Next Article in Journal
Study of the Activity of the Staphylococcus aureus Phage vB_SaS_GE1 Against MRSA Clinical Isolates and Its Impact on the Formation of Dual-Species Biofilms with P. aeruginosa
Previous Article in Journal
Sociodemographic Associations and COVID-19 Symptoms Following One Year of Molecular Screening for SARS-CoV-2 Among Healthcare Workers
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Does Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) Represent a Poly-Herpesvirus Post-Virus Infectious Disease?

by
Maria Eugenia Ariza
1,2,
Irene Mena Palomo
1,3 and
Marshall V. Williams
1,2,*
1
Institute of Brain, Behavior and Immunology (IBBI), The Ohio State University Wexner Center, Columbus, OH 43210, USA
2
Department of Cancer Biology and Genetics (CBG), The Ohio State University Wexner Center, Columbus, OH 43210, USA
3
Biomedical Sciences Graduate Program, The Ohio State University College of Medicine, Columbus, OH 43210, USA
*
Author to whom correspondence should be addressed.
Viruses 2025, 17(12), 1624; https://doi.org/10.3390/v17121624
Submission received: 21 November 2025 / Revised: 9 December 2025 / Accepted: 12 December 2025 / Published: 16 December 2025
(This article belongs to the Section Human Virology and Viral Diseases)

Abstract

Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is a debilitating multisystem illness with unknown etiology. An estimated 17–24 million people representing approximately 1% of the population are afflicted worldwide. In over half of cases, ME/CFS onset is associated with acute “flu-like” symptoms, suggesting a role for viruses. However, no single virus has been identified as the only etiological agent. This may reflect the approach employed or more strongly the central dogma associated with herpesviruses replication, which states that a herpesvirus exists in two states, either lytic or latent. The purpose of this review is to address the role that abortive lytic replication may have in the pathogenesis of ME/CFS and other post-acute viral infections and also to raise awareness that these syndromes might be poly-herpesviruses mediated diseases.

1. Introduction

Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is a debilitating, multisystem illness characterized by post-exertional malaise (PEM), immune dysfunction, muscle pain and weakness, metabolic defects, cognitive impairment, sleep disturbances, and orthostatic intolerance [1,2,3,4,5]. Prior to the COVID pandemic, ME/CFS was estimated to affect 17–24 million people worldwide, representing approximately 1% of the population [6]. However, dismissal and limited awareness of the disease among some general practitioners coupled with inconsistent diagnostic criteria suggest that the true number may be far higher [7,8,9,10,11,12,13,14].
For decades, clinicians and researchers have developed separate case definitions and diagnostic criteria for ME [15,16,17,18,19,20,21,22]. Over this time period, four major sets of clinical case criteria have been established for the diagnosis of ME/CFS: the Fukuda criteria in 1994 [21] with and without the Reeves modification [22], the Canadian Consensus criteria (CCC) in 2003 [17], the International Consensus criteria (ICC) in 2011 [23], and the IOM criteria in 2015 [7], which is the current diagnostic criteria used by the CDC. In 2015, the IOM proposed that the Canadian Consensus criteria be employed for research purposes [7], but the Fukuda criteria has been employed in most studies [24] in spite of criticisms that this case definition is overtly inclusive [25,26]. Jason et al. [26] indicated that those subjects meeting the research criteria were significantly more impaired on a wide variety of symptoms and functional areas when compared to those meeting the clinical criteria. There are no specific biomarkers available to confirm a diagnosis of ME/CFS. Instead, patients are diagnosed by ruling out other illnesses or conditions that could explain their symptoms and by determining whether those symptoms meet established case criteria. At present, there are no approved treatments for ME/CFS.
Research into ME/CFS began long before formal case definitions were established. Early studies date back to the 1930s, when the outbreak of a polio-like illness known as benign myalgic encephalomyelitis was reported [27]. Because of the epidemic nature of the studies, clinicians and researchers suspected a viral cause. Prior to 1980, much of the focus was on persistent enterovirus infections, a hypothesis that remains under consideration as potential etiological agents of ME/CFS today [28,29]. Later, following the Nevada and New York outbreaks, the attention shifted to a human herpesvirus, Epstein–Barr virus (EBV), as patients exhibited symptoms resembling a chronic mononucleosis-like syndrome. These investigations centered on individuals with prolonged fatigue, and the term “post-fatigue syndrome” was coined. Reports indicated that more than 70% of chronic fatigue cases occurred following an “infectious, flu-like illness” [30,31,32], reinforcing the premise that a single virus might be the cause of ME/CFS.

