Skip to content
BY 4.0 license Open Access Published by De Gruyter July 10, 2023

The elusive role of herpesviruses in Alzheimer’s disease: current evidence and future directions

  • Stacey L. Piotrowski , Allison Tucker and Steven Jacobson EMAIL logo

Abstract

Alzheimer’s disease (AD) is the most common cause of dementia. While pathologic hallmarks, such as extracellular beta-amyloid plaques, are well-characterized in affected individuals, the pathogenesis that causes plaque formation and eventual cognitive decline is not well understood. A recent resurgence of the decades-old “infectious hypothesis” has garnered increased attention on the potential role that microbes may play in AD. In this theory, it is thought that pathogens such as viruses may act as seeds for beta-amyloid aggregation, ultimately leading to plaques. Interest in the infectious hypothesis has also spurred further investigation into additional characteristics of viral infection that may play a role in AD progression, such as neuroinflammation, latency, and viral DNA integration. While a flurry of research in this area has been recently published, with herpesviruses being of particular interest, the role of pathogens in AD remains controversial. In this review, the insights gained thus far into the possible role of herpesviruses in AD are summarized. The challenges and potential future directions of herpesvirus research in AD and dementia are also discussed.

Introduction

Alzheimer’s disease (AD) is the leading cause of dementia, with an estimated 50 million people affected worldwide [13]. AD is a multifactorial neurodegenerative disease, where characteristics such as genes and environment are believed to play important roles [4, 5]. A pathologic hallmark of AD is extracellular senile plaques, which are comprised of beta-amyloid (Aβ) and accumulate in regions of the brain such as the hippocampus [5, 6]. While these pathologic changes are well-characterized, the pathogenesis leading to Aβ plaque formation and ultimately cognitive decline in AD is poorly understood [2, 5, 7].

Multiple hypotheses have been investigated in efforts to better understand the pathogenesis of AD [5, 8, 9]. For decades, the amyloid cascade hypothesis has spurred extensive research with the idea that Aβ deposition is the initial event in AD [8, 10]. Aβ accumulation is suspected to lead to further downstream effects such as intracellular tau neurofibrillary tangle formation, neuroinflammation, neuronal cell loss, neurodegeneration, and cognitive decline [7, 11]. While the reasons for the continued failures of clinical therapeutic trials that target Aβ are likely complex, including the current lack of biomarkers needed to identify patients early in disease progression and lack of objective clinical trial endpoint measures, it suggests that AD pathogenesis is likely not simply a function of the amyloid cascade [1215]. Recently, interest in the role of infections and infectious triggers or contributors in AD has garnered renewed attention [16].

The idea that a pathogen may play a role in AD dates back to the early 1900s, where it was originally proposed by Alois Alzheimer and Oskar Fischer [17, 18]. Since then, numerous bacteria, fungi, parasites, and viruses have been investigated for an association with AD [17, 19]. While a variety of viruses have been investigated for their potential role in AD, including human immunodeficiency virus (HIV), hepatitis C virus (HCV), and the recent severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2, COVID-19), herpesviruses have been a consistent focus of attention in the AD field (Table 1) [17, 1925]. However, evidence for the role of herpesviruses in AD, and the infectious hypothesis in general, remains highly debated [26, 27].

Table 1:

Timeline of selected publications investigating the role of viruses in Alzheimer’s disease (AD).

Pathogen Year of Publications
Herpes simplex virus type 1 (HSV-1) 1982 [67]
1997 [82]
2008 [79]
2009 [68,78,155]
2011 [73]
2018 [69]
2019 [70, 71, 75]
2020 [72, 76, 81]
2021 [74, 77, 83]
2022 [84]
Human herpesvirus 6 (HHV-6) 2014 [104]
2015 [102]
2017 [103]
2018 [46, 69]
2020 [98]
Epstein–Barr virus (EBV) 2014 [104]
2021 [110, 118]
Cytomegalovirus (CMV) 2018 [123]
Varicella zoster virus (VZV) 2018 [120]
2021 [119, 121]
2022 [122]
Hepatitis C virus (HCV) 2014 [24]
Human immunodeficiency virus (HIV) 2017 [23]
2021 [25]
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2, COVID-19) 2022 [21]

Infectious hypothesis of AD

Neurons are likely the primary source of Aβ in the brain [28]. Aβ is generated via the cleavage of amyloid precursor protein (APP) by β-secretase, followed by γ-secretase [29, 30]. An imbalance of Aβ production and clearance is thought to contribute to the excessive accumulation of Aβ in AD [31]. While the accumulation of Aβ is potentially neurotoxic, evidence suggests that Aβ may initially be beneficial [32].

Aβ is an evolutionary conserved peptide, suggesting it serves an important physiological role [33, 34]. LL-37, the human antimicrobial peptide (AMP), is structurally similar to Aβ, with both proteins able to form oligomers and insoluble fibrils [35]. Evidence that Aβ functions as an innate AMP has recently grown, with both in vitro and in vivo studies showing that microbial infection can increase Aβ production and aggregation [36]. Additionally, in vitro assays have also demonstrated the antimicrobial activity of Aβ against a variety of pathogens [35, 36].

One avenue of investigation in the infectious hypothesis theorizes that pathogens are the primary cause of AD, triggering the amyloid cascade and ultimately leading to plaque formation and other neurodegenerative changes associated with AD [16, 19]. Due to Aβ′s function as an AMP, it has been proposed that microbes, such as herpesviruses, that enter the brain can function as “seeds” for Aβ aggregation and fibrillation (Figure 1) [37, 38]. When viruses enter the brain, microglia and astrocytes are stimulated to produce cytokines such as interferon, which may increase Aβ production from neurons through interferon-induced transmembrane protein 3 (IFITM3)-mediated γ-secretase activity [39]. Aβ is then nucleated by the pathogen or a portion of the pathogen, such as glycoproteins in the case of viruses, triggering fibril aggregation and organization around the invading infectious agent, subsequently protecting the brain from further infection but ultimately causing the Aβ accumulation and plaque formation seen in AD [18].

Figure 1: 
Herpesviruses and the infectious origin hypothesis of Alzheimer’s disease (AD). The infectious origin hypothesis of AD theorizes that the following general events may occur in response to pathogens that enter the brain, ultimately leading to beta-amyloid (Aβ) plaque formation. 1 The pathogen, such as a herpesvirus, infects the brain. 2 In response to the viral infection, microglia and astrocytes secrete pro-inflammatory cytokines, such as interferon. 3 Neurons respond to the binding of these cytokines by increasing Aβ production, via mechanisms like interferon-induced transmembrane protein 3 (IFITM3)-mediated γ-secretase activity. 4 Aβ is “seeded” and nucleated by the virus or a portion of the virus such as glycoproteins, with Aβ fibrils aggregating around the virus and trapping it. 5 Aβ plaques that are seen in AD are ultimately formed as Aβ continues to aggregate around the virus. 6 Aβ aggregation stimulates further microglial activation and neuroinflammation, further contributing to continued Aβ production. Figure 1 was created with BioRender.com.
Figure 1:

Herpesviruses and the infectious origin hypothesis of Alzheimer’s disease (AD). The infectious origin hypothesis of AD theorizes that the following general events may occur in response to pathogens that enter the brain, ultimately leading to beta-amyloid (Aβ) plaque formation. 1 The pathogen, such as a herpesvirus, infects the brain. 2 In response to the viral infection, microglia and astrocytes secrete pro-inflammatory cytokines, such as interferon. 3 Neurons respond to the binding of these cytokines by increasing Aβ production, via mechanisms like interferon-induced transmembrane protein 3 (IFITM3)-mediated γ-secretase activity. 4 Aβ is “seeded” and nucleated by the virus or a portion of the virus such as glycoproteins, with Aβ fibrils aggregating around the virus and trapping it. 5 Aβ plaques that are seen in AD are ultimately formed as Aβ continues to aggregate around the virus. 6 Aβ aggregation stimulates further microglial activation and neuroinflammation, further contributing to continued Aβ production. Figure 1 was created with BioRender.com.

In addition to the potential role of microbes as the “infectious origin of AD,” there has been growing interest in the possible contributions of viral infection to AD progression, the “infectious comorbidity in AD” theory [40]. As in other chronic neurodegenerative diseases, the idea of “two hits” or “multiple hits” contributing to disease manifestation, including genetic predispositions, oxidative stress, neuroinflammation and environmental factors such as pathogen exposures, has gained traction in AD research [4143]. In addition to the direct “seeding” of plaque formation by viruses as a potential causative element, the multifactorial nature of AD suggests that viruses and other infectious agents may act as contributory factors in some subtypes of AD patients through a variety of mechanisms [4, 44].

Some viruses have a unique ability to integrate into human host cell chromosomes as a part of their life cycle or incidentally, potentially allowing for disruptions in gene expression and other effects for the host cell that may contribute to the development of disease [45, 46]. Some pathogens, such as herpesviruses, result in lifelong latent infection that may cause intermittent damage that contributes to AD progression after reactivation [40, 47, 48]. Both acute and chronic viral infection can activate microglia and stimulate cytokine release, with resulting neuroinflammation that may drive and influence both Aβ and tau pathology [4952]. Aβ aggregation has been shown to subsequently further induce microglial activation and stimulate inflammatory mediators, creating a cycle of enhanced protein deposition and continued neuroinflammation [37, 53, 54]. Microglial activation has also been shown to activate A1 astrocytes and contribute to a neurotoxic response, resulting in the death of neurons and oligodendrocytes [52, 55]. Neuroinflammation, which can be mediated by viral infections, may ultimately drive neurodegeneration and disease progression in AD [56].

Additional mechanisms by which viruses may contribute to AD are also being explored. Dysregulated autophagy, which is associated with the progressive pathology of AD, has also been associated with neurotropic viruses [57]. While microRNAs (miRNAs) have been investigated as a potential biomarker of AD, viral infection may also contribute to the dysregulation of miRNA activity that has been linked to AD [58, 59].

Due to their ubiquitous nature, their association with other neurodegenerative diseases, and their life cycle characteristics, herpesviruses have been intensely investigated and scrutinized for their potential role in AD [18, 26, 60, 61]. This interest has spanned across multiple decades and has increasingly focused on specific herpesviruses in recent years, with most attention concentrated on herpes simplex virus type 1 (HSV-1), human herpesvirus 6 (HHV-6), and Epstein–Barr virus (EBV). Varying associations in a multitude of studies highlight the need for additional research and further scrutiny of the possible role of herpesviruses in AD pathogenesis.

Herpes simplex virus type 1 (HSV-1)

Herpes simplex virus type 1 (HSV-1), a neurotropic virus that can establish a latent infection with subsequent reactivation, has been investigated for decades as a possible factor in AD (Table 2) [62, 63]. Infection usually occurs in childhood, with approximately 70 % of the worldwide population having antibodies to HSV-1 [64]. HSV-1 infection can result in a wide-variety of clinical manifestations, including infection of the central nervous system and encephalitis [65, 66].

Table 2:

Selected publications investigating the possible role of herpes simplex virus type 1 (HSV-1) in Alzheimer’s disease (AD).

