 11.Manfredo Vieira S, Hiltensperger M, Kumar V, et al. Translocation of a gut pathobiont drives autoimmunity in mice and humans. Science 2018; 359:1156–1161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Konig MF, Abusleme L, Reinholdt J, et al. Aggregatibacter actinomycetemcomitans-induced hypercitrullination links periodontal infection to autoimmunity in rheumatoid arthritis. Sci Transl Med 2016; 8:369ra176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Johnson TP, Antiochos B, Rosen A. Robert R, Rich TAF, Harry W. Schroeder, Jr, Cornelia M. Weyand, David B. Corry, Jennifer M. Puck. Mechanisms of autoimmunity. Clinical Immunology, Principles and Practice 6th Edition ed2022. [Google Scholar]
- 14.Dorcet G, Benaiteau M, Bost C, et al. Two cases of late-onset anti-NMDAr auto-immune encephalitis after herpes simplex virus 1 encephalitis. Front Neurol 2020; 11:38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Leypoldt F, Titulaer MJ, Aguilar E, et al. Herpes simplex virus-1 encephalitis can trigger anti-NMDA receptor encephalitis: case report. Neurology 2013; 81:1637–1639. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Le Govic Y, Demey B, Cassereau J, et al. Pathogens infecting the central nervous system. PLoS Pathog 2022; 18:e1010234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Nath A, Johnson TP. Mechanisms of viral persistence in the brain and therapeutic approaches. FEBS J 2022; 289:2145–2161. [DOI] [PubMed] [Google Scholar]
- 18.Gisslen M, Keating SM, Spudich S, et al. Compartmentalization of cerebrospinal fluid inflammation across the spectrum of untreated HIV-1 infection, central nervous system injury and viral suppression. PLoS One 2021; 16:e0250987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Navia BA, Cho ES, Petito CK, Price RW. The AIDS dementia complex: II. Neuropathology. Ann Neurol 1986; 19:525–535. [DOI] [PubMed] [Google Scholar]
- 20.Johnson TP, Nath A. Biotypes of HIV-associated neurocognitive disorders based on viral and immune pathogenesis. Curr Opin Infect Dis 2022; 35:223–230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Galvez C, Urrea V, Dalmau J, et al. Extremely low viral reservoir in treated chronically HIV-1-infected individuals. EBioMedicine 2020; 57:102830. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Imamichi H, Smith M, Adelsberger JW, et al. Defective HIV-1 proviruses produce viral proteins. Proc Natl Acad Sci U S A 2020; 117:3704–3710. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Henderson LJ, Johnson TP, Smith BR, et al. Presence of Tat and transactivation response element in spinal fluid despite antiretroviral therapy. AIDS 2019; 33:S145–S157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Johnson TP, Patel K, Johnson KR, et al. Induction of IL-17 and nonclassical T-cell activation by HIV-Tat protein. Proc Natl Acad Sci U S A 2013; 110:13588–13593. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Wahl A, Al-Harthi L. HIV infection of nonclassical cells in the brain. Retrovirology 2023; 20:1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.McArthur JC, Johnson TP. Chronic inflammation mediates brain injury in HIV infection: relevance for cure strategies. Curr Opin Neurol 2020; 33:397–404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Saylor D, Dickens AM, Sacktor N, et al. HIV-associated neurocognitive disorder--pathogenesis and prospects for treatment. Nat Rev Neurol 2016; 12:234–248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Fraser NW, Lawrence WC, Wroblewska Z, et al. Herpes simplex type 1 DNA in human brain tissue. Proc Natl Acad Sci U S A 1981; 78:6461–6465. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Harrison KS, Zhu L, Thunuguntla P, Jones C. Herpes simplex virus 1 regulates beta-catenin expression in TG neurons during the latency-reactivation cycle. PLoS One 2020; 15:e0230870. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Portegies P, Corssmit N. Epstein-Barr virus and the nervous system. Curr Opin Neurol 2000; 13:301–304. [DOI] [PubMed] [Google Scholar]
- 31.Murata T, Sugimoto A, Inagaki T, et al. Molecular basis of epstein-barr virus latency establishment and lytic reactivation. Viruses 2021; 13: doi: 10.3390/v13122344. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.De Chiara G, Piacentini R, Fabiani M, et al. Recurrent herpes simplex virus-1 infection induces hallmarks of neurodegeneration and cognitive deficits in mice. PLoS Pathog 2019; 15:e1007617. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Singh N, Tscharke DC. Herpes simplex virus latency is noisier the closer we look. J Virol 2020; 94:e01701-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34▪.Bjornevik K, Cortese M, Healy BC, et al. Longitudinal analysis reveals high prevalence of Epstein-Barr virus associated with multiple sclerosis. Science 2022; 375:296–301. [DOI] [PubMed] [Google Scholar]; Using samples from the Department of Defense Serum Repository this study showed that Epstein-Barr virus infection increased the risk of later development of multiple sclerosis. These data support the role of EBV as a driver of multiple sclerosis.