2. Are the Symptoms of a Subset of Patients with ME/CSF the Result of a Post-Virus Infection?

Several viruses including the coronaviruses (SARS, SAR-C0V-2), influenza virus, Ebola virus, Dengue virus, enteroviruses (polio and Coxsackie B), Chikungunya virus, West Nile virus, Ross fever virus, and several herpesviruses (EBV, human herpesvirus 6 (HHV-6), human herpesvirus 7 (HHV-7), and varicella-zoster virus (VZV)) have been associated with post-acute infection syndromes (PAISs) [31]. While severity is variable, patients with PAIS exhibit flu-like symptoms of exertion intolerance, disproportionate levels of fatigue, neurocognitive and sensory impairment, unrefreshing sleep, and myalgia/arthralgia [31]. In the case of ME/CFS, the viruses most closely associated with this syndrome have been enteroviruses and several human herpesviruses. Enterovirus, a genus within the family Picornaviridae, comprises enteroviruses, coxsackieviruses, rhinoviruses, polioviruses, and echoviruses. These viruses cause a wide variety of illnesses ranging from the common cold to poliomyelitis and aseptic meningitis. In humans, they are among the most common infectious agents worldwide. More than 90% of primary infections are asymptomatic or result in a mild febrile illness. Primary infections usually occur by 5 years of age, and over 90% of the cases occur in children younger than 14 years. Enteroviruses produce acute cytolytic infections generally cleared by the host adaptive immune response. While somewhat controversial [33,34], it has been proposed that the development of persistence of non-cytolytic variants may occur following acute infections with enterovirus B serotypes (coxsackievirus and echovirus) [35,36,37]. Prior to 2010, there were numerous studies investigating the possible relationship between enterovirus infection and ME/CFS [38]. Since 2010, the number of studies has subsided, with only four studies, two of which were reviews [28,29,39,40], suggesting the interest might be fading.
Conversely, in the past decade or so, there has been an increased research interest regarding the potential role(s) of some herpesviruses, particularly EBV and HHV-6A/B, in ME/CFS. Numerous studies, both historical and ongoing, reflect this heightened focus, underscoring the persistent effort to clarify their role in the syndrome’s development.
There are nine members of the Herpesviridae that infect humans, and they are classified into three subfamilies: the α-herpesviruses (herpes simplex types 1 and 2 [HSV-1 and 2] and VZV], the β-herpesviruses (human cytomegalovirus [HCMV], HHV-6A and 6B and human herpesvirus-7 [HHV-7]), and the γ-herpesviruses (EBV and human herpesvirus-8 [HHV-8]) (Table 1). While individual herpesviruses can be distinguished based on cellular tropism, serology, and genomic DNA sequence, a common feature shared by members of the Herpesviridae family is the ability to establish life-long persistent infections. For most herpesviruses, primary infection occurs in childhood and is typically asymptomatic, with prevalence increasing with age. More than 90% of the adult population is persistently infected with EBV, HHV-6A/B, HHV-7, and VZV. Notably, these viruses are associated with a wide range of diseases in both immune-competent and immune-suppressed individuals, posing a greater burden than once recognized. Numerous studies have explored EBV and HHV-6 as possible triggers of ME/CFS, but conflicting findings regarding their presence in patients led Soto and Straus [41] to conclude that evidence for herpesviruses involvement in ME/CFS was fading. More recent research, however, suggests otherwise, as discussed in this manuscript.

3. If ME/CFS Is Triggered by a Virus in Some Cases, Why Has It Not Been Identified? Factors to Consider

Research investigating the viral etiology of ME/CFS has generally been epidemiological in nature, relying on serological evidence, increased viral load, or both to suggest causation. Most of these studies sought to identify a single etiological agent, yet none has been definitively identified/established. Several factors contributed to this problem: Variations in the case criteria used for diagnosis have resulted in extreme heterogeneity in the study populations, the cohorts’ size is often too small for robust statistical analysis, and the different endpoints measured. Perhaps more importantly are the experimental approaches and clinical samples employed. Most studies have focused on detecting antibodies, typically IgG, against specific viral proteins (antigens), or on viral load assessment using plasma or PBMCs. Each approach has inherent limitations, complicating efforts to reach firm conclusions.

3.1. Lack of Standardized Criteria to Designate Individuals as Healthy Controls

Most studies define healthy control populations as individuals lacking evidence of the disease under investigation. Yet, the virome of healthy controls is not characterized prior to their inclusion in a study, which is a critical oversight when studying virus-associated illnesses. Without such screening, biases may be introduced that further complicate interpretations of results. The human virome is a highly complex ecosystem, unique to each person, and shaped by factors such as age, diet, gender, and interactions with other members of the microbiome [49,50]. Despite its importance, research into the virome and its interplay with the broader microbiome is very limited. Metagenomic analyses have revealed that herpesviruses are commonly present in healthy individuals across multiple body compartments, including blood, gastrointestinal tract, lungs, oral cavity, nervous system, reproductive organs, skin, and urinary tract, while picornaviruses have been found primarily in the gastrointestinal system [49,50,51]. However, there is no evidence that control groups included in ME/CFS studies are screened for persistent infections which could influence baseline values. Consistent with this, RNA sequencing (RNA-seq) and metagenomic studies of ME/CFS patients’ feces, blood, and saliva have not identified significant differences between their virome and those of healthy controls [51,52,53,54,55,56,57]. Given the high prevalence of some herpesvirus infections (>90% for EBV, HHV-6, HHV-7, and VZV) in the adult population, it is not surprising that RNA-seq and metagenomic analyses fail to show statistically significant differences between healthy controls and ME/CFS cases. This aligns with two recent comprehensive studies reporting that ME/CFS cases and healthy controls exhibit similar exposure to most common pathogens [58,59].