Reference Results Methodology
Wozniak et al. [68]. Increased HSV-1 DNA co-localization with Aβ plaques in AD patients In situ PCR and immunohistochemistry
Eimer et al. [69]. Aβ fibrillization and oligomerization mediated by HSV-1

Aβ protects against HSV-1 infection
In vitro culture

5XFAD transgenic and wild-type mice
Linard et al. [81]. APOE4 carriers with IgM or high IgG for HSV-1 had increased risk for AD Prospective cohort and chemiluminescence immunoassay
Bocharova et al. [74]. Aβ does not protect against HSV-1 infection 5XFAD transgenic and wild-type mice
Murphy et al. [83]. HSV-1 seropositivity associated with cognitive decline, not dementia and AD Prospective population-based cohort and chemiluminescence immunoassay
Zhang et al. [84] Anti-HSV-1 IgG not associated with risk of AD Multigenomics analysis and two-sample Mendelian randomization analysis

In 1982, reactivation of HSV-1 infection was initially suggested as a possible cause of neurodegeneration and AD due to similarly affected regions of the brain [67]. HSV-1 DNA was later shown to co-localize with Aβ plaques to a greater extent in AD patients than age-matched controls, suggesting that HSV-1 may be the cause of AD-related plaque formation [68]. More recently, in vitro assays showed Aβ fibrillization and oligomerization mediated by HSV-1 [69]. The interaction between the viral protein corona of HSV-1 and the extracellular environment has been demonstrated as a critical component of HSV-1 induced Aβ accumulation [70]. A 3D human brain-like tissue model comprised of human-induced neural stem cells also demonstrated extensive Aβ fibrillary plaque-like formations upon lower dose inoculation of HSV-1, coupled with an upregulation in presenilin 1 (PSEN1) and presenilin 2 (PSEN2), the catalytic subunits of γ-secretase, and an increase in glial activity and markers of neuroinflammation [71]. Additionally, HSV-1 has been shown to depolarize the plasma membrane of mouse cortical neurons and increase intracellular calcium, which initiates APP production similar to a transgenic mouse model of AD [72, 73]. However, in vivo models have led to varying results, with one study showing protection against HSV-1 encephalitis in 5XFAD transgenic mice that express human Aβ, while another study with a lower dose of viral inoculum failed to show Aβ protection from HSV-1 infection in the same transgenic strain, coupled with a lack of evidence of HSV-1-triggered Aβ aggregation [69, 74].

Repeated infection of HSV-1 has been associated with AD-like neurodegeneration. In transgenic mice, non-Aβ AD biomarkers, such as hyperphosphorylated tau protein and astrogliosis, increased with recurrent HSV-1 infection [75]. The impact of HSV-1 infection on the hippocampus includes the accumulation of AD biomarkers in the dentate gyrus (DG) alongside increased neuroinflammation and lymphocytic infiltration in mouse models [7577]. In both in vitro and in vivo models, proliferation and differentiation of neural stem cells (NSCs) in the DG decreased post-HSV-1 infection [7577]. Changes in gene expression contribute to immunological cascades associated with AD pathogenesis and increase interferon-gamma, with some of these cascades linked to the upregulation of miRNA-146a [78, 79]. miRNAs expressed by HSV-1 can dysregulate host miRNA activity; however, these changes are often restricted to cell type or environment [80].

Studies investigating HSV-1 as a risk factor for AD have suggested that individuals with both HSV-1 positivity in the brain and apolipoprotein E4 (APOE4) allele, a genetic susceptibility factor for AD, have an increased risk for AD [81, 82]. However, a recent population-based study did not find an association between HSV-1 seropositivity and risk of dementia, but seropositivity was associated with subtle cognitive decline [83]. Additionally, a multigenomics analysis and two-sample Mendelian randomization study found no association between anti-HSV-1 immunoglobulins and risk of AD [84]. The often-disparate results between studies emphasizes the need for continued investigation using a multitude of methodologies to more completely characterize the potential role of HSV-1 in AD.

Human herpesvirus 6 (HHV-6)

Over 95 % of people over the age of 2 are seropositive for HHV-6, a betaherpesvirus [85]. HHV-6A and HHV-6B are two distinct variants, with evidence suggesting that HHV-6A is more neurotropic [86]. While primary childhood infection with HHV-6B can be asymptomatic or the cause of febrile illness, HHV-6A and -6B have both been associated with neurological diseases such as mesial temporal lobe epilepsy, encephalitis, and multiple sclerosis (MS) [87, 88]. Interestingly, the brain regions impacted by HHV-6 infection in diseases such as mesial temporal lobe epilepsy can be co-localized to regions of interest in AD, like the hippocampus [8991].

The notion that HHV-6 could be associated with Alzheimer’s disease was strongly ignited by two recent publications (Table 3) [46, 69]. In vitro assays showed Aβ fibrillization and oligomerization mediated by HHV-6 and its viral glycoproteins [69]. Using advanced multiomics analyses, increased HHV-6A and human herpesvirus 7 (HHV-7) were found in brains of individuals with Alzheimer’s disease [46]. Dysregulation of miRNA155 was identified as a potential contributor to neuroinflammation in AD, with additional studies showing that HHV-6A infected immune cells actively suppress miRNA155 activity [46, 92, 93]. While intriguing and exciting, these bioinformatics analyses and their implications have been questioned, spurring further analyses and varying interpretations based on the computational pipelines utilized on the omics data [9497].

Table 3:

Selected publications investigating the possible role of human herpesvirus 6 (HHV-6) in Alzheimer’s disease (AD).

Reference Results Methodology
Carbone et al. [104]. Increased HHV-6 positivity in peripheral blood leukocytes (PBL) in AD patients; PBL positivity increased in patients who developed AD Nested and/or quantitative real-time PCR (qPCR)
Agostini et al. [102]. No correlation between HHV-6 IgG and AD Enzyme-linked immunoassay (ELISA)
Westman et al. [103]. Decreased HHV-6 IgG in AD Multiplex immunoassay
Eimer et al. [69]. Aβ fibrillization and oligomerization mediated by HHV-6 In vitro culture
Readhead et al. [46]. Increased HHV-6A in brains of individuals with AD Multiomic and computational analyses
Allnutt et al. [98]. No association between HHV-6 and AD RNA-seq analysis and digital droplet PCR (ddPCR)

Despite using many of the same cohorts, a subsequent study failed to support an association between HHV-6 and AD when HHV-6 PCR reactivity of DNA extracted from AD and control brains was quantified, with low frequency of HHV-6 detection and low magnitude of HHV-6 PCR reactivity in cohorts [98]. In this study, the Broad Institute tool PathSeq was also used to screen the RNA-seq data for over 25,000 microbes including multiple herpesviruses, and it demonstrated no difference in viral detection between AD patients and controls [98]. A similar RNA-seq analysis in a different cohort also found similar HHV-6 abundance in AD and control brains [99]. Other transcriptomic profile comparisons revealed shared gene expression between AD patients and those with active HHV-6 infection [100, 101]. These seemingly disparate results have fueled discussions on the varying methodologies and analyses utilized in bioinformatics and omics-based approaches, their sensitivities for viral detection when compared to other more traditional assays such as PCR, and the different results anticipated from direct methods, such as PCR, and indirect methods, including serology [46, 9498].

Other studies investigating the association between HHV-6 and AD via immunological measures have also achieved divergent results. While one study found decreased HHV-6 IgG in AD patients, suggesting a possible impaired humoral immunity to HHV-6 in AD, another study failed to find any association between the humoral response against HHV-6 and AD [102, 103]. A study in Italy found increased HHV-6 positivity in peripheral blood leukocytes of AD patients, which was also associated with clinical AD progression and suggested possible peripheral reactivation of HHV-6 may play a role in AD patients [104]. However, this relationship between leukocyte positivity in AD patients was not present in a similar study utilizing a different cohort [103].

Changes in AD associated pathology have been observed in cell cultures post-exposure to HHV-6A, with microglia infected with HHV-6A increasing Aβ1-42 expression [105]. Additionally, HHV-6A infection of peripheral blood mononuclear-microglial cells resulted in an increase in both tau and hyperphosphorylated tau [105]. While mice are not naturally competent to HHV-6 infection, the closely related murine roseolovirus failed to increase cortical Aβ deposition in the transgenic 5XFAD mouse model [99]. However, like most studies in this field, these results don’t refute the infectious hypothesis of AD or the antimicrobial protection hypothesis of Aβ [99].

Epstein–Barr virus (EBV)

Human herpesvirus 4, more commonly known as Epstein–Barr virus (EBV), has long been investigated as a contributor to the demyelinating disease MS, with recent publications lending further credence to its role in MS pathogenesis [88, 106], [107], [108]. Approximately 90 % of the worldwide population is estimated to be infected with EBV, with clinical syndromes varying in part due to age of initial infection [109]. Few studies have investigated the role of EBV in AD, with one study in 2014 suggesting that EBV, in addition to the previously mentioned HHV-6, may be a risk factor for AD, and it may contribute to AD progression based on PCR positivity for EBV in peripheral blood leukocytes from AD patients [104] (Table 4). A recently published Mendelian randomization study observed an association between mononucleosis, the result of primary EBV infection later in life, and AD, suggesting that further investigation of EBV and its possible role in AD pathogenesis is warranted and that antiviral therapies may be beneficial in AD prevention or treatment [110].

Table 4:

Selected publications investigating the possible role of Epstein–Barr virus (EBV) in Alzheimer’s disease (AD).

Reference Results Methodology
Carbone et al. [104]. PBL positivity for EBV increased in patients who developed AD

IgG levels for EBV increased in patients who developed AD
Nested and/or quantitative real-time PCR (qPCR)

Enzyme-linked immunoassay (ELISA)
Huang et al. [110]. Significant association between mononucleosis and risk of AD Two-sample Mendelian randomization study
Tiwari et al. [118]. EBV peptides can potentially form cytotoxic aggregates In silico bioinformatics analysis

Based on cell cycle dysregulation mediated by EBV infection, it has been proposed that this dysregulation could also contribute to neurodegeneration in AD [111]. EBV may contribute to neuroinflammation in AD through the infection of monocytes and peripheral blood mononuclear cells [112115]. Two known EBV antigens have been associated with an adaptive immune response in the cerebrospinal fluid of AD patients, with their presence triggering a cytotoxic immune cascade mediated by the proinflammatory activity of CD8+ T cells [116, 117]. In silico analysis of EBV proteins have revealed potential peptides capable of forming cytotoxic aggregates, with further in vitro and in vivo experiments needed [118].

Other herpesviruses

Other herpesviruses have been implicated for their potential role in AD due to hypothesized interactions with other viruses [17]. Varicella zoster virus (VZV, human herpesvirus 3) has been found at an increased prevalence in AD patients, and antiviral therapy after VZV diagnosis has been associated with a reduced risk of dementia [119121]. However, in vitro studies suggest VZV does not directly trigger Aβ pathology, but rather, it can reactivate HSV-1 and indirectly contribute to Aβ production [122]. Similarly, the presence of both cytomegalovirus (CMV) and HSV-1 antibodies, not either virus alone, has been described as a significant risk factor for AD, suggesting interaction of multiple herpesviruses may be key to AD [123].

The challenges of demonstrating the potential role of herpesviruses in AD

Obtaining definitive evidence for the role of herpesviruses in AD has thus far remained elusive, highlighting the inherent difficulties in this area of investigation. The association of herpesviruses with AD does not prove causation, but it can and should stimulate further investigation, as in the case of the infectious hypothesis and AD [124126]. A recent publication describes the evidence that may suggest a causal role for microbes like HHV-6 in disease, including, among others, viral nucleic acid or antibody levels that correlate to disease severity, lack of detection of other infectious agents in the diseased tissue, and specific antiviral therapy that decreases viral load and is followed by clinical improvement [27]. However, whether all of these criteria are widely-applicable to proving the role of herpesvirus infection in AD is unknown, particularly as some criteria, such as presence of viral nucleic acids, does not immediately allow for characterization of a virus as a cause, cofactor, or bystander in the disease process [27]. Additionally, since the infectious hypothesis suggests that many infectious agents may play a role in AD, antimicrobials directed at one specific agent may not be widely successful or beneficial if it is not combined with more general therapeutics aimed at other contributors to AD such as neuroinflammation [27, 36, 127]. As the necessary specific criteria and threshold to prove causation remains undefined, evidence for the role of herpesviruses in AD can only grow with continued investigation and with replication of results through various methodologies and in multiple different cohorts [27].