- 35.Itzhaki RF. Overwhelming evidence for a major role for herpes simplex virus type 1 (HSV1) in Alzheimer's Disease (AD); underwhelming evidence against. Vaccines (Basel) 2021; 9:679. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Peluso MJ, Deveau TM, Munter SE, et al. Chronic viral coinfections differentially affect the likelihood of developing long COVID. J Clin Invest 2023; 133:e163669. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Cairns DM, Rouleau N, Parker RN, et al. A 3D human brain-like tissue model of herpes-induced Alzheimer's disease. Sci Adv 2020; 6:eaay8828. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Eimer WA, Vijaya Kumar DK, Navalpur Shanmugam NK, et al. Alzheimer's disease-associated beta-amyloid is rapidly seeded by herpesviridae to protect against brain infection. Neuron 2018; 99:56–63.e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Kumar DK, Choi SH, Washicosky KJ, et al. Amyloid-beta peptide protects against microbial infection in mouse and worm models of Alzheimer's disease. Sci Transl Med 2016; 8:340ra72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Coronel-Castello SP, Lepennetier G, Diddens J, et al. Intrathecally expanding B cell clones in herpes simplex encephalitis: a case report. Neurol Ther 2022; 11:905–913. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41▪▪.Lanz TV, Brewer RC, Ho PP, et al. Clonally expanded B cells in multiple sclerosis bind EBV EBNA1 and GlialCAM. Nature 2022; 603:321–327. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study provides mechanistic links between EBV and multiple sclerosis (MS). By examining the antibodies and sequencing the B-cells from the CSF in patients with MS this study found antibodies that recognize the EBV nuclear antigen 1 (EBNA1) protein cross react with the host protein glial cell adhesion molecule (GlialCAM). Importantly, through longitudinal analyses the authors were able to demonstrate that the autoantibodies arose from continued affinity maturation of viral specific lymphocytes. This provides a mechanism by which a persistent infection gives rise to an autoimmune process.
- 42.Bocharova OV, Fisher A, Pandit NP, et al. Abeta plaques do not protect against HSV-1 infection in a mouse model of familial Alzheimer's disease, and HSV-1 does not induce Abeta pathology in a model of late onset Alzheimer's disease. Brain Pathol 2023; 33:e13116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Johannesdottir Schmidt SA, Veres K, Sorensen HT, et al. Incident herpes zoster and risk of dementia: a population-based Danish cohort study. Neurology 2022; 99:e660–e668. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Tran DN, Bakx A, van Dis V, et al. No evidence of aberrant amyloid beta and phosphorylated tau expression in herpes simplex virus-infected neurons of the trigeminal ganglia and brain. Brain Pathol 2022; 32:e13044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Wozniak MA, Mee AP, Itzhaki RF. Herpes simplex virus type 1 DNA is located within Alzheimer's disease amyloid plaques. J Pathol 2009; 217:131–138. [DOI] [PubMed] [Google Scholar]
- 46.Pisani G, Cristiano K, Pupella S, Liumbruno GM. West Nile virus in Europe and safety of blood transfusion. Transfus Med Hemother 2016; 43:158–167. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Agudelo S, Ventresca M. Modeling the spread of the Zika virus by sexual and mosquito transmission. PLoS One 2022; 17:e0270127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Pierson TC, Diamond MS. The continued threat of emerging flaviviruses. Nat Microbiol 2020; 5:796–812. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Lebov JF, Brown LM, MacDonald PDM, et al. Review: evidence of neurological sequelae in children with acquired zika virus infection. Pediatr Neurol 2018; 85:16–20. [DOI] [PubMed] [Google Scholar]
- 50.Mavigner M, Raper J, Kovacs-Balint Z, et al. Postnatal Zika virus infection is associated with persistent abnormalities in brain structure, function, and behavior in infant macaques. Sci Transl Med 2018; 10: doi: 10.1126/scitranslmed.aao6975. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Sigera PC, Rajapakse S, Weeratunga P, et al. Dengue and postinfection fatigue: findings from a prospective cohort-the Colombo Dengue Study. Trans R Soc Trop Med Hyg 2021; 115:669–676. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Umakanth M, Narayanasami E, Suganthan N. Profuse sweating and hot flashes: an unusual presentation of post-dengue fatigue syndrome. Cureus 2020; 12:e10024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Seet RC, Quek AM, Lim EC. Postinfectious fatigue syndrome in dengue infection. J Clin Virol 2007; 38:1–6. [DOI] [PubMed] [Google Scholar]