3.2. Serological Studies

In general, most studies investigating the potential role of enteroviruses and herpesviruses as triggers for ME/CFS have relied on commercially available ELISAs or ELISA-like platforms to detect antibodies, typically IgG, against specific virus proteins. In some cases, these assays use highly purified viral antigens such as VP1 (a capsid protein of enteroviruses) or viral capsid antigen (VCA) of EBV. In other cases, however, they employ relative crude preparations (“purified viral lysates”). Because the viral lysates contain a mixture of many viral antigens, these ELISAs cannot determine nor distinguish the host’s immune response to any single virus protein. This is important since the antigenic properties of virus proteins are not the same. As demonstrated by Vaider-Shalt et al. [60], over the course of their evolution, HSV-1, EBV, and human cytomegalovirus (HCMV) have decreased the number of epitopes present in virus proteins to help them avoid immune detection. Thus, the ability of a virus protein to generate an antibody response is dependent on the amount of protein present in the host and its antigenicity. Not surprisingly, serological studies employing viral lysates have led to conflicting results. Recent studies using advanced methodologies such as peptide microarray [61] and suspension multiplex immunoassay [62] have shown that the EBV-specific IgG response in ME/CFS patients did not differ significantly from that of healthy controls. These findings are consistent with numerous reports highlighting that the interpretation of EBV serological data [63] and, by extension, serological data for other herpesviruses and potentially enteroviruses [64] is inherently complex and requires caution. Although enterovirus-specific IgM and IgG assays have been developed, their practical utility is limited due to antigenic cross-reactivity among different serotypes and the high prevalence of enteroviruses in the general population [64,65,66,67,68,69,70,71].

3.3. Use of Viral Load as a Requirement to Demonstrate a Viral Infection

A range of platforms has been employed to directly determine viral genome levels in ME/CFS patients and healthy controls. Although no studies to date have specifically examined enterovirus load in ME/CFS patients, several studies have consistently reported no increase in EBV and/or HHV-6 viral load among affected ME/CFS patients [72,73,74,75]. This raises an important question: do these findings reflect limitations in methodologies/approaches, or are they shaped by the prevailing dogma/assumptions about how these viruses replicate and persist in vivo?

3.4. Abortive Lytic Replication (ALR)

A central concept regarding the biology of herpesviruses is that two distinct phases of viral gene expression exist, either latency or lytic replication where virus progeny are produced. However, there is accumulating data to suggest that in vivo a third state exists in which the virus undergoes abortive lytic replication (ALR). In ALR, the virus initiates the lytic cycle but does not fully complete it, resulting in limited viral replication and no production of infectious virions. During this process, some immediate-early and early lytic genes are expressed, though the specific genes expressed vary across herpesviruses. Although early studies showed ALR in EBV [76,77,78,79,80,81,82], subsequent studies have shown that ALR also occurs in cells infected with HCMV [83,84], HHV-6 [85], HHV-8 [86,87,88], HSV-1 [89,90,91,92], and VZV [93,94,95]. An essential aspect of ALR is that it does not lead to an increase in virus load [76,78,80,81,83,90,96,97]. This characteristic of ALR provides a clear rationale for the lack of significant differences in viral load between individuals with ME/CFS cases and healthy controls. Despite evidence of lytic gene activity, the replication process aborts before producing infectious virions, thus preventing measurable changes in overall viral burden [76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97].
More importantly, although these immediate-early and early genes regulate virus replication, these same proteins also modulate host genes involved in immune evasion, cell proliferation, the establishment and maintenance of latency, and apoptosis. Elevated antibodies to the deoxyuridine triphosphate nucleotidohydrolases (dUTPase) encoded by the early genes of EBV (BLLF3), HHV-6 (U45), and VZV (ORF8) were reported in both longitudinal and single serum samples from patients with ME/CFS compared to controls [96,97], suggesting that ALR may be occurring in these patients. Moreover, it was recently reported that the EBV dUTPase protein is expressed in postmortem brain tissues of ME/CFS cases but not in non-ME/CFS control tissues [98], highlighting the potential of herpesvirus dUTPases as distinctive biomarkers for ME/CFS.