The lifecycle of herpesviruses provides additional obstacles for proving their potential role in AD. Herpesviruses cause a lifelong infection, with initial infection usually occurring at an early age [27]. If herpesviruses play a role in AD, the interval from primary infection to clinical disease onset is decades. Current analytical methods, including omics-related analyses and more traditional diagnostic assays such as PCR, may not be specific or sensitive enough to detect the remnants of viral infection that may be present in AD tissue decades later [18, 27]. Additionally, the host-herpesvirus interaction allows for two different life cycles, latent and lytic cycles, where primary infection and reactivation could potentially play different roles in disease pathogenesis [19, 60, 128]. More recently developed investigative strategies and techniques, such as extracellular vesicle interrogation, and novel analytical methods may be necessary to help elucidate further evidence of herpesvirus infection and its role in AD patients [129132]. Extracellular vesicles, membrane-bound structures that contain a wide-variety of cargo, may serve as sensitive and specific biomarkers for viral infection and AD, while also serving as a potential treatment modality to deliver therapeutic cargo [133].

Herpesviruses are also ubiquitous, with all humans infected with at least one herpesvirus in their lifetime [128]. A ubiquitous pathogen like HHV-6 can cause a specific disease in a subset of infected individuals, as many factors, such as genetics and comorbidities, may contribute and ultimately culminate in disease manifestation [27]. AD is multifactorial and heterogenous, with factors like genetics, inflammation, and environmental aspects contributing to various extents in different subgroups of patients [4]. While they may not be the sole cause of AD, herpesviruses could be a contributing factor in some, but not all, AD patients [44].

Multiple in vitro and in vivo studies have shown that Aβ can act as an antimicrobial peptide against a variety of pathogens, and Aβ plaque formation can also be seeded by many microbes [35, 36, 134, 135]. Some meta-analyses have associated a variety of herpesviruses with an increased risk of AD, highlighting a possible lack of specificity for a single herpesvirus as a cause of AD [136, 137]. Multi-pathogen infections, including both single-taxon and multi-taxon combinations, and overall infectious burden may be greater contributors to AD, and the relevant combinations of beneficial antimicrobial therapeutics may differ from patient to patient [138141]. Studies that focus on a single microbe in AD, such as HHV-6 or HSV-1 as a sole cause or contributory factor in AD, may erroneously simplify the clinical scenario where multiple infectious agents, in combination with other attributes such as genetics, contribute to disease manifestation and progression. While studies analyzing multiple contributory agents or other components may be more difficult to design and interpret, they are likely necessary to more fully understand the effects of herpesvirus infection in AD. The proper methodology for determining which patients may have viral infection as a factor in their AD pathogenesis is not clear, but it is likely essential for the success of any forthcoming antiviral-based AD treatment clinical trials. The establishment of pathogen-based biomarkers may aid in identifying patients with viral contributors to AD and help distinguish appropriate trial and treatment populations [56].

Potential future directions of herpesvirus research in AD

Failure of Aβ-targeting therapeutics has recently called into question whether Aβ is the major contributor to AD progression [11]. The challenges of successfully targeting Aβ, such as determining the appropriate timeframe and adequate dosages for therapeutic administration, have also led to the diversification of targets for AD treatments [12], [13], [14], [15, 142, 143]. In addition to Aβ plaques, neurofibrillary tangles, intraneuronal aggregates of hyperphosphorylated and misfolded tau protein, are also a pathologic hallmark of AD [144, 145]. While accumulation of each protein is likely vital to AD initiation and progression, evidence suggests that the interplay between Aβ and tau is complex and likely drives neuronal disease [146]. For example, while Aβ has been shown to trigger pathologic tau modifications, other evidence suggests that tau also enhances the toxicity of Aβ [146150]. Recent studies have more closely associated tau pathology to cognitive decline in AD [151153]. This has brought increased attention to the tau hypothesis, which theorizes that tau is the initiator of downstream AD neurodegeneration via pathologic modifications, like hyperphosphorylation, that make the protein prone to aggregation and prion-like propagation [7, 8, 154]. However, if tau plays a primary role in AD pathogenesis, that does not preclude infectious agents, like herpesviruses, from contributing to AD. The few studies investigating the effects of viral infection on tau have shown a transient tau increase in neurons after HSV-1 infection and HSV-1 induced phosphorylation [155157]. Additionally, viral glycoproteins may promote release of extracellular vesicles and the transfer of misfolded tau proteins between cells [129]. As the tau hypothesis continues to be placed near the forefront of theories of AD pathogenesis, further research on the effects of viral infection on tau aggregation and propagation is warranted, as infectious agents may affect the accumulation of multiple neurodegenerative proteins involved in AD.

Alpha-synuclein (α-syn) is a synapse-associated protein that can abnormally accumulate, forming Lewy bodies, and is linked to a group of neurodegenerative diseases called synucleinopathies, which includes Parkinson’s disease, multiple system atrophy, and dementia with Lewy bodies [158161]. Multiple studies have demonstrated that in addition to Aβ and tau, α-syn pathology can also be found in over 50 % of AD patients, suggesting that α-syn may also be involved in AD pathogenesis or that many AD patients have other neurodegenerative disease co-morbidities [162165]. Few studies thus far have investigated the relationship between viral infection and α-syn; however, those studies suggest a possible role for α-syn as an inhibitor of viral infection, similar to Aβ [166169]. The effects of herpesvirus infection on α-syn aggregation in AD patients and other neurodegenerative diseases has not yet been investigated, but this research is likely necessary for a deeper understanding of the role of microbes in AD and dementia pathogenesis.

In vivo studies can be useful in the investigation of the effects of herpesviruses in AD, as the capacity of viruses to influence disease pathology in a more complete animal model system can provide important evidence of causation [27]. Transgenic mouse models that result in Aβ plaque and tau tangle formation are the most common animal models of AD [170, 171]. However, many experimental treatments that are successful in pre-clinical studies utilizing rodent models fail in clinical human trials, which suggests that animal models may not be good surrogates of human AD or better animal models that more closely mimic human clinical AD are needed for better result translation [170, 172, 173]. While most of the in vivo research on the role of herpesviruses in AD has utilized rodent models, other animal models may better replicate the effects of viral infection on Aβ and related protein accumulation in AD. Both spontaneous and induced models of AD have been described in nonhuman primates [174]. While these models also have some shortcomings when compared to human AD, they may provide an essential link between pre-clinical studies and clinical trial results due to their phylogenetic relationship to humans and their similarities in aging pathologies [174, 175]. Non-human primates can be infected with human herpesviruses of interest in AD research, such as HHV-6A and HHV-6B, and they also harbor closely related viruses such as rhesus lymphocryptovirus, Callitrichine herpesvirus 3, and Simian varicella virus [176179]. Aged dogs also naturally develop AD-related pathology, including Aβ plaques, and a clinically similar syndrome called canine cognitive dysfunction [170, 180, 181]. The continued accumulation of a large number of longitudinal biosamples through the NIH-funded Canine Longevity Consortium and the Dog Aging Project provides a plethora of opportunities for investigating AD-related biomarkers and phenotypes in the canine model [182]. These longitudinal studies in an animal model relevant to human AD may provide additional insights into the association between risk factors like viral infection and cognitive decline. The utility of more translationally relevant animal models may strengthen the evidence for viral infection as a factor in AD.

The countless recent failures of later stage clinical trials targeting Aβ highlights the necessity for different therapeutic strategies in the treatment of AD [12, 13]. Based on the multifactorial nature of AD, polypharmaceutical strategies are likely to be most effective in treating AD [183]. If the infectious hypothesis of AD proves true, antiviral and antimicrobial agents, in combination with therapeutics targeting the overproduction of Aβ, may provide opportunities to dampen neuroinflammation secondary to microbial infection and reduce the Aβ burden [184]. A recent study based in Sweden showed that antiviral treatment was associated with a decreased risk of dementia, while herpesvirus infection without antiviral treatment resulted in an increased risk of dementia [121]. A population study in Taiwan revealed reduced risk of dementia in HSV-1 infected patients who received anti-herpetic medications [185]. A clinical trial of the antiviral valacyclovir in AD patients who are positive for HSV-1 or HSV-2 has been initiated [186]. A 4-week trial in early-stage AD patients determined a tolerable and safe high-dose treatment of valacyclovir; however, no significant change in biomarkers was noted in that time period [187]. The results of these initial clinical studies, coupled with continued in vitro and in vivo investigation of the role of herpesviruses in AD, will help dictate the future utility of antiviral strategies in AD treatment and prevention, and the fulfillment of arguably the most important criteria for herpesviruses as causative agents in AD: that elimination or reduction of the virus changes disease outcome [27, 188].

Summary

Based on the current literature and evidence, the exact role of herpesviruses, and infectious agents in general, in AD is uncertain and not definitive. While it is possible that herpesviruses may play a role in some AD patients, whether they can function as an initiator of the disease cascade, drivers of disease progression, or both remains unclear. As the population of AD-afflicted individuals continues to grow and research-related support in the field increases, further investigation is warranted to better understand the contributions of microbes to AD pathogenesis. When the relationship between pathogens such as herpesviruses and AD becomes clearer, it will help dictate potential future avenues for disease prevention and treatment, such as antiherpetic therapeutics.


Corresponding author: Steven Jacobson, PhD, Senior Investigator, Viral Immunology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Building 10, Room 5C103, 10 Center Drive 20892-1400, Bethesda, MD, USA, E-mail:

Award Identifier / Grant number: Intramural Research Program

Funding source: National Institute of Neurological Disorders and Stroke

Award Identifier / Grant number: Unassigned

Funding source: Purdue University

Award Identifier / Grant number: Unassigned

Acknowledgments

SP is a NIH Comparative Biomedical Scientist Training Program Fellow, in partnership with the National Institute of Neurological Disorders and Stroke and Purdue University. Figure 1 was created with BioRender.com.

  1. Research funding: SP, AT, and SJ are supported by the Intramural Research Program, National Institute of Neurological Disorders and Stroke, National Institutes of Health. AT is also supported by the National Institutes of Health Undergraduate Scholarship Program. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.

  2. Author contributions: SP and SJ conceptualized the work. SP, AT, and SJ contributed to the manuscript writing and revisions. All authors have accepted responsibility for the entire content of this manuscript and approved its submission.

  3. Competing interests: The authors declare that this manuscript was written in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. The authors state no conflict of interest.