- 54.Fulton CDM, Beasley DWC, Bente DA, Dineley KT. Long-term, West Nile virus-induced neurological changes: a comparison of patients and rodent models. Brain Behav Immun Health 2020; 7:100105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Mehta R, Gerardin P, de Brito CAA, et al. The neurological complications of chikungunya virus: a systematic review. Rev Med Virol 2018; 28:e1978. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Garber C, Soung A, Vollmer LL, et al. T cells promote microglia-mediated synaptic elimination and cognitive dysfunction during recovery from neuropathogenic flaviviruses. Nat Neurosci 2019; 22:1276–1288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Levine KS, Leonard HL, Blauwendraat C, et al. Virus exposure and neurodegenerative disease risk across national biobanks. Neuron 2023; S0896-6273:01147–1153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Chapagain S, Pal Singh P, Le K, et al. Japanese encephalitis virus persists in the human reproductive epithelium and porcine reproductive tissues. PLoS Negl Trop Dis 2022; 16:e0010656. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Aid M, Abbink P, Larocca RA, et al. Zika virus persistence in the central nervous system and lymph nodes of rhesus monkeys. Cell 2017; 169:610–620.e14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Hsu DC, Chumpolkulwong K, Corley MJ, et al. Neurocognitive impact of Zika virus infection in adult rhesus macaques. J Neuroinflammation 2022; 19:40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Johnson TP, Larman HB, Lee MH, et al. Chronic dengue virus panencephalitis in a patient with progressive dementia with extrapyramidal features. Ann Neurol 2019; 86:695–703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Appler KK, Brown AN, Stewart BS, et al. Persistence of West Nile virus in the central nervous system and periphery of mice. PLoS One 2010; 5:e10649. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Amuzu C, James PB, Bah AJ, et al. Post-Ebola sequelae among Ebola child survivors in Sierra Leone. BMC Pediatr 2021; 21:482. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Bond NG, Grant DS, Himmelfarb ST, et al. Post-Ebola syndrome presents with multiple overlapping symptom clusters: evidence from an ongoing cohort study in eastern Sierra Leone. Clin Infect Dis 2021; 73:1046–1054. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Scott JT, Sesay FR, Massaquoi TA, et al. Post-Ebola Syndrome, Sierra Leone. Emerg Infect Dis 2016; 22:641–646. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Billioux BJ, Smith B, Nath A. Neurological complications of Ebola virus infection. Neurotherapeutics 2016; 13:461–470. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Wohl DA, Fischer WA, Mei W, et al. Post-Ebola symptoms 7 years after infection: the natural history of Long Ebola. Clin Infect Dis 2022; 76:e835–e840. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Deen GF, Broutet N, Xu W, et al. Ebola RNA persistence in semen of Ebola virus disease survivors - final report. N Engl J Med 2017; 377:1428–1437. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Jacobs M, Rodger A, Bell DJ, et al. Late Ebola virus relapse causing meningoencephalitis: a case report. Lancet 2016; 388:498–503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Varkey JB, Shantha JG, Crozier I, et al. Persistence of Ebola virus in ocular fluid during convalescence. N Engl J Med 2015; 372:2423–2427. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71▪.Liu J, Trefry JC, Babka AM, et al. Ebola virus persistence and disease recrudescence in the brains of antibody-treated nonhuman primate survivors. Sci Transl Med 2022; 14:eabi5229. [DOI] [PubMed] [Google Scholar]; In a rhesus macaque model this study demonstrated that Ebola virus could persist in the CNS in animals treated with monoclonal antibodies. The virus was able to re-emerge and cause severe CNS inflammation culminating in death.