4. Is ME/CFS the Outcome of Synergistic Interactions Between Multiple Herpesviruses?

The premise that ME/CFS may be the result of a poly-herpesvirus infection is not new. Lerner et al. [99,100] proposed that ME/CFS is caused by EBV, HCMV, and HHV-6 in single or multiple virus co-infections. While no research has been specifically designed to directly test this hypothesis, several serological and viral load-based studies have shown evidence of increased reactivation of several herpesviruses in ME/CFS patients. Manian [101] found that among ME/CFS patients (n = 20) diagnosed according to the Holmes et al. [18] criteria, 20% exhibited elevated IgG antibodies against EBV, HHV-6, and HSV1/2. However, these findings were not statistically different from age- and gender-matched controls. Similarly, Cameron et al. [72], studying ME/CFS patients (n = 10; Age 34 ± 15; 7 females 3) from the Dubbo Infection Outcome Study (post-infectious fatigue), reported that all patients were IgG seropositive for HHV-6, and 50% were seropositive for HCMV, yet these rates did not differ significantly from the control group. In contrast, a larger study by Chapenko et al. [102] which assessed both serological and viral load found that 26.85% of ME/CFS patients (n = 108) diagnosed using the Fukuda case definition [21] exhibited active co-infections with HHV-6, HHV-7, or parvovirus B19, whereas none of the controls (n = 90) showed evidence of such active co-infections. Similar findings were reported using a larger cohort of ME/CFS patients (n = 200) diagnosed using the Fukuda case definition [21]. In these studies, latent infection or co-infection with HHV-6A/B, HHV-7, and parvovirus B19V was observed in 51.5% of patients and in 76.7% of healthy control individuals (n = 150). In contrast, active infection was observed in approximately 45% of ME/CFS patients compared to only 8.7% of healthy controls [103]. Likewise, Briese et al. [56], using a metagenomics study of saliva from ME/CFS patients (n = 106), found that 30% of cases were coinfected with two herpesviruses and 21% with at least three. Similar results were observed in the control group (n = 91; 34 and 26%, respectively). Halpin et al. [96] analyzed both single serum samples (n = 55) and longitudinal samples (n = 74, 4 samples each) from ME/CFS patients diagnosed using the Fukuda with Reeves modification as well as the Canadian case definitions. Their findings revealed that patients could be stratified into two distinct subgroups: those exhibiting IgG antibodies to the herpesviruses encoded dUTPase and those who did not. Furthermore, the study showed that a significant percentage of ME/CFS patients (30.91–52.7%) were simultaneously producing IgG antibodies against multiple human herpesviruses (EBV, HHV-6, and VZV) dUTPases compared to controls (n = 151; 17.21%) [96]. In a follow-up study involving a different patient cohort, the same group found evidence of significant reactivation of multiple herpesviruses (EBV, HHV-6, and VZV) in 50% of ME/CFS female cases (n = 40; 873 longitudinal samples over 46–80 days), based on elevated dUTPase IgG antibodies. In contrast, only 6% of the control cohort (n = 16) showed reactivation [97]. More importantly, anti-dUTPase antibody levels directly correlated with fatigue and pain, two hallmark symptoms of ME/CFS. Finally, there have been several additional reports of simultaneous reactivation of herpesviruses (EBV, HSV-1, HHV-6, and VZV) in ME/CFS patients, but these studies did not provide individual patient data [62,104,105].
Additional support for the hypothesis that ME/CFS may represent a poly-herpesvirus syndrome comes from several double-blind, placebo-controlled studies demonstrating that long-term therapy (6 months) with the herpesvirus-targeting antivirals valaciclovir or valganciclovir led to improved clinical symptoms and reduced serum antibody titers among ME/CFS patients infected with EBV or with combinations of EBV, HCMV, and HHV-6 [98,106,107,108,109,110,111].
Interestingly, adolescents who develop heterophile antibody-positive infectious mononucleosis (IM) caused by EBV [112] have a 12–23% probability of developing severe ME/CFS if they meet >1 set of criteria (Fukuda, Canadian, and Institute of Medicine) for ME/CFS [113,114,115,116,117,118]. Serological studies have also reported that, in addition to EBV, these patients may also be co-infected with HCMV, HSV-1, and/or HHV-6 [119,120,121,122,123]. Bertram et al. [120] reported that 39.5% of the serum samples from 215 patients with infectious mononucleosis showed active co-infections of EBV and HHV-6. Furthermore, Al Tabaa et al. demonstrated that ALR is a key feature of EBV in patients diagnosed with infectious mononucleosis [78]. However, the potential contribution of these viral interactions to the pathogenesis of ME/CFS remains unclear and requires further investigation.
Interactions between viruses and members of the microbiome in vivo is an understudied area; however, evidence suggests that such interactions can be beneficial, neutral, or detrimental, thereby influencing the clinical course of disease [124,125,126,127,128,129,130,131]. Similarly, concurrent reactivation of multiple herpesviruses in patients is considered a rare event and has only been reported in cases where clinical symptoms are present. The best example of a syndrome caused by the sequential reactivation of multiple herpesviruses is Drug Induced Hypersensitivity Syndrome (DIHS)/Drug Reaction with Eosinophilia and Systemic Symptoms (DRESS). DIHS/DRESS is a rare multiorgan, systemic, severe hypersensitivity reaction to a drug that can affect the skin, blood, and any internal organs, most commonly the liver, kidneys, lungs, and heart [128,129]. The cause of DISH/DRESS, which occurs in immunocompetent individuals, is multifactorial, involving drug-exposure, genetic predisposition, viral reactivation, and the immune system response. The sequential reactivation of HHV-6 followed by EBV and HCMV is a hallmark feature implicated in the etiology of DIHS/DRESS. While EBV reactivation can occur initially in some cases, HHV-6 is most often the initial herpesvirus reactivated [129,132]. Systemic complications occur in 15–25% of patients and are classified into two categories: infectious and autoimmune. Among the infectious complications, HCMV-induced pneumonia is associated with poor outcomes. In contrast, reactivation of HHV-6 and EBV has been linked to the development of autoimmune disease [133].
There are multiple reports in the literature documenting concurrent reactivation of multiple herpesviruses in vitro and in vivo [96,97,134,135,136,137]. With the exception of DIHS/DRESS patients, it remains unclear whether multiple herpesviruses reactivate in vivo simultaneously or in sequence. Concurrent reactivation of multiple herpesviruses in vivo may be the result of immunosuppression driven by an expansion of regulatory CD4+ T suppressor cells (Tregs) or by CD8+ T cell exhaustion. While herpesviruses have been reported to increase Treg levels following infection [138,139,140,141], studies examining ME/CFS patients have reported conflicting results regarding whether Tregs are elevated in this population [142,143,144].
Loebel et al. [74] reported an impaired EBV-specific T cell response and diminished IFN-γ production in ME/CFS patients. A recent study conducted in post-infectious-ME/CFS (PI-ME/CFS) [59] and ME/CFS patients revealed CD8+ T cell dysfunction [145] which may result in T cell exhaustion [146,147]. Notably, we have previously shown that EBV dUTPase alters T cell function in vitro and in vitro [148,149,150]. These dUTPase effects are consistent with ALR, and we proposed that the ALR of a herpesvirus, likely HHV-6, results in the chronic production of the dUTPase along with other virus proteins. These proteins, through the expression of cross-reactive epitopes, may drive T cell exhaustion [151,152,153], thereby triggering the sequential reactivation of other herpesviruses that contribute to hallmark ME/CFS symptoms, including fatigue, PEM, and cognitive impairment. Furthermore, disease severity is related not only to the number of herpesviruses reactivated but also the amplitude of this reactivation cascade and the duration of CD8+ T cell exhaustion (Figure 1). Nonetheless, further studies are needed to substantiate this premise.