  4. Informed consent: Not applicable.

  5. Ethical approval: Not applicable.

References

1. Scheltens, P, De Strooper, B, Kivipelto, M, Holstege, H, Chételat, G, Teunissen, CE, et al.. Alzheimer’s disease. Lancet 2021;397:1577–90. https://doi.org/10.1016/s0140-6736(20)32205-4.Search in Google Scholar

2. Ferrari, C, Sorbi, S. The complexity of Alzheimer’s disease: an evolving puzzle. Physiol Rev 2021;101:1047–81. https://doi.org/10.1152/physrev.00015.2020.Search in Google Scholar PubMed

3. Knopman, DS, Amieva, H, Petersen, RC, Chételat, G, Holtzman, DM, Hyman, BT, et al.. Alzheimer disease. Nat Rev Dis Prim 2021;7:33. https://doi.org/10.1038/s41572-021-00269-y.Search in Google Scholar PubMed PubMed Central

4. Iqbal, K, Grundke-Iqbal, I. Alzheimer’s disease, a multifactorial disorder seeking multitherapies. Alzheimer’s Dement 2010;6:420–4. https://doi.org/10.1016/j.jalz.2010.04.006.Search in Google Scholar PubMed PubMed Central

5. Breijyeh, Z, Karaman, R. Comprehensive review on Alzheimer’s disease: causes and treatment. Molecules 2020;25:5789. https://doi.org/10.3390/molecules25245789.Search in Google Scholar PubMed PubMed Central

6. DeTure, MA, Dickson, DW. The neuropathological diagnosis of Alzheimer’s disease. Mol Neurodegener 2019;14:32. https://doi.org/10.1186/s13024-019-0333-5.Search in Google Scholar PubMed PubMed Central

7. Du, X, Wang, X, Geng, M. Alzheimer’s disease hypothesis and related therapies. Transl Neurodegener 2018;7:2. https://doi.org/10.1186/s40035-018-0107-y.Search in Google Scholar PubMed PubMed Central

8. Liu, P-P, Xie, Y, Meng, X-Y, Kang, J-S. History and progress of hypotheses and clinical trials for Alzheimer’s disease. Signal Transduction Targeted Ther 2019;4:29. https://doi.org/10.1038/s41392-019-0063-8.Search in Google Scholar PubMed PubMed Central

9. Bondi, MW, Edmonds, EC, Salmon, DP. Alzheimer’s disease: past, present, and future. J Int Neuropsychol Soc 2017;23:818–31. https://doi.org/10.1017/s135561771700100x.Search in Google Scholar PubMed PubMed Central

10. Fan, L, Mao, C, Hu, X, Zhang, S, Yang, Z, Hu, Z, et al.. New insights into the pathogenesis of alzheimer’s disease. Front Neurol 2020;10:1312. https://doi.org/10.3389/fneur.2019.01312.Search in Google Scholar PubMed PubMed Central

11. Kametani, F, Hasegawa, M. Reconsideration of amyloid hypothesis and tau hypothesis in alzheimer’s disease. Front Neurosci 2018;12:25. https://doi.org/10.3389/fnins.2018.00025.Search in Google Scholar PubMed PubMed Central

12. Oxford, AE, Stewart, ES, Rohn, TT. Clinical trials in alzheimer’s disease: a hurdle in the path of remedy. Int J Alzheimer’s Dis 2020;2020:5380346. https://doi.org/10.1155/2020/5380346.Search in Google Scholar PubMed PubMed Central

13. Yiannopoulou, KG, Anastasiou, AI, Zachariou, V, Pelidou, S-H. Reasons for failed trials of disease-modifying treatments for alzheimer disease and their contribution in recent research. Biomedicines 2019;7:97. https://doi.org/10.3390/biomedicines7040097.Search in Google Scholar PubMed PubMed Central

14. Yiannopoulou, KG, Papageorgiou, SG. Current and future treatments in alzheimer disease: an update. J Cent Nerv Syst Dis 2020;12:1179573520907397. https://doi.org/10.1177/1179573520907397.Search in Google Scholar PubMed PubMed Central

15. Jeremic, D, Jiménez-Díaz, L, Navarro-López, JD. Past, present and future of therapeutic strategies against amyloid-β peptides in Alzheimer’s disease: a systematic review. Ageing Res Rev 2021;72:101496. https://doi.org/10.1016/j.arr.2021.101496.Search in Google Scholar PubMed

16. Fulop, T, Witkowski, JM, Bourgade, K, Khalil, A, Zerif, E, Larbi, A, et al.. Can an infection hypothesis explain the beta amyloid hypothesis of alzheimer’s disease? Front Aging Neurosci 2018;10:224. https://doi.org/10.3389/fnagi.2018.00224.Search in Google Scholar PubMed PubMed Central

17. Vojtechova, I, Machacek, T, Kristofikova, Z, Stuchlik, A, Petrasek, T. Infectious origin of Alzheimer’s disease: amyloid beta as a component of brain antimicrobial immunity. PLoS Pathog 2022;18:e1010929. https://doi.org/10.1371/journal.ppat.1010929.Search in Google Scholar PubMed PubMed Central

18. Allnutt, MA, Jacobson, S. Do herpesviruses play a role in Alzheimer’s disease pathogenesis? Drug Discovery Today Dis. Models 2020;32:21–6. https://doi.org/10.1016/j.ddmod.2019.10.006.Search in Google Scholar

19. Sochocka, M, Zwolińska, K, Leszek, J. The infectious etiology of alzheimer’s disease. Curr Neuropharmacol 2017;15:996–1009. https://doi.org/10.2174/1570159x15666170313122937.Search in Google Scholar

20. Chen, F, Chen, Y, Wang, Y, Ke, Q, Cui, L. The COVID-19 pandemic and Alzheimer’s disease: mutual risks and mechanisms. Transl Neurodegener 2022;11:40. https://doi.org/10.1186/s40035-022-00316-y.Search in Google Scholar PubMed PubMed Central

21. Wang, L, Davis, PB, Volkow, ND, Berger, NA, Kaelber, DC, Xu, R. Association of COVID-19 with new-onset alzheimer’s disease. J Alzheimer’s Dis 2022;89:411–4. https://doi.org/10.3233/jad-220717.Search in Google Scholar PubMed PubMed Central

22. Fulop, T, Witkowski, JM, Larbi, A, Khalil, A, Herbein, G, Frost, EH. Does HIV infection contribute to increased beta-amyloid synthesis and plaque formation leading to neurodegeneration and Alzheimer’s disease? J Neurovirol 2019;25:634–47. https://doi.org/10.1007/s13365-019-00732-3.Search in Google Scholar PubMed

23. Hategan, A, Bianchet, MA, Steiner, J, Karnaukhova, E, Masliah, E, Fields, A, et al.. HIV Tat protein and amyloid-β peptide form multifibrillar structures that cause neurotoxicity. Nat Struct Mol Biol 2017;24:379–86. https://doi.org/10.1038/nsmb.3379.Search in Google Scholar PubMed PubMed Central

24. Chiu, WC, Tsan, YT, Tsai, SL, Chang, CJ, Wang, JD, Chen, PC. Hepatitis C viral infection and the risk of dementia. Eur J Neurol 2014;21:1068–e59. https://doi.org/10.1111/ene.12317.Search in Google Scholar PubMed

25. Andrea, C, Luigi, C, Mattia, T, Giancarlo, O, Daniele, I, Cristiana, A, et al.. Alzheimer dementia in people living with HIV. Neurol: Clin Pract 2021;11:e627. https://doi.org/10.1212/cpj.0000000000001060.Search in Google Scholar PubMed PubMed Central

26. Rizzo, R. Controversial role of herpesviruses in Alzheimer’s disease. PLoS Pathog 2020;16:e1008575. https://doi.org/10.1371/journal.ppat.1008575.Search in Google Scholar PubMed PubMed Central

27. Komaroff, A, Pellett, P, Jacobson, S. Human herpesviruses 6A and 6B in brain diseases: association versus causation. Clin Microbiol Rev 2020;34. https://doi.org/10.1128/cmr.00143-20.Search in Google Scholar

28. Wang, H, Kulas, JA, Wang, C, Holtzman, DM, Ferris, HA, Hansen, SB. Regulation of beta-amyloid production in neurons by astrocyte-derived cholesterol. Proc Natl Acad Sci 2021;118:e2102191118. https://doi.org/10.1073/pnas.2102191118.Search in Google Scholar PubMed PubMed Central

29. Chen, G-F, Xu, T-H, Yan, Y, Zhou, Y-R, Jiang, Y, Melcher, K, et al.. Amyloid beta: structure, biology and structure-based therapeutic development. Acta Pharmacol Sin 2017;38:1205–35. https://doi.org/10.1038/aps.2017.28.Search in Google Scholar PubMed PubMed Central

30. Sun, X, Chen, W-D, Wang, Y-D. β-Amyloid: the key peptide in the pathogenesis of Alzheimer’s disease. Front Pharmacol 2015;6:221. https://doi.org/10.3389/fphar.2015.00221.Search in Google Scholar PubMed PubMed Central

31. Tarasoff-Conway, JM, Carare, RO, Osorio, RS, Glodzik, L, Butler, T, Fieremans, E, et al.. Clearance systems in the brain-implications for Alzheimer disease. Nat Rev Neurol 2015;11:457–70. https://doi.org/10.1038/nrneurol.2015.119.Search in Google Scholar PubMed PubMed Central

32. Brothers, HM, Gosztyla, ML, Robinson, SR. The physiological roles of amyloid-β peptide hint at new ways to treat alzheimer’s disease. Front Aging Neurosci 2018;10:118. https://doi.org/10.3389/fnagi.2018.00118.Search in Google Scholar PubMed PubMed Central

33. Tharp, WG, Sarkar, IN. Origins of amyloid-β. BMC Genom 2013;14:290. https://doi.org/10.1186/1471-2164-14-290.Search in Google Scholar PubMed PubMed Central

34. Moir, RD, Tanzi, RE. Low evolutionary selection pressure in senescence does not explain the persistence of Aβ in the vertebrate genome. Front Aging Neurosci 2019;11:70. https://doi.org/10.3389/fnagi.2019.00070.Search in Google Scholar PubMed PubMed Central

35. Soscia, SJ, Kirby, JE, Washicosky, KJ, Tucker, SM, Ingelsson, M, Hyman, B, et al.. The alzheimer’s disease-associated amyloid β-protein is an antimicrobial peptide. PLoS One 2010;5:e9505. https://doi.org/10.1371/journal.pone.0009505.Search in Google Scholar PubMed PubMed Central

36. Gosztyla, ML, Brothers, HM, Robinson, SR. Alzheimer’s amyloid-β is an antimicrobial peptide: a review of the evidence. J Alzheimer’s Dis 2018;62:1495–506. https://doi.org/10.3233/jad-171133.Search in Google Scholar

37. Seaks, CE, Wilcock, DM. Infectious hypothesis of Alzheimer disease. PLoS Pathog 2020;16:e1008596. https://doi.org/10.1371/journal.ppat.1008596.Search in Google Scholar PubMed PubMed Central

38. Kayed, R. Infectious etiology and amyloidosis in Alzheimer’s disease: the puzzle continues. J Biol Chem 2021;297:100936. https://doi.org/10.1016/j.jbc.2021.100936.Search in Google Scholar PubMed PubMed Central

39. Hur, JY, Frost, GR, Wu, X, Crump, C, Pan, SJ, Wong, E, et al.. The innate immunity protein IFITM3 modulates γ-secretase in Alzheimer’s disease. Nature 2020;586:735–40. https://doi.org/10.1038/s41586-020-2681-2.Search in Google Scholar PubMed PubMed Central

40. Itzhaki, RF, Golde, TE, Heneka, MT, Readhead, B. Do infections have a role in the pathogenesis of Alzheimer disease? Nat Rev Neurol 2020;16:193–7. https://doi.org/10.1038/s41582-020-0323-9.Search in Google Scholar PubMed

41. Patrick, KL, Bell, SL, Weindel, CG, Watson, RO. Exploring the “multiple-hit hypothesis” of neurodegenerative disease: bacterial infection comes up to bat. Front Cell Infect Microbiol 2019;9:138. https://doi.org/10.3389/fcimb.2019.00138.Search in Google Scholar PubMed PubMed Central

42. Zhu, X, Lee, HG, Perry, G, Smith, MA. Alzheimer disease, the two-hit hypothesis: an update. Biochim Biophys Acta 2007;1772:494–502. https://doi.org/10.1016/j.bbadis.2006.10.014.Search in Google Scholar PubMed

43. Armstrong, RA. Risk factors for Alzheimer’s disease. Folia Neuropathol 2019;57:87–105. https://doi.org/10.5114/fn.2019.85929.Search in Google Scholar PubMed