- 72.Gascon GG. Subacute sclerosing panencephalitis. Semin Pediatr Neurol 1996; 3:260–269. [DOI] [PubMed] [Google Scholar]
- 73.Watanabe S, Shirogane Y, Suzuki SO, et al. Mutant fusion proteins with enhanced fusion activity promote measles virus spread in human neuronal cells and brains of suckling hamsters. J Virol 2013; 87:2648–2659. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74▪.Campbell H, Lopez Bernal J, Bukasa A, et al. A Re-emergence of subacute sclerosing panencephalitis in the United Kingdom. Pediatr Infect Dis J 2023; 42:82–84. [DOI] [PubMed] [Google Scholar]; Using national data from the UK Health security Agency this study identified that after 15 years without SSPE there have now been six cases of SSPE beginning in 2017. The authors demonstrate that the incidence of SSPE correlates with decreased vaccination rates for measles. This paper demonstrates that SSPE incidence is on the rise, due to decreased vaccination rates, and reminds clinicians to be aware of this rare and previously absent complication of measles.
- 75.Wendorf KA, Winter K, Zipprich J, et al. Subacute sclerosing panencephalitis: the devastating measles complication that might be more common than previously estimated. Clin Infect Dis 2017; 65:226–232. [DOI] [PubMed] [Google Scholar]
- 76.Lam T, Ranjan R, Newark K, et al. A recent surge of fulminant and early onset subacute sclerosing panencephalitis (SSPE) in the United Kingdom: An emergence in a time of measles. Eur J Paediatr Neurol 2021; 34:43–49. [DOI] [PubMed] [Google Scholar]
- 77.Khetsuriani N, Sanadze K, Abuladze M, Tatishvili N. High risk of subacute sclerosing panencephalitis following measles outbreaks in Georgia. Clin Microbiol Infect 2020; 26:737–742. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Dales LG, Kizer KW, Rutherford GW, et al. Measles epidemic from failure to immunize. West J Med 1993; 159:455–464. [PMC free article] [PubMed] [Google Scholar]
- 79.Li SG, Liang H, Chen YW, Pang YS. Death in children with influenza A (H3N2) virus infection-associated encephalopathy: two case reports. J Int Med Res 2023; 51:3000605221149879. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Fowler A, Ygberg S, Svensson E, et al. Prospective evaluation of childhood encephalitis: predisposing factors, prevention and outcome. Pediatr Infect Dis J 2020; 39:e417–e422. [DOI] [PubMed] [Google Scholar]
- 81.Chen LW, Teng CK, Tsai YS, et al. Influenza-associated neurological complications during 2014-2017 in Taiwan. Brain Dev 2018; 40:799–806. [DOI] [PubMed] [Google Scholar]
- 82.Jang H, Boltz D, Sturm-Ramirez K, et al. Highly pathogenic H5N1 influenza virus can enter the central nervous system and induce neuroinflammation and neurodegeneration. Proc Natl Acad Sci U S A 2009; 106:14063–14068. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Marreiros R, Muller-Schiffmann A, Trossbach SV, et al. Disruption of cellular proteostasis by H1N1 influenza A virus causes alpha-synuclein aggregation. Proc Natl Acad Sci U S A 2020; 117:6741–6751. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.McCall S, Henry JM, Reid AH, Taubenberger JK. Influenza RNA not detected in archival brain tissues from acute encephalitis lethargica cases or in postencephalitic Parkinson cases. J Neuropathol Exp Neurol 2001; 60:696–704. [DOI] [PubMed] [Google Scholar]
- 85.Rohn TT, Catlin LW. Immunolocalization of influenza A virus and markers of inflammation in the human Parkinson's disease brain. PLoS One 2011; 6:e20495. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86▪▪.Stein SR, Ramelli SC, Grazioli A, et al. SARS-CoV-2 infection and persistence in the human body and brain at autopsy. Nature 2022; 612:758–763. [DOI] [PMC free article] [PubMed] [Google Scholar]; In this study the authors describe the presence of SARS-CoV-2 in multiple anatomical sites including the CNS. The tissues included in this study were from 44 individuals who died from COVID-19. Virus was detected in the CNS during acute infection but could also be detected as far out as 230 days postinfection in a single patient. This paper demonstrates that SARS-CoV-2 is widely distributed throughout the body during infection and is not restricted to the respiratory tissues.