5. Could Other Multisystem Illnesses Such as Gulf War Illness (GWI) and Post-Acute Sequelae of COVID-19 (PASC) Be Poly-Herpesvirus Infections?

5.1. Gulf War Illness (GWI)

Approximately one-third of the US veterans returning from the Operation Desert Storm conflict (8 August 1990–31 July 1991) are afflicted with a chronic multisystem disorder known as Gulf War Illness (GWI) [154,155]. Characterized by severe and debilitating symptoms including fatigue, musculoskeletal pain, and cognitive problems, GWI cases exhibit a significant overlap in symptoms with ME/CFS. Multiple hypotheses regarding the etiology of these symptoms have been proposed, but the etiological agents remain unknown. Studies to examine the potential role of viruses in GWI are limited. Vojdani and Thrasher [156] showed a statistically significant increase in IgM and IgG antibodies against EBV (VCA-IgM); HCMV (IgG); HSV-1 (IgG) HSV-2 (IgG); HHV-6 (IgG); and VZV (IgG) in GWI patients compared to controls. However, individual patient values were not reported. Maloney et al. [157] proposed a latent herpesvirus immune inflammatory response model for chronic multisystem illnesses (CMIs). CMI is a broad term used by the Department of Veterans Affairs to refer to complex conditions of unknown etiology characterized by persistent symptoms that lack a clear medical diagnosis and include veterans diagnosed with GWI and/or ME/CFS. Bast E et al. [158] suggested that GWI might be a post-viral syndrome resulting from pre-exposure to Middle East respiratory syndrome coronavirus (MERS). Halpin et al. [96] showed that a significant percentage of patients with GWI (29.34%) were simultaneously producing antibodies against multiple human herpesviruses-encoded dUTPases compared to controls (17.21%). Notably, GWI patients exhibited significantly higher levels of antibodies to the HHV-6 dUTPase than controls (p = 0.0053). Further analyses of longitudinal sera samples from two additional military veteran cohorts diagnosed with CMI or GWI revealed an increased prevalence of IgG (55% and 83%, respectively) and IgM antibodies (80–90% and 100%, respectively) to the dUTPase protein encoded by EBV, HHV-6A, and VZV compared to age- and gender-matched healthy controls (5% and 7% for IgG and IgM, respectively, p < 0.001) [159]. These findings indicate the importance of virus dUTPase, expressed as an early gene product during both lytic (productive) and abortive lytic (non-productive) replication of herpesviruses and which has novel immune- and neuro-modulatory functions, as a potential biomarker and key contributor to the pathophysiology of patients with CMI including GWI and ME/CFS [148,160,161,162,163,164,165,166,167,168,169,170,171,172].