44. Fülöp, T, Itzhaki, RF, Balin, BJ, Miklossy, J, Barron, AE. Role of microbes in the development of Alzheimer’s disease: state of the art – an international symposium presented at the 2017 IAGG congress in san francisco. Front Genet 2018;9:362. https://doi.org/10.3389/fgene.2018.00362.Search in Google Scholar PubMed PubMed Central

45. Desfarges, S, Ciuffi, A. Viral integration and consequences on host gene expression. Viruses: Essential Agents of Life 2012;25:147–75. https://doi.org/10.1007/978-94-007-4899-6_7.Search in Google Scholar

46. Readhead, B, Haure-Mirande, J-V, Funk, CC, Richards, MA, Shannon, P, Haroutunian, V, et al.. Multiscale analysis of independent Alzheimer’s cohorts finds disruption of molecular, genetic, and clinical networks by human herpesvirus. Neuron 2018;99:64–82.e7. https://doi.org/10.1016/j.neuron.2018.05.023.Search in Google Scholar PubMed PubMed Central

47. Tanaka, S, Nagashima, H. Establishment of an Alzheimer’s disease model with latent herpesvirus infection using PS2 and Tg2576 double transgenic mice. Exp Anim 2018;67:185–92. https://doi.org/10.1538/expanim.17-0066.Search in Google Scholar PubMed PubMed Central

48. Laval, K, Enquist, LW. The potential role of herpes simplex virus type 1 and neuroinflammation in the pathogenesis of alzheimer’s disease. Front Neurol 2021;12:658695. https://doi.org/10.3389/fneur.2021.658695.Search in Google Scholar PubMed PubMed Central

49. Kinney, JW, Bemiller, SM, Murtishaw, AS, Leisgang, AM, Salazar, AM, Lamb, BT. Inflammation as a central mechanism in Alzheimer’s disease. Alzheimer’s Dementia 2018;4:575–90. https://doi.org/10.1016/j.trci.2018.06.014.Search in Google Scholar PubMed PubMed Central

50. Li, L, Mao, S, Wang, J, Ding, X, Zen, JY. Viral infection and neurological disorders—potential role of extracellular nucleotides in neuroinflammation. ExRNA 2019;1:26. https://doi.org/10.1186/s41544-019-0031-z.Search in Google Scholar

51. Filgueira, L, Larionov, A, Lannes, N. The influence of virus infection on microglia and accelerated brain aging. Cells 2021;10:1836. https://doi.org/10.3390/cells10071836.Search in Google Scholar PubMed PubMed Central

52. Leng, F, Edison, P. Neuroinflammation and microglial activation in Alzheimer disease: where do we go from here? Nat Rev Neurol 2021;17:157–72. https://doi.org/10.1038/s41582-020-00435-y.Search in Google Scholar PubMed

53. Cai, Z, Hussain, MD, Yan, LJ. Microglia, neuroinflammation, and beta-amyloid protein in Alzheimer’s disease. Int J Neurosci 2014;124:307–21. https://doi.org/10.3109/00207454.2013.833510.Search in Google Scholar PubMed

54. Minter, MR, Taylor, JM, Crack, PJ. The contribution of neuroinflammation to amyloid toxicity in Alzheimer’s disease. J Neurochem 2016;136:457–74. https://doi.org/10.1111/jnc.13411.Search in Google Scholar PubMed

55. Liddelow, SA, Guttenplan, KA, Clarke, LE, Bennett, FC, Bohlen, CJ, Schirmer, L, et al.. Neurotoxic reactive astrocytes are induced by activated microglia. Nature 2017;541:481–7. https://doi.org/10.1038/nature21029.Search in Google Scholar PubMed PubMed Central

56. Naughton, SX, Raval, U, Pasinetti, GM. The viral hypothesis in alzheimer’s disease: novel insights and pathogen-based biomarkers. J Pers Med 2020;10:74. https://doi.org/10.3390/jpm10030074.Search in Google Scholar PubMed PubMed Central

57. Romeo, MA, Faggioni, A, Cirone, M. Could autophagy dysregulation link neurotropic viruses to Alzheimer’s disease? Neural Regener Res 2019;14:1503–6. https://doi.org/10.4103/1673-5374.253508.Search in Google Scholar PubMed PubMed Central

58. Dobricic, V, Schilling, M, Schulz, J, Zhu, L-S, Zhou, C-W, Fuß, J, et al.. Differential microRNA expression analyses across two brain regions in Alzheimer’s disease. Transl Psychiatry 2022;12:352. https://doi.org/10.1038/s41398-022-02108-4.Search in Google Scholar PubMed PubMed Central

59. Wen, Q, Verheijen, M, Wittens, MMJ, Czuryło, J, Engelborghs, S, Hauser, D, et al.. Lead-exposure associated miRNAs in humans and Alzheimer’s disease as potential biomarkers of the disease and disease processes. Sci Rep 2022;12:15966. https://doi.org/10.1038/s41598-022-20305-5.Search in Google Scholar PubMed PubMed Central

60. Wainberg, M, Luquez, T, Koelle, DM, Readhead, B, Johnston, C, Darvas, M, et al.. The viral hypothesis: how herpesviruses may contribute to Alzheimer’s disease. Mol Psychiatry 2021;26:5476–80. https://doi.org/10.1038/s41380-021-01138-6.Search in Google Scholar PubMed PubMed Central

61. Balin, BJ, Hudson, AP. Herpes viruses and Alzheimer’s disease: new evidence in the debate. Lancet Neurol 2018;17:839–41. https://doi.org/10.1016/s1474-4422(18)30316-8.Search in Google Scholar

62. Piacentini, R, De Chiara, G, Li Puma, DD, Ripoli, C, Marcocci, ME, Garaci, E, et al.. HSV-1 and Alzheimer’s disease: more than a hypothesis. Front Pharmacol 2014;5:97. https://doi.org/10.3389/fphar.2014.00097.Search in Google Scholar PubMed PubMed Central

63. Itzhaki, RF. Overwhelming evidence for a major role for herpes simplex virus type 1 (HSV1) in Alzheimer’s disease (AD). underwhelming evidence against. Vaccines 2021;9:679. https://doi.org/10.3390/vaccines9060679.Search in Google Scholar PubMed PubMed Central

64. Looker, KJ, Magaret, AS, May, MT, Turner, KME, Vickerman, P, Gottlieb, SL, et al.. Global and regional estimates of prevalent and incident herpes simplex virus type 1 infections in 2012. PLoS One 2015;10:e0140765. https://doi.org/10.1371/journal.pone.0140765.Search in Google Scholar PubMed PubMed Central

65. Duarte, LF, Farías, MA, Álvarez, DM, Bueno, SM, Riedel, CA, González, PA. Herpes simplex virus type 1 infection of the central nervous system: insights into proposed interrelationships with neurodegenerative disorders. Front Cell Neurosci 2019;13:46. https://doi.org/10.3389/fncel.2019.00046.Search in Google Scholar PubMed PubMed Central

66. Meyding-Lamadé, U, Strank, C. Herpesvirus infections of the central nervous system in immunocompromised patients. Ther Adv Neurol Disord 2012;5:279–96. https://doi.org/10.1177/1756285612456234.Search in Google Scholar PubMed PubMed Central

67. Ball, MJ. Limbic predilection in alzheimer dementia: is reactivated herpesvirus involved? Can J Neurol Sci/J Can Sci Neurol 1982;9:303–6. https://doi.org/10.1017/s0317167100044115.Search in Google Scholar PubMed

68. Wozniak, MA, Mee, AP, Itzhaki, RF. Herpes simplex virus type 1 DNA is located within Alzheimer’s disease amyloid plaques. J Pathol 2009;217:131–8. https://doi.org/10.1002/path.2449.Search in Google Scholar PubMed

69. Eimer, WA, Vijaya Kumar, DK, Navalpur Shanmugam, NK, Rodriguez, AS, Mitchell, T, Washicosky, KJ, et al.. Alzheimer’s disease-associated β-amyloid is rapidly seeded by herpesviridae to protect against brain infection. Neuron 2018;99:56–63.e3. https://doi.org/10.1016/j.neuron.2018.06.030.Search in Google Scholar PubMed PubMed Central

70. Ezzat, K, Pernemalm, M, Pålsson, S, Roberts, TC, Järver, P, Dondalska, A, et al.. The viral protein corona directs viral pathogenesis and amyloid aggregation. Nat Commun 2019;10:2331. https://doi.org/10.1038/s41467-019-10192-2.Search in Google Scholar PubMed PubMed Central

71. Cairns, DM, Rouleau, N, Parker, RN, Walsh, KG, Gehrke, L, Kaplan, DL. A 3D human brain–like tissue model of herpes-induced Alzheimer’s disease. Sci Adv 2020;6:eaay8828. https://doi.org/10.1126/sciadv.aay8828.Search in Google Scholar PubMed PubMed Central

72. Marcocci, ME, Napoletani, G, Protto, V, Kolesova, O, Piacentini, R, Li Puma, DD, et al.. Herpes simplex virus-1 in the brain: the dark side of a sneaky infection. Trends Microbiol 2020;28:808–20. https://doi.org/10.1016/j.tim.2020.03.003.Search in Google Scholar PubMed

73. Piacentini, R, Civitelli, L, Ripoli, C, Marcocci, ME, De Chiara, G, Garaci, E, et al.. HSV-1 promotes Ca2+ -mediated APP phosphorylation and Aβ accumulation in rat cortical neurons. Neurobiol Aging 2011;32:2323.e13–26. https://doi.org/10.1016/j.neurobiolaging.2010.06.009.Search in Google Scholar PubMed

74. Bocharova, O, Pandit, NP, Molesworth, K, Fisher, A, Mychko, O, Makarava, N, et al.. Alzheimer’s disease-associated β-amyloid does not protect against herpes simplex virus 1 infection in the mouse brain. J Biol Chem 2021;297:100845. https://doi.org/10.1016/j.jbc.2021.100845.Search in Google Scholar PubMed PubMed Central

75. De Chiara, G, Piacentini, R, Fabiani, M, Mastrodonato, A, Marcocci, ME, Limongi, D, et al.. Recurrent herpes simplex virus-1 infection induces hallmarks of neurodegeneration and cognitive deficits in mice. PLoS Pathog 2019;15:e1007617. https://doi.org/10.1371/journal.ppat.1007617.Search in Google Scholar PubMed PubMed Central

76. Toscano, ECB, Sousa, L, Lima, GK, Mesquita, LA, Vilela, MC, Rodrigues, DH, et al.. Neuroinflammation is associated with reduced SOCS2 and SOCS3 expression during intracranial HSV-1 infection. Neurosci Lett 2020;736:135295. https://doi.org/10.1016/j.neulet.2020.135295.Search in Google Scholar PubMed

77. Yong, SJ, Yong, MH, Teoh, SL, Soga, T, Parhar, I, Chew, J, et al.. The hippocampal vulnerability to herpes simplex virus type I infection: relevance to alzheimer’s disease and memory impairment. Front Cell Neurosci 2021;15:695738. https://doi.org/10.3389/fncel.2021.695738.Search in Google Scholar PubMed PubMed Central

78. Hill, JM, Zhao, Y, Clement, C, Neumann, DM, Lukiw, WJ. HSV-1 infection of human brain cells induces miRNA-146a and Alzheimer-type inflammatory signaling. Neuroreport 2009;20:1500–5. https://doi.org/10.1097/wnr.0b013e3283329c05.Search in Google Scholar

79. Marques, CP, Cheeran, MC, Palmquist, JM, Hu, S, Urban, SL, Lokensgard, JR. Prolonged microglial cell activation and lymphocyte infiltration following experimental herpes encephalitis. J Immunol 2008;181:6417–26. https://doi.org/10.4049/jimmunol.181.9.6417.Search in Google Scholar PubMed PubMed Central