- 87.Blagova O, Lutokhina Y, Kogan E, et al. Chronic biopsy proven post-COVID myoendocarditis with SARS-Cov-2 persistence and high level of antiheart antibodies. Clin Cardiol 2022; 45:952–959. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Gaebler C, Wang Z, Lorenzi JCC, et al. Evolution of antibody immunity to SARS-CoV-2. Nature 2021; 591:639–644. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Song E, Zhang C, Israelow B, et al. Neuroinvasion of SARS-CoV-2 in human and mouse brain. J Exp Med 2021; 218:e20202135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Zhao Y, Xia Z, Liang W, et al. SARS-CoV-2 persisted in lung tissue despite disappearance in other clinical samples. Clin Microbiol Infect 2020; 26:1424–1425. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Burgi JJ, Rosslein M, Hornung H, et al. Divergent humoral responses in mild to moderate SARS-CoV-2 infection over time - indication of persistence of the virus? J Infect 2022; 84:418–467. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Vibholm LK, Nielsen SSF, Pahus MH, et al. SARS-CoV-2 persistence is associated with antigen-specific CD8 T-cell responses. EBioMedicine 2021; 64:103230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Walitt B, Johnson TP. The pathogenesis of neurologic symptoms of the postacute sequelae of severe acute respiratory syndrome coronavirus 2 infection. Curr Opin Neurol 2022; 35:384–391. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Choutka J, Jansari V, Hornig M, Iwasaki A. Unexplained postacute infection syndromes. Nat Med 2022; 28:911–923. [DOI] [PubMed] [Google Scholar]
- 95.Spudich S, Nath A. Nervous system consequences of COVID-19. Science 2022; 375:267–269. [DOI] [PubMed] [Google Scholar]
- 96.O’Mahoney LL, Routen A, Gillies C, et al. The prevalence and long-term health effects of Long Covid among hospitalised and nonhospitalised populations: a systematic review and meta-analysis. EClinicalMedicine 2023; 55:101762. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97▪▪.Craddock V, Mahajan A, Spikes L, et al. Persistent circulation of soluble and extracellular vesicle-linked spike protein in individuals with post-acute sequelae of COVID-19. J Med Virol 2023; 95:e28568. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study demonstrates the presence of viral RNA and viral Spike protein in patients with PASC. Importantly, the authors further demonstrate that levels of viral RNA and protein increase from levels observed during acute infection in patients who develop PASC whereas in patients who fully recover from COVID levels of viral RNA and protein decrease.
- 98▪.Swank Z, Senussi Y, Manickas-Hill Z, et al. Persistent circulating severe acute respiratory syndrome coronavirus 2 spike is associated with postacute coronavirus disease 2019 sequelae. Clin Infect Dis 2023; 76:e487–e490. [DOI] [PMC free article] [PubMed] [Google Scholar]; In a study of 63 patients with PASC, this study found detectable viral Spike protien in approximately 60% of patients up to a year after infection.