5.2. Post-Acute Sequelae of COVID-19 (PASC)

Following SARS-CoV-2 infections, a significant number of individuals develop “Post-acute Sequelae of COVID-19 (PASC),” a syndrome characterized by profound fatigue as well as immune and neurocognitive dysfunction that persists for at least 3 months following diagnosis without any alternative explanation for the symptoms. While PASC is initiated by SARS-CoV-2, the underlying mechanisms contributing to the development of PASC remain unclear. Gentilotti et al. [173] identified four clinical phenotypes of PASC, with the dominant one resembling a chronic fatigue-like syndrome phenotype. PASC patients in this phenotype exhibited symptoms such as PEM, fatigue, and cognitive dysfunction identical to those seen in patients with ME/CFS [174,175,176]. Furthermore, it has been estimated that more than 40% of patients diagnosed with PASC could also meet the clinical criteria for ME/CFS [177,178,179,180]. Although several hypotheses have been proposed to explain the mechanisms underlying PASC, at this time, none have been validated. One hypothesis is that the clinical symptoms in PASC may not be a direct result from SARS-CoV-2 but rather from EBV reactivation triggered by COVID-19-induced inflammation [181]. In support of this hypothesis, numerous studies have reported serological and viral load evidence of reactivation of EBV, and other herpesviruses, during both acute SARS-CoV-2 infection and in PASC patients [182,183,184,185]. Furthermore, Su et al. [186], using a longitudinal multi-omics approach, reported that EBV reactivation was one of four biological factors associated with PASC development. Similarly, Peluso et al. [187] found a strong association between EBV reactivation and the fatigue associated with PASC. Verma et al. [188] further showed that EBV lytic replication induced the expression of ACE2 and enhanced SARS-CoV-2 entry into epithelial cells, suggesting that EBV reactivation may also contribute to COVID-19 and possibly PASC. Several studies have reported a correlation between EBV reactivation and disease severity in both COVID-19 and PASC [182,189,190,191,192,193,194,195,196,197,198]; however, conflicting data have also been reported [98,194]. Interestingly, several studies, including ours, have linked EBV reactivation with a subgroup of ME/CFS patients whose symptoms resemble those shown in some PASC patients [60,71,72,73,96,97,180,181,182,183,184,185,186,187,199,200,201].

6. Future Directions

There is sufficient data to demonstrate that ME/CFS patients can be divided into at least two subgroups: those whose illness is not triggered by a virus infection and those whose are. The latter group should be referred to as patients with post-infective ME/CFS (PI-ME/CFS). This classification is necessary because while the symptomology exhibited by patients in these subsets might be similar, the mechanism(s) driving the disease process will be different, and thus, different therapeutic approaches might be required. Putative viruses associated with PI-ME/CFS must be identified not based on viral load but instead on the concepts that abortive lytic replication and co-infections are responsible for this disease. This will require appropriate screening of ME/CFS patients for the presence of abortive-lytic replication. Screening can be performed using either qRT-PCR to quantitate expression of early genes or by using a serological approach to determine the antibody response to multiple proteins encoded by early genes (Figure 2). Early proteins that could be used for this purpose include viral DNA polymerase, viral DNA polymerase accessory protein, and the dUTPase, to name a few. Likewise, individuals classified as “healthy controls” should also be screened, since the expression of early virus proteins would suggest they are asymptomatic carriers and therefore not truly representative of “healthy controls.” To improve research consistency, a definitive ME/CFS case definition for researchers must be established, possibly using a combination of the Canadian and IOM case criteria, to reduce patient population heterogeneity and enhance data reproducibility.
It is essential to pursue more mechanistic and hypothesis-driven research to elucidate the pathophysiological effects of these viruses in PI-ME/CFS. Priority should be given to studies investigating synergistic interactions within the virome, particularly between members of the herpesviruses, as they play a role in the initiation and/or progression of ME/CFS. These investigations face limitations, since appropriate animal models for ME/CFS do not exist and human herpesviruses generally do not naturally infect common laboratory rodents. Consequently, such interactions must be identified and studied directly in patients. Advanced techniques such as bulk RNA sequencing and single-cell RNA sequencing (scRNA-seq) offer powerful tools to explore viral synergies and host–virus interactions. Insights gained from these approaches could improve diagnostic accuracy and guide the development of targeted therapeutics.
It has been reported that there is a positive correlation between the levels of antibodies against the herpesvirus dUTPases and the severity of pain and fatigue in patients with ME/CFS [97]. This raises the question of whether disease severity in these patients may be linked to the number of herpesviruses reactivated—a critical issue that requires further investigation.
It has been suggested that future studies into viral infections in ME/CFS should “focus on adaptive immune responses rather than surveillance for viral gene products” [56]. However, this perspective rests on the assumption that an increase in viral load is necessary to demonstrate viral pathogenesis. This assumption overlooks evidence from advanced technologies showing that many dynamic viral infections are abortive lytic in nature. In such cases, numerous viral proteins are expressed that may contribute to disease pathogenesis even without an increase in viral load.
DNA viruses, including human herpesviruses, encode a wide array of proteins. These proteins have diverse effects on host cells, such as promoting cell proliferation, inhibiting apoptosis, and evading immune responses. Identifying the specific viral gene products responsible for fatigue, pain, cognitive dysfunction, and post-exertional malaise (PEM) will be essential for developing reliable biomarkers and targeted therapies. Continued research in this area is therefore critical.
Finally, if simultaneous abortive lytic replication of multiple herpesviruses is a key driver in certain ME/CFS patients, then targeted treatment protocols could be developed for this PI-ME/CFS subgroup. Such approaches might include combinations of antiviral agents (e.g., ganciclovir), cell-depleting therapies such as rituximab, checkpoint inhibitors, or agents designed to prevent viral reactivation.