80. Cokarić Brdovčak, M, Zubković, A, Jurak, I. Herpes simplex virus 1 deregulation of host MicroRNAs. Non-Coding RNA 2018;4:36. https://doi.org/10.3390/ncrna4040036.Search in Google Scholar PubMed PubMed Central

81. Linard, M, Letenneur, L, Garrigue, I, Doize, A, Dartigues, JF, Helmer, C. Interaction between APOE4 and herpes simplex virus type 1 in Alzheimer’s disease. Alzheimer’s Dement 2020;16:200–8. https://doi.org/10.1002/alz.12008.Search in Google Scholar PubMed

82. Itzhaki, RF, Lin, W-R, Shang, D, Wilcock, GK, Faragher, B, Jamieson, GA. Herpes simplex virus type 1 in brain and risk of Alzheimer’s disease. Lancet 1997;349:241–4. https://doi.org/10.1016/s0140-6736(96)10149-5.Search in Google Scholar

83. Murphy, MJ, Fani, L, Ikram, MK, Ghanbari, M, Ikram, MA. Herpes simplex virus 1 and the risk of dementia: a population-based study. Sci Rep 2021;11:8691. https://doi.org/10.1038/s41598-021-87963-9.Search in Google Scholar PubMed PubMed Central

84. Zhang, Y, Qu, J, Luo, L, Xu, Z, Zou, X. Multigenomics reveals the causal effect of herpes simplex virus in Alzheimer’s disease: a two-sample mendelian randomization study. Front Genet 2022;12:773725. https://doi.org/10.3389/fgene.2021.773725.Search in Google Scholar PubMed PubMed Central

85. Braun, DK, Dominguez, G, Pellett, PE. Human herpesvirus 6. Clin Microbiol Rev 1997;10:521–67. https://doi.org/10.1128/cmr.10.3.521.Search in Google Scholar

86. Ablashi, D, Agut, H, Alvarez-Lafuente, R, Clark, DA, Dewhurst, S, DiLuca, D, et al.. Classification of HHV-6A and HHV-6B as distinct viruses. Arch Virol 2014;159:863–70. https://doi.org/10.1007/s00705-013-1902-5.Search in Google Scholar PubMed PubMed Central

87. Yao, K, Crawford, JR, Komaroff, AL, Ablashi, DV, Jacobson, S. Review part 2: human herpesvirus-6 in central nervous system diseases. J Med Virol 2010;82:1669–78. https://doi.org/10.1002/jmv.21861.Search in Google Scholar PubMed PubMed Central

88. Leibovitch, EC, Jacobson, S. Viruses in chronic progressive neurologic disease. Mult Scler J 2018;24:48–52. https://doi.org/10.1177/1352458517737392.Search in Google Scholar PubMed PubMed Central

89. Epstein, LG, Shinnar, S, Hesdorffer, DC, Nordli, DR, Hamidullah, A, Benn, EK, et al.. Human herpesvirus 6 and 7 in febrile status epilepticus: the FEBSTAT study. Epilepsia 2012;53:1481–8. https://doi.org/10.1111/j.1528-1167.2012.03542.x.Search in Google Scholar PubMed PubMed Central

90. Kawamura, Y, Sugata, K, Ihira, M, Mihara, T, Mutoh, T, Asano, Y, et al.. Different characteristics of human herpesvirus 6 encephalitis between primary infection and viral reactivation. J Clin Virol 2011;51:12–9. https://doi.org/10.1016/j.jcv.2011.02.002.Search in Google Scholar PubMed

91. Santpere, G, Telford, M, Andrés-Benito, P, Navarro, A, Ferrer, I. The presence of human herpesvirus 6 in the brain in health and disease. Biomolecules 2020;10:1520. https://doi.org/10.3390/biom10111520.Search in Google Scholar PubMed PubMed Central

92. Readhead, B, Haure-Mirande, J-V, Mastroeni, D, Audrain, M, Fanutza, T, Kim, SH, et al.. miR155 regulation of behavior, neuropathology, and cortical transcriptomics in Alzheimer’s disease. Acta Neuropathol 2020;140:295–315. https://doi.org/10.1007/s00401-020-02185-z.Search in Google Scholar PubMed PubMed Central

93. Romanescu, C, Schreiner, TG, Mukovozov, I. The role of human herpesvirus 6 infection in alzheimer’s disease pathogenicity-A theoretical mosaic. J Clin Med 2022;11:3061. https://doi.org/10.3390/jcm11113061.Search in Google Scholar PubMed PubMed Central

94. Jeong, H-H, Liu, Z. Are HHV-6A and HHV-7 really more abundant in Alzheimer’s disease? Neuron 2019;104:1034–5. https://doi.org/10.1016/j.neuron.2019.11.009.Search in Google Scholar PubMed

95. Readhead, B, Haure-Mirande, J-V, Ehrlich, ME, Gandy, S, Dudley, JT. Clarifying the potential role of microbes in Alzheimer’s disease. Neuron 2019;104:1036–7. https://doi.org/10.1016/j.neuron.2019.11.008.Search in Google Scholar PubMed PubMed Central

96. Readhead, B, Haure-Mirande, J-V, Ehrlich, ME, Gandy, S, Dudley, JT. Further evidence of increased human Herpesvirus in Alzheimer’s disease. bioRxiv 2019:858050.10.1101/858050Search in Google Scholar

97. Chorlton, SD. Reanalysis of Alzheimer’s brain sequencing data reveals absence of purported HHV6A and HHV7. J Bioinf Comput Biol 2020;18:2050012. https://doi.org/10.1142/s0219720020500122.Search in Google Scholar PubMed

98. Allnutt, MA, Johnson, K, Bennett, DA, Connor, SM, Troncoso, JC, Pletnikova, O, et al.. Human herpesvirus 6 detection in Alzheimer’s disease cases and controls across multiple cohorts. Neuron 2020;105:1027–35.e2. https://doi.org/10.1016/j.neuron.2019.12.031.Search in Google Scholar PubMed PubMed Central

99. Bigley, TM, Xiong, M, Ali, M, Chen, Y, Wang, C, Serrano, JR, et al.. Murine roseolovirus does not accelerate amyloid-β pathology and human roseoloviruses are not over-represented in Alzheimer disease brains. Mol Neurodegener 2022;17:10. https://doi.org/10.1186/s13024-021-00514-8.Search in Google Scholar PubMed PubMed Central

100. Costa, SAC, Madsen, H, Brown, JR. Shared molecular signatures across neurodegenerative diseases and herpes virus infections highlights potential mechanisms for maladaptive innate immune responses. Sci Rep 2019;9:8795. https://doi.org/10.1038/s41598-019-45129-8.Search in Google Scholar PubMed PubMed Central

101. Carter, CJ. Alzheimer’s disease: a pathogenetic autoimmune disorder caused by herpes simplex in a gene-dependent manner. Int J Alzheimer’s Dis 2010;2010:140539. https://doi.org/10.4061/2010/140539.Search in Google Scholar PubMed PubMed Central

102. Agostini, S, Mancuso, R, Baglio, F, Cabinio, M, Hernis, A, Guerini, FR, et al.. Lack of evidence for a role of HHV-6 in the pathogenesis of Alzheimer’s disease. J Alzheimer’s Dis 2016;49:229–35. https://doi.org/10.3233/jad-150464.Search in Google Scholar

103. Westman, G, Blomberg, J, Yun, Z, Lannfelt, L, Ingelsson, M, Eriksson, B-M. Decreased HHV-6 IgG in Alzheimer’s disease. Front Neurol 2017;8:40. https://doi.org/10.3389/fneur.2017.00040.Search in Google Scholar PubMed PubMed Central

104. Carbone, I, Lazzarotto, T, Ianni, M, Porcellini, E, Forti, P, Masliah, E, et al.. Herpes virus in Alzheimer’s disease: relation to progression of the disease. Neurobiol Aging 2014;35:122–9. https://doi.org/10.1016/j.neurobiolaging.2013.06.024.Search in Google Scholar PubMed

105. Bortolotti, D, Gentili, V, Rotola, A, Caselli, E, Rizzo, R. HHV-6A infection induces amyloid-beta expression and activation of microglial cells. Alzheimer’s Res Ther 2019;11:104. https://doi.org/10.1186/s13195-019-0552-6.Search in Google Scholar PubMed PubMed Central

106. Bar-Or, A, Pender, MP, Khanna, R, Steinman, L, Hartung, HP, Maniar, T, et al.. Epstein-barr virus in multiple sclerosis: theory and emerging immunotherapies. Trends Mol Med 2020;26:296–310. https://doi.org/10.1016/j.molmed.2019.11.003.Search in Google Scholar PubMed PubMed Central

107. Lanz, TV, Brewer, RC, Ho, PP, Moon, J-S, Jude, KM, Fernandez, D, et al.. Clonally expanded B cells in multiple sclerosis bind EBV EBNA1 and GlialCAM. Nature 2022;603:321–7. https://doi.org/10.1038/s41586-022-04432-7.Search in Google Scholar PubMed PubMed Central

108. Bjornevik, K, Cortese, M, Healy Brian, C, Kuhle, J, Mina Michael, J, Leng, Y, et al.. Longitudinal analysis reveals high prevalence of Epstein-Barr virus associated with multiple sclerosis. Science 2022;375:296–301. https://doi.org/10.1126/science.abj8222.Search in Google Scholar PubMed

109. Dowd, JB, Palermo, T, Brite, J, McDade, TW, Aiello, A. Seroprevalence of Epstein-Barr virus infection in U.S. children ages 6-19, 2003–2010. PLoS One 2013;8:e64921. https://doi.org/10.1371/journal.pone.0064921.Search in Google Scholar PubMed PubMed Central

110. Huang, S-Y, Yang, Y-X, Kuo, K, Li, H-Q, Shen, X-N, Chen, S-D, et al.. Herpesvirus infections and Alzheimer’s disease: a Mendelian randomization study. Alzheimer’s Res Ther 2021;13:158. https://doi.org/10.1186/s13195-021-00905-5.Search in Google Scholar PubMed PubMed Central

111. Tiwari, D, Mittal, N, Jha, HC. Unraveling the links between neurodegeneration and Epstein-Barr virus-mediated cell cycle dysregulation. Curr Res Neurobiol 2022;3:100046. https://doi.org/10.1016/j.crneur.2022.100046.Search in Google Scholar

112. Dezfulian, M. A new Alzheimer’s disease cell model using B cells to induce beta amyloid plaque formation and increase TNF alpha expression. Int Immunopharm 2018;59:106–12. https://doi.org/10.1016/j.intimp.2018.04.012.Search in Google Scholar

113. Kanakry, JA, Hegde, AM, Durand, CM, Massie, AB, Greer, AE, Ambinder, RF, et al.. The clinical significance of EBV DNA in the plasma and peripheral blood mononuclear cells of patients with or without EBV diseases. Blood 2016;127:2007–17. https://doi.org/10.1182/blood-2015-09-672030.Search in Google Scholar

114. Zhang, N, Zuo, Y, Jiang, L, Peng, Y, Huang, X, Zuo, L. Epstein-barr virus and neurological diseases. Front Mol Biosci 2022;8:81. https://doi.org/10.3389/fmolb.2021.816098.Search in Google Scholar

115. Jakhmola, S, Jha, HC. Glial cell response to Epstein-Barr Virus infection: a plausible contribution to virus-associated inflammatory reactions in the brain. Virology 2021;559:182–95. https://doi.org/10.1016/j.virol.2021.04.005.Search in Google Scholar