- 99.Blennow O, Vesterbacka J, Tovatt T, Nowak P. Successful combination treatment for persistent SARS-CoV-2 infection. Clin Infect Dis 2023; ciad085. [DOI] [PubMed] [Google Scholar]
- 100.Trottier CA, Wong B, Kohli R, et al. Dual antiviral therapy for persistent COVID-19 and associated organizing pneumonia in an immunocompromised host. Clin Infect Dis 2022; 76:923–925. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Peluso MJ, Anglin K, Durstenfeld MS, et al. Effect of oral nirmatrelvir on long COVID symptoms: 4 cases and rationale for systematic studies. Pathog Immun 2022; 7:95–103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Louveau A, Herz J, Alme MN, et al. CNS lymphatic drainage and neuroinflammation are regulated by meningeal lymphatic vasculature. Nat Neurosci 2018; 21:1380–1391. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Laman JD, Weller RO. Drainage of cells and soluble antigen from the CNS to regional lymph nodes. J Neuroimmune Pharmacol 2013; 8:840–856. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Smolders J, Heutinck KM, Fransen NL, et al. Tissue-resident memory T cells populate the human brain. Nat Commun 2018; 9:4593. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Calzascia T, Masson F, Di Berardino-Besson W, et al. Homing phenotypes of tumor-specific CD8 T cells are predetermined at the tumor site by crosspresenting APCs. Immunity 2005; 22:175–184. [DOI] [PubMed] [Google Scholar]
- 106▪▪.Frieser D, Pignata A, Khajavi L, et al. Tissue-resident CD8(+) T cells drive compartmentalized and chronic autoimmune damage against CNS neurons. Sci Transl Med 2022; 14:eabl6157. [DOI] [PubMed] [Google Scholar]; This study used orthogonal approaches, including human tissues and murine models, to demonstrate that tissue-resident CD8+ T cells are the drivers of chronic CNS autoimmunity. Within the context of CNS diseases this work further showed that tissue-resident CD8+ T cells, independent of peripheral CD8+ T cells, mediated neuroinflammation and loss of neurons.
- 107.Yenyuwadee S, Sanchez-Trincado Lopez JL, Shah R, et al. The evolving role of tissue-resident memory T cells in infections and cancer. Sci Adv 2022; 8:eabo5871. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Lee HG, Lee JU, Kim DH, et al. Pathogenic function of bystander-activated memory-like CD4(+) T cells in autoimmune encephalomyelitis. Nat Commun 2019; 10:709. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109▪.Trujillo-Gomez J, Tsokani S, Arango-Ferreira C, et al. Biofire FilmArray Meningitis/Encephalitis panel for the aetiological diagnosis of central nervous system infections: a systematic review and diagnostic test accuracy meta-analysis. EClinicalMedicine 2022; 44:101275. [DOI] [PMC free article] [PubMed] [Google Scholar]; This is a systematic review and meta-analyses of the utility of the Biofire FilmArray panel for diagnosis CNS infections. Estimated sensitivity and specificity data were generated.
- 110.Lakeman FD, Whitley RJ. Diagnosis of herpes simplex encephalitis: application of polymerase chain reaction to cerebrospinal fluid from brain-biopsied patients and correlation with disease. National Institute of Allergy and Infectious Diseases Collaborative Antiviral Study Group. J Infect Dis 1995; 171:857–863. [DOI] [PubMed] [Google Scholar]
- 111.Aurelius E, Johansson B, Skoldenberg B, Forsgren M. Encephalitis in immunocompetent patients due to herpes simplex virus type 1 or 2 as determined by type-specific polymerase chain reaction and antibody assays of cerebrospinal fluid. J Med Virol 1993; 39:179–186. [DOI] [PubMed] [Google Scholar]
- 112.Nagel MA, Forghani B, Mahalingam R, et al. The value of detecting anti-VZV IgG antibody in CSF to diagnose VZV vasculopathy. Neurology 2007; 68:1069–1073. [DOI] [PubMed] [Google Scholar]
- 113.Cinque P, Koralnik IJ, Clifford DB. The evolving face of human immunodeficiency virus-related progressive multifocal leukoencephalopathy: defining a consensus terminology. J Neurovirol 2003; 9:88–92. [DOI] [PubMed] [Google Scholar]