Author Contributions

Conceptualization, M.E.A. and M.V.W.; writing—original draft preparation, M.E.A. and M.V.W.; writing—review and editing, M.E.A., I.M.P., and M.V.W. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the National Institutes of Health grant RO1 A1084898.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Missailidis, D.; Annesley, S.J.; Fisher, P.R. Pathological Mechanisms Underlying Myalgic Encephalomyelitis/Chronic Fatigue Syndrome. Diagnostics 2019, 9, 80. [Google Scholar] [CrossRef] [PubMed]
  2. Anderson, G.; Maes, M. Mitochondria and immunity in chronic fatigue syndrome. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2020, 103, 109976. [Google Scholar] [CrossRef]
  3. Nakatomi, Y.; Mizuno, K.; Ishii, A.; Wada, Y.; Tanaka, M.; Tazawa, S.; Onoe, K.; Fukuda, S.; Kawabe, J.; Takahashi, M.; et al. Neuroinflammation in Patients with Chronic Fatigue Syndrome/Myalgic Encephalomyelitis: An 11C-(R)-PK11195 PET Study. J. Nucl. Med. 2014, 55, 945–950. [Google Scholar] [CrossRef] [PubMed]
  4. Wirth, K.J.; Scheibenbogen, C. Pathophysiology of skeletal muscle disturbances in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS). J. Transl. Med. 2021, 19, 162. [Google Scholar] [CrossRef] [PubMed]
  5. Wirth, K.J.; Scheibenbogen, C.; Paul, F. An attempt to explain the neurological symptoms of Myalgic Encephalomyelitis/Chronic Fatigue Syndrome. J. Transl. Med. 2021, 19, 471. [Google Scholar] [CrossRef]
  6. Lim, E.J.; Ahn, Y.C.; Jang, E.S.; Lee, S.W.; Lee, S.H.; Son, C.G. Systematic review and meta-analysis of the prevalence of chronic fatigue syndrome/myalgic encephalomyelitis (CFS/ME). J. Transl. Med. 2020, 18, 100. [Google Scholar] [CrossRef]
  7. Institute of Medicine of the National Academies. Encephalomyelitis/Chronic Fatigue Syndrome: Redefining an Illness; The National Academies Press: Washington, DC, USA, 2015; ISBN 978-0-309-31689-7. [Google Scholar]
  8. Brimmer, D.J.; Jones, J.F.; Boneva, R.; Campbell, C.; Lin, J.S.; Unger, E.R. Assessment of ME/CFS (Myalgic Encephalomyelitis/Chronic Fatigue Syndrome): A Case study for health care providers. MedEdPORTAL 2016, 12, 10527. [Google Scholar] [CrossRef]
  9. Friedberg, F.; Sunnquist, M.; Nacul, L. Rethinking the standard of care for Myalgic Encephalomyelitis/Chronic Fatigue Syndrome. J. Gen. Intern. Med. 2020, 35, 906–909. [Google Scholar]
  10. Araja, D.; Berkis, U.; Lunga, A.; Murovska, M. Shadow burden of undiagnosed Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) on society: Retrospective and prospective-in light of COVID-19. J. Clin. Med. 2021, 10, 3017. [Google Scholar] [CrossRef]
  11. Mirin, A.A.; Dimmock, M.E.; Jason, L.A. Updated ME/CFS prevalence estimates reflecting post-COVID increases and associated economic costs and funding implications. Fatigue Biomed. Health Behav. 2022, 10, 83–93. [Google Scholar] [CrossRef]
  12. Pheby, D.F.H.; Araja, D.; Berkis, U.; Brenna, E.; Cullinan, J.; de Korwin, J.D.; Gitto, L.; Hughes, D.A.; Hunter, R.M.; Trepel, D.; et al. The Role of prevention in reducing the economic impact of ME/CFS in Europe: A report from the socioeconomics working group of the European Network on ME/CFS (EUROMENE). Medicina 2021, 57, 388. [Google Scholar] [CrossRef]
  13. Solomon, L.; Reeves, W.C. Factors influencing the diagnosis of chronic fatigue syndrome. Arch. Intern. Med. 2004, 164, 2241–2245. [Google Scholar] [CrossRef]
  14. Vardaman, M.; Gilmour, S. Letter: Time to correct the record on the global burden of myalgic encephalomyelitis/chronic fatigue syndrome. J. Transl. Med. 2025, 25, 331. [Google Scholar] [CrossRef]
  15. Ramsay, A.M. Myalgic encephalomyelitis, or what. Lancet 1988, 2, 100. [Google Scholar] [CrossRef]