116. Athanasiou, E, Gargalionis, AN, Anastassopoulou, C, Tsakris, A, Boufidou, F. New insights into the molecular interplay between human herpesviruses and Alzheimer’s disease—a narrative review. Brain Sci 2022;12:1010. https://doi.org/10.3390/brainsci12081010.Search in Google Scholar

117. Gate, D, Saligrama, N, Leventhal, O, Yang, AC, Unger, MS, Middeldorp, J, et al.. Clonally expanded CD8 T cells patrol the cerebrospinal fluid in Alzheimer’s disease. Nature 2020;577:399–404. https://doi.org/10.1038/s41586-019-1895-7.Search in Google Scholar

118. Tiwari, D, Singh, VK, Baral, B, Pathak, DK, Jayabalan, J, Kumar, R, et al.. Indication of neurodegenerative cascade initiation by amyloid-like aggregate-forming EBV proteins and peptide in Alzheimer’s disease. ACS Chem Neurosci 2021;12:3957–67. https://doi.org/10.1021/acschemneuro.1c00584.Search in Google Scholar

119. Bae, S, Yun, SC, Kim, MC, Yoon, W, Lim, JS, Lee, SO, et al.. Association of herpes zoster with dementia and effect of antiviral therapy on dementia: a population-based cohort study. Eur Arch Psychiatry Clin Neurosci 2021;271:987–97. https://doi.org/10.1007/s00406-020-01157-4.Search in Google Scholar

120. Chen, VC, Wu, SI, Huang, KY, Yang, YH, Kuo, TY, Liang, HY, et al.. Herpes zoster and dementia: a nationwide population-based cohort study. J Clin Psychiatry 2018;79:16m11312. https://doi.org/10.4088/jcp.16m11312.Search in Google Scholar

121. Lopatko Lindman, K, Hemmingsson, ES, Weidung, B, Brännström, J, Josefsson, M, Olsson, J, et al.. Herpesvirus infections, antiviral treatment, and the risk of dementia-a registry-based cohort study in Sweden. Alzheimer’s Dement 2021;7:e12119. https://doi.org/10.1002/trc2.12119.Search in Google Scholar PubMed PubMed Central

122. Cairns, DM, Itzhaki, RF, Kaplan, DL. Potential involvement of varicella zoster virus in Alzheimer’s disease via reactivation of quiescent herpes simplex virus type 1. J Alzheimer’s Dis 2022;88:1189–200. https://doi.org/10.3233/jad-220287.Search in Google Scholar

123. Lövheim, H, Olsson, J, Weidung, B, Johansson, A, Eriksson, S, Hallmans, G, et al.. Interaction between cytomegalovirus and herpes simplex virus type 1 associated with the risk of Alzheimer’s disease development. J Alzheimers Dis 2018;61:939–45. https://doi.org/10.3233/jad-161305.Search in Google Scholar PubMed

124. Osborne, V, Shakir, SAW. What is the difference between observed association and causal association, signals and evidence? Examples related to COVID-19. Front Pharmacol 2021;11:569189. https://doi.org/10.3389/fphar.2020.569189.Search in Google Scholar PubMed PubMed Central

125. Wakeford, R. Association and causation in epidemiology – half a century since the publication of Bradford Hill’s interpretational guidance. J R Soc Med 2015;108:4–6. https://doi.org/10.1177/0141076814562713.Search in Google Scholar PubMed PubMed Central

126. Altman, N, Krzywinski, M. Association, correlation and causation. Nat Methods 2015;12:899–900. https://doi.org/10.1038/nmeth.3587.Search in Google Scholar PubMed

127. Fu, W-Y, Wang, X, Ip, NY. Targeting neuroinflammation as a therapeutic strategy for Alzheimer’s disease: mechanisms, drug candidates, and new opportunities. ACS Chem Neurosci 2019;10:872–9. https://doi.org/10.1021/acschemneuro.8b00402.Search in Google Scholar PubMed

128. Sehrawat, S, Kumar, D, Rouse, BT. Herpesviruses: harmonious pathogens but relevant cofactors in other diseases? Front Cell Infect Microbiol 2018;8:177. https://doi.org/10.3389/fcimb.2018.00177.Search in Google Scholar PubMed PubMed Central

129. Liu, S, Hossinger, A, Heumüller, S-E, Hornberger, A, Buravlova, O, Konstantoulea, K, et al.. Highly efficient intercellular spreading of protein misfolding mediated by viral ligand-receptor interactions. Nat Commun 2021;12:5739. https://doi.org/10.1038/s41467-021-25855-2.Search in Google Scholar PubMed PubMed Central

130. Watson, LS, Hamlett, ED, Stone, TD, Sims-Robinson, C. Neuronally derived extracellular vesicles: an emerging tool for understanding Alzheimer’s disease. Mol Neurodegener 2019;14:22. https://doi.org/10.1186/s13024-019-0317-5.Search in Google Scholar PubMed PubMed Central

131. Lee, S, Mankhong, S, Kang, J-H. Extracellular vesicle as a source of Alzheimer’s biomarkers: opportunities and challenges. Int J Mol Sci 2019;20:1728. https://doi.org/10.3390/ijms20071728.Search in Google Scholar PubMed PubMed Central

132. Martins, ST, Alves, LR. Extracellular vesicles in viral infections: two sides of the same coin? Front Cell Infect Microbiol 2020;10:593170. https://doi.org/10.3389/fcimb.2020.593170.Search in Google Scholar PubMed PubMed Central

133. Garcia-Contreras, M, Thakor, AS. Extracellular vesicles in Alzheimer’s disease: from pathology to therapeutic approaches. Neural Regener Res 2023;18:18–22. https://doi.org/10.4103/1673-5374.343882.Search in Google Scholar PubMed PubMed Central

134. Spitzer, P, Condic, M, Herrmann, M, Oberstein, TJ, Scharin-Mehlmann, M, Gilbert, DF, et al.. Amyloidogenic amyloid-β-peptide variants induce microbial agglutination and exert antimicrobial activity. Sci Rep 2016;6:32228. https://doi.org/10.1038/srep32228.Search in Google Scholar PubMed PubMed Central

135. Kumar, DKV, Choi, SH, Washicosky, KJ, Eimer, WA, Tucker, S, Ghofrani, J, et al.. Amyloid-β peptide protects against microbial infection in mouse and worm models of Alzheimer’s disease. Sci Transl Med 2016;8:340ra72. https://doi.org/10.1126/scitranslmed.aaf1059.Search in Google Scholar PubMed PubMed Central

136. Steel, AJ, Eslick, GD. Herpes viruses increase the risk of Alzheimer’s disease: a meta-analysis. J Alzheimer’s Dis 2015;47:351–64. https://doi.org/10.3233/jad-140822.Search in Google Scholar PubMed

137. Warren-Gash, C, Forbes, HJ, Williamson, E, Breuer, J, Hayward, AC, Mavrodaris, A, et al.. Human herpesvirus infections and dementia or mild cognitive impairment: a systematic review and meta-analysis. Sci Rep 2019;9:4743. https://doi.org/10.1038/s41598-019-41218-w.Search in Google Scholar PubMed PubMed Central

138. Vigasova, D, Nemergut, M, Liskova, B, Damborsky, J. Multi-pathogen infections and Alzheimer’s disease. Microb Cell Factories 2021;20:25. https://doi.org/10.1186/s12934-021-01520-7.Search in Google Scholar PubMed PubMed Central

139. Carrasco, L, Pisa, D, Alonso, R. Polymicrobial infections and neurodegenerative diseases. Curr Clin Microbiol Rep 2020;7:20–30. https://doi.org/10.1007/s40588-020-00139-3.Search in Google Scholar

140. Bu, XL, Yao, XQ, Jiao, SS, Zeng, F, Liu, YH, Xiang, Y, et al.. A study on the association between infectious burden and Alzheimer’s disease. Eur J Neurol 2015;22:1519–25. https://doi.org/10.1111/ene.12477.Search in Google Scholar PubMed

141. Douros, A, Santella, C, Dell’Aniello, S, Azoulay, L, Renoux, C, Suissa, S, et al.. Infectious disease burden and the risk of Alzheimer’s disease: a population-based study. J Alzheimer’s Dis 2021;81:329–38. https://doi.org/10.3233/jad-201534.Search in Google Scholar

142. Cummings, J, Lee, G, Zhong, K, Fonseca, J, Taghva, K. Alzheimer’s disease drug development pipeline: 2021. Alzheimer’s Dement 2021;7:e12179. https://doi.org/10.1002/trc2.12179.Search in Google Scholar PubMed PubMed Central

143. Crous-Bou, M, Minguillón, C, Gramunt, N, Molinuevo, JL. Alzheimer’s disease prevention: from risk factors to early intervention. Alzheimer’s Res Ther 2017;9:71. https://doi.org/10.1186/s13195-017-0297-z.Search in Google Scholar PubMed PubMed Central

144. Serrano-Pozo, A, Frosch, MP, Masliah, E, Hyman, BT. Neuropathological alterations in Alzheimer disease. Cold Spring Harbor Perspect Med 2011;1:a006189. https://doi.org/10.1101/cshperspect.a006189.Search in Google Scholar PubMed PubMed Central

145. Perl, DP. Neuropathology of Alzheimer’s disease. Mt Sinai J Med 2010;77:32–42. https://doi.org/10.1002/msj.20157.Search in Google Scholar PubMed PubMed Central

146. Bloom, GS. Amyloid-β and tau: the trigger and bullet in alzheimer disease pathogenesis. JAMA Neurol 2014;71:505–8. https://doi.org/10.1001/jamaneurol.2013.5847.Search in Google Scholar PubMed

147. Götz, J, Chen, F, van Dorpe, J, Nitsch, RM. Formation of neurofibrillary tangles in P301l tau transgenic mice induced by Abeta 42 fibrils. Science 2001;293:1491–5. https://doi.org/10.1126/science.1062097.Search in Google Scholar PubMed

148. Lewis, J, Dickson, DW, Lin, WL, Chisholm, L, Corral, A, Jones, G, et al.. Enhanced neurofibrillary degeneration in transgenic mice expressing mutant tau and APP. Science 2001;293:1487–91. https://doi.org/10.1126/science.1058189.Search in Google Scholar PubMed

149. Hurtado, DE, Molina-Porcel, L, Iba, M, Aboagye, AK, Paul, SM, Trojanowski, JQ, et al.. Aβ accelerates the spatiotemporal progression of tau pathology and augments tau amyloidosis in an Alzheimer mouse model. Am J Pathol. 2010;177:1977–88. https://doi.org/10.2353/ajpath.2010.100346.Search in Google Scholar PubMed PubMed Central

150. Leroy, K, Ando, K, Laporte, V, Dedecker, R, Suain, V, Authelet, M, et al.. Lack of tau proteins rescues neuronal cell death and decreases amyloidogenic processing of APP in APP/PS1 mice. Am J Pathol 2012;181:1928–40. https://doi.org/10.1016/j.ajpath.2012.08.012.Search in Google Scholar PubMed

151. Bejanin, A, Schonhaut, DR, La Joie, R, Kramer, JH, Baker, SL, Sosa, N, et al.. Tau pathology and neurodegeneration contribute to cognitive impairment in Alzheimer’s disease. Brain 2017;140:3286–300. https://doi.org/10.1093/brain/awx243.Search in Google Scholar PubMed PubMed Central

152. Desai, P, Evans, D, Dhana, K, Aggarwal, NT, Wilson, RS, McAninch, E, et al.. Longitudinal association of total tau concentrations and physical activity with cognitive decline in a population sample. JAMA Netw Open 2021;4:e2120398. https://doi.org/10.1001/jamanetworkopen.2021.20398.Search in Google Scholar PubMed PubMed Central