- 114.Fong IW, Toma E. The natural history of progressive multifocal leukoencephalopathy in patients with AIDS. Canadian PML Study Group. Clin Infect Dis 1995; 20:1305–1310. [DOI] [PubMed] [Google Scholar]
- 115.Berger JR, Danaher RJ, Dobbins J, et al. Dynamic expression of JC virus in urine and its relationship to serostatus. Mult Scler Relat Disord 2020; 41:101972. [DOI] [PubMed] [Google Scholar]
- 116.Ramachandran PS, Wilson MR. Diagnostic testing of neurologic infections. Neurol Clin 2018; 36:687–703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Garg RK. Subacute sclerosing panencephalitis. Postgrad Med J 2002; 78:63–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Galardi MM, Sowa GM, Crockett CD, et al. Pathogen and antibody identification in children with encephalitis in Myanmar. Ann Neurol 2022; 93:615–628. [DOI] [PubMed] [Google Scholar]
- 119.Wilson MR, Sample HA, Zorn KC, et al. Clinical metagenomic sequencing for diagnosis of meningitis and encephalitis. N Engl J Med 2019; 380:2327–2340. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Leon KE, Schubert RD, Casas-Alba D, et al. Genomic and serologic characterization of enterovirus A71 brainstem encephalitis. Neurol Neuroimmunol Neuroinflamm 2020; 7:e703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Li X, Yang L, Li D, et al. Diagnosis of neurological infections in pediatric patients from cell-free DNA specimens by using metagenomic next-generation sequencing. Microbiol Spectr 2023; 11:e0253022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Yu L, Zhang Y, Zhou J, et al. Metagenomic next-generation sequencing of cell-free and whole-cell DNA in diagnosing central nervous system infections. Front Cell Infect Microbiol 2022; 12:951703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.McGill F, Tokarz R, Thomson EC, et al. Viral capture sequencing detects unexpected viruses in the cerebrospinal fluid of adults with meningitis. J Infect 2022; 84:499–510. [DOI] [PubMed] [Google Scholar]
- 124.Cinque P, Scarpellini P, Vago L, et al. Diagnosis of central nervous system complications in HIV-infected patients: cerebrospinal fluid analysis by the polymerase chain reaction. AIDS 1997; 11:1–17. [DOI] [PubMed] [Google Scholar]
- 125.Fong IW, Britton CB, Luinstra KE, et al. Diagnostic value of detecting JC virus DNA in cerebrospinal fluid of patients with progressive multifocal leukoencephalopathy. J Clin Microbiol 1995; 33:484–486. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Berger JR, Aksamit AJ, Clifford DB, et al. PML diagnostic criteria: consensus statement from the AAN Neuroinfectious Disease Section. Neurology 2013; 80:1430–1438. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Aurelius E, Johansson B, Skoldenberg B, et al. Rapid diagnosis of herpes simplex encephalitis by nested polymerase chain reaction assay of cerebrospinal fluid. Lancet 1991; 337:189–192. [DOI] [PubMed] [Google Scholar]
- 128.Nagel MA, Cohrs RJ, Mahalingam R, et al. The varicella zoster virus vasculopathies: clinical, CSF, imaging, and virologic features. Neurology 2008; 70:853–860. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Jarius S, Pache F, Kortvelyessy P, et al. Cerebrospinal fluid findings in COVID-19: a multicenter study of 150 lumbar punctures in 127 patients. J Neuroinflammation 2022; 19:19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Tuma RL, Guedes BF, Carra R, et al. Clinical, cerebrospinal fluid, and neuroimaging findings in COVID-19 encephalopathy: a case series. Neurol Sci 2021; 42:479–489. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Billioux BJ, Nath A, Stavale EJ, et al. Cerebrospinal fluid examination in survivors of Ebola virus disease. JAMA Neurol 2017; 74:1141–1143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Marra CM, Tantalo LC, Maxwell CL, et al. The rapid plasma reagin test cannot replace the venereal disease research laboratory test for neurosyphilis diagnosis. Sex Transm Dis 2012; 39:453–457. [DOI] [PMC free article] [PubMed] [Google Scholar]