153. Digma, LA, Madsen, JR, Reas, ET, Dale, AM, Brewer, JB, Banks, SJ, et al.. Tau and atrophy: domain-specific relationships with cognition. Alzheimer’s Res Ther 2019;11:65. https://doi.org/10.1186/s13195-019-0518-8.Search in Google Scholar PubMed PubMed Central

154. Li, H, Liu, C-C, Zheng, H, Huang, TY. Amyloid, tau, pathogen infection and antimicrobial protection in Alzheimer’s disease –conformist, nonconformist, and realistic prospects for AD pathogenesis. Transl Neurodegener 2018;7:34. https://doi.org/10.1186/s40035-018-0139-3.Search in Google Scholar PubMed PubMed Central

155. Wozniak, MA, Frost, AL, Itzhaki, RF. Alzheimer’s disease-specific tau phosphorylation is induced by herpes simplex virus type 1. J Alzheimer’s Dis 2009;16:341–50. https://doi.org/10.3233/jad-2009-0963.Search in Google Scholar

156. Powell-Doherty, RD, Abbott, ARN, Nelson, LA, Bertke, AS. Amyloid-β and p-tau anti-threat response to herpes simplex virus 1 infection in primary adult murine hippocampal neurons. J Virol 2020;94:e01874-19. https://doi.org/10.1128/jvi.01874-19.Search in Google Scholar

157. Sait, A, Angeli, C, Doig, AJ, Day, PJR. Viral involvement in Alzheimer’s disease. ACS Chem Neurosci 2021;12:1049–60. https://doi.org/10.1021/acschemneuro.0c00719.Search in Google Scholar PubMed PubMed Central

158. Bernal-Conde, LD, Ramos-Acevedo, R, Reyes-Hernández, MA, Balbuena-Olvera, AJ, Morales-Moreno, ID, Argüero-Sánchez, R, et al.. Alpha-synuclein physiology and pathology: a perspective on cellular structures and organelles. Front Neurosci 2020;13:1399. https://doi.org/10.3389/fnins.2019.01399.Search in Google Scholar PubMed PubMed Central

159. Lashuel, HA, Overk, CR, Oueslati, A, Masliah, E. The many faces of α-synuclein: from structure and toxicity to therapeutic target. Nat Rev Neurosci 2013;14:38–48. https://doi.org/10.1038/nrn3406.Search in Google Scholar PubMed PubMed Central

160. Bennett, MC. The role of alpha-synuclein in neurodegenerative diseases. Pharmacol Ther 2005;105:311–31. https://doi.org/10.1016/j.pharmthera.2004.10.010.Search in Google Scholar PubMed

161. Goedert, M. Alpha-synuclein and neurodegenerative diseases. Nat Rev Neurosci 2001;2:492–501. https://doi.org/10.1038/35081564.Search in Google Scholar PubMed

162. Twohig, D, Nielsen, HM. α-synuclein in the pathophysiology of Alzheimer’s disease. Mol Neurodegener 2019;14:23. https://doi.org/10.1186/s13024-019-0320-x.Search in Google Scholar PubMed PubMed Central

163. Irwin, DJ, Hurtig, HI. The contribution of tau, amyloid-beta and alpha-synuclein pathology to dementia in Lewy body disorders. J Alzheimers Dis Parkinsonism 2018;8:444. https://doi.org/10.4172/2161-0460.1000444.Search in Google Scholar PubMed PubMed Central

164. Moussaud, S, Jones, DR, Moussaud-Lamodière, EL, Delenclos, M, Ross, OA, McLean, PJ. Alpha-synuclein and tau: teammates in neurodegeneration? Mol Neurodegener 2014;9:43. https://doi.org/10.1186/1750-1326-9-43.Search in Google Scholar PubMed PubMed Central

165. Mukaetova-Ladinska, EB, Hurt, J, Jakes, R, Xuereb, J, Honer, WG, Wischik, CM. α-Synuclein inclusions in alzheimer and Lewy body diseases. J Neuropathol Exp Neurol 2000;59:408–17. https://doi.org/10.1093/jnen/59.5.408.Search in Google Scholar PubMed

166. Bantle, CM, Phillips, AT, Smeyne, RJ, Rocha, SM, Olson, KE, Tjalkens, RB. Infection with mosquito-borne alphavirus induces selective loss of dopaminergic neurons, neuroinflammation and widespread protein aggregation. NPJ Parkinson’s Dis 2019;5:20. https://doi.org/10.1038/s41531-019-0090-8.Search in Google Scholar PubMed PubMed Central

167. Beatman, EL, Massey, A, Shives, KD, Burrack, KS, Chamanian, M, Morrison, TE, et al.. Alpha-synuclein expression restricts RNA viral infections in the brain. J Virol 2015;90:2767–82. https://doi.org/10.1128/jvi.02949-15.Search in Google Scholar PubMed PubMed Central

168. Massey, AR, Beckham, JD. Alpha-synuclein, a novel viral restriction factor hiding in plain sight. DNA Cell Biol 2016;35:643–5. https://doi.org/10.1089/dna.2016.3488.Search in Google Scholar PubMed

169. Ait Wahmane, S, Achbani, A, Ouhaz, Z, Elatiqi, M, Belmouden, A, Nejmeddine, M. The possible protective role of α-synuclein against severe acute respiratory syndrome coronavirus 2 infections in patients with Parkinson’s disease. Mov Disord 2020;35:1293–4. https://doi.org/10.1002/mds.28185.Search in Google Scholar PubMed PubMed Central

170. Drummond, E, Wisniewski, T. Alzheimer’s disease: experimental models and reality. Acta Neuropathol 2017;133:155–75. https://doi.org/10.1007/s00401-016-1662-x.Search in Google Scholar PubMed PubMed Central

171. Vitek, MP, Araujo, JA, Fossel, M, Greenberg, BD, Howell, GR, Rizzo, SJS, et al.. Translational animal models for Alzheimer’s disease: an alzheimer’s association business Consortium think tank. Alzheimers Dement 2020;6:e12114. https://doi.org/10.1002/trc2.12114.Search in Google Scholar PubMed PubMed Central

172. Banik, A, Brown, RE, Bamburg, J, Lahiri, DK, Khurana, D, Friedland, RP, et al.. Translation of pre-clinical studies into successful clinical trials for alzheimer’s disease: what are the roadblocks and how can they Be overcome? J Alzheimers Dis 2015;47:815–43. https://doi.org/10.3233/jad-150136.Search in Google Scholar PubMed

173. Cummings, JL, Morstorf, T, Zhong, K. Alzheimer’s disease drug-development pipeline: few candidates, frequent failures. Alzheimer’s Res Ther 2014;6:37. https://doi.org/10.1186/alzrt269.Search in Google Scholar PubMed PubMed Central

174. Li, H-W, Zhang, L, Qin, C. Current state of research on non-human primate models of Alzheimer’s disease. Anim Models Exp Med 2019;2:227–38. https://doi.org/10.1002/ame2.12092.Search in Google Scholar PubMed PubMed Central

175. Haque, RU, Levey, AI. Alzheimer’s disease: a clinical perspective and future nonhuman primate research opportunities. Proc Natl Acad Sci USA 2019;116:26224. https://doi.org/10.1073/pnas.1912954116.Search in Google Scholar PubMed PubMed Central

176. Estes, JD, Wong, SW, Brenchley, JM. Nonhuman primate models of human viral infections. Nat Rev Immunol 2018;18:390–404. https://doi.org/10.1038/s41577-018-0005-7.Search in Google Scholar PubMed PubMed Central

177. Wang, F. Nonhuman primate models for Epstein-Barr virus infection. Curr Opin Virol 2013;3:233–7. https://doi.org/10.1016/j.coviro.2013.03.003.Search in Google Scholar PubMed PubMed Central

178. Leibovitch, E, Wohler, JE, Cummings Macri, SM, Motanic, K, Harberts, E, Gaitán, MI, et al.. Novel marmoset (Callithrix jacchus) model of human Herpesvirus 6A and 6B infections: immunologic, virologic and radiologic characterization. PLoS Pathog 2013;9:e1003138. https://doi.org/10.1371/journal.ppat.1003138.Search in Google Scholar PubMed PubMed Central

179. Reynaud, J, Horvat, B. Animal models for human herpesvirus 6 infection. Front Microbiol 2013;4:174. https://doi.org/10.3389/fmicb.2013.00174.Search in Google Scholar PubMed PubMed Central

180. Schmidt, F, Boltze, J, Jäger, C, Hofmann, S, Willems, N, Seeger, J, et al.. Detection and quantification of β-amyloid, pyroglutamyl Aβ, and tau in aged canines. J Neuropathol Exp Neurol 2015;74:912–23. https://doi.org/10.1097/nen.0000000000000230.Search in Google Scholar

181. Prpar Mihevc, S, Majdič, G. Canine cognitive dysfunction and Alzheimer’s disease - two facets of the same disease? Front Neurosci 2019;13:604. https://doi.org/10.3389/fnins.2019.00604.Search in Google Scholar PubMed PubMed Central

182. Kaeberlein, M, Creevy, KE, Promislow, DEL. The dog aging project: translational geroscience in companion animals. Mamm Genome 2016;27:279–88. https://doi.org/10.1007/s00335-016-9638-7.Search in Google Scholar PubMed PubMed Central

183. Carreiras, CM, Mendes, E, Perry, JM, Francisco, PA, Marco-Contelles, J. The multifactorial nature of Alzheimer’s disease for developing potential therapeutics. Curr Top Med Chem 2013;13:1745–70. https://doi.org/10.2174/15680266113139990135.Search in Google Scholar PubMed

184. Iqbal, UH, Zeng, E, Pasinetti, GM. The use of antimicrobial and antiviral drugs in Alzheimer’s disease. Int J Mol Sci 2020;21:4920. https://doi.org/10.3390/ijms21144920.Search in Google Scholar PubMed PubMed Central

185. Tzeng, N-S, Chung, C-H, Lin, F-H, Chiang, C-P, Yeh, C-B, Huang, S-Y, et al.. Anti-herpetic medications and reduced risk of dementia in patients with herpes simplex virus infections—a nationwide, population-based cohort study in taiwan. Neurotherapeutics 2018;15:417–29. https://doi.org/10.1007/s13311-018-0611-x.Search in Google Scholar PubMed PubMed Central

186. Devanand, DP, Andrews, H, Kreisl, WC, Razlighi, Q, Gershon, A, Stern, Y, et al.. Antiviral therapy: valacyclovir Treatment of Alzheimer’s Disease (VALAD) trial: protocol for a randomised, double-blind,placebo-controlled, treatment trial. BMJ Open 2020;10:e032112. https://doi.org/10.1136/bmjopen-2019-032112.Search in Google Scholar PubMed PubMed Central

187. Weidung, B, Hemmingsson, E-S, Olsson, J, Sundström, T, Blennow, K, Zetterberg, H, et al.. VALZ-Pilot: high-dose valacyclovir treatment in patients with early-stage Alzheimer’s disease. Alzheimer’s & Dementia. Transl Res Clin Inter 2022;8:e12264. https://doi.org/10.1002/trc2.12264.Search in Google Scholar PubMed PubMed Central

188. Haas, JG, Lathe, R. Microbes and Alzheimer’s disease: new findings call for a paradigm change. Trends Neurosci 2018;41:570–3. https://doi.org/10.1016/j.tins.2018.07.001.Search in Google Scholar PubMed

Received: 2023-05-22
Accepted: 2023-06-26
Published Online: 2023-07-10
Published in Print: 2023-09-26

© 2023 the author(s), published by De Gruyter, Berlin/Boston

This work is licensed under the Creative Commons Attribution 4.0 International License.

Downloaded on 9.6.2024 from https://www.degruyter.com/document/doi/10.1515/nipt-2023-0011/html
Scroll to top button