  16. Hyde, B.M. The Nightingale, Mylagic Encephalomyelitis (ME) Definition; Nightingale Research Foundation: Ottawa, ON, Canada, 2007. [Google Scholar]
  17. Carruthers, B.M.; Jain, A.K.; DeMeirleir, K.L.; Peterson, D.L.; Klimas, N.G.; Lerner, A.M.; Bested, A.C.; Flor-Henry, P.; Joshi, P.; Powles, A.C.P.; et al. Myalgic encephalomyelitis/chronic fatigue syndrome: Clinical working case definition, diagnostic and treatment protocols. J. Chronic Fatigue Syndr. 2003, 11, 7–115. [Google Scholar] [CrossRef]
  18. Holmes, G.P.; Kaplan, J.E.; Gantz, N.M.; Komaroff, A.L.; Schonberger, L.B.; Straus, S.E.; Jones, J.F.; Dubois, R.E.; Cunningham-Rundles, C.; Pahwa, S.; et al. Chronic fatigue syndrome: A working case definition. Ann. Intern. Med. 1988, 108, 387–389. [Google Scholar] [CrossRef]
  19. Sharpe, M.C. A report-chronic fatigue syndrome: Guidelines for research. J. R. Soc. Med. 1991, 84, 118–121. [Google Scholar] [CrossRef]
  20. Kitani, T.; Kuratsune, H.; Yamaguchi, K. Diagnostic criteria for chronic fatigue syndrome by the CFS study group in Japan. Nippon. Rinsho Jpn. J. Clin. Med. 1992, 50, 2600–2605. [Google Scholar]
  21. Fukuda, K.; Straus, S.E.; Hickie, I.; Sharpe, M.C.; Dobbins, J.G.; Komaroff, A.; Schluederberg, A.; Jones, J.F.; Lloyd, A.R.; Wessely, S.; et al. The chronic fatigue syndrome: A comprehensive approach to its definition and study. Ann. Intern. Med. 1994, 121, 953–959. [Google Scholar] [CrossRef]
  22. Reeves, W.C.; Wagner, D.; Nisenbaum, R.; Jones, J.F.; Gurbaxani, B.; Solomon, L.; Papanicolaou, D.A.; Unger, E.R.; Vernon, S.D.; Heim, C. Chronic fatigue syndrome—A clinically empirical approach to its definition and study. BMC Med. 2005, 3, 19. [Google Scholar] [CrossRef] [PubMed]
  23. Carruthers, B.M.; van de Sande, M.I.; De Meirleir, K.L.; Klimas, N.G.; Broderick, G.; Staines, D.; Powles, A.C.P.; Speight, N.; Vallings, R.; Bateman, L.; et al. Myalgic encephalomyelitis: International Consensus Criteria. J. Intern. Med. 2011, 270, 327–338. [Google Scholar] [CrossRef]
  24. Todhunter-Brown, A.; Campbell, P.; Broderick, C.; Cowie, J.; Davis, B.; Fenton, C.; Markham, S.; Sellers, C.; Thomson, K. Recent research in myalgic encephalomyelitis/chronic fatigue syndrome: An evidence map. Health Technol. Assess. 2025. ahead of print. [Google Scholar] [CrossRef]
  25. Jason, L.A.; Najar, N.; Porter, N.; Reh, C. Evaluating the centers for disease control’s empirical chronic fatigue syndrome case definition. J. Disabil. Policy Stud. 2009, 20, 93–100. [Google Scholar] [CrossRef]
  26. Jason, L.A.; Fox, P.A.; Gleason, K.D. The importance of a research definition. Fatigue Biomed. Health Behav. 2018, 6, 52–58. [Google Scholar]
  27. Acheson, E.D. The clinical syndrome variously called beign malgic encephalomyelitis, Iceland disease and epidemic neuromyasthenia. Am. J. Med. 1959, 26, 569–595. [Google Scholar]
  28. O’Neal, A.J.; Hanson, M.R. The enterovirus theory of disease in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome: A critical review. Front. Med. 2021, 8, 688486. [Google Scholar]
  29. Hanson, M.R. The viral origin of myalgic encephalomyelitis/chronic fatigue syndrome. PLoS Pathog. 2023, 18, e1011523. [Google Scholar]
  30. Naess, H.; Sundal, E.; Myhr, K.M.; Nyland, H.I. Postinfectious and chronic fatigue syndromes: Clinical experience from a tertiary-referral centre in Norway. In Vivo 2010, 24, 185–188. [Google Scholar]
  31. Choutka, J.; Jansari, V.; Hornig, M.; Iwasaki, A. Unexplained post-acute infection syndromes. Nat. Med. 2022, 28, 911–923. [Google Scholar] [CrossRef] [PubMed]
  32. Jason, L.A.; Yoo, S.; Bhatia, S. Patient preceptions of infectious illness preceeding Myalgic Encephalomyelitis/Chronic Fatigue Syndrome. Chronic Illn. 2022, 18, 901–910. [Google Scholar] [PubMed]
  33. Melchers, W.; Zoll, J.; van Kuppeveld, F.; Swanink, C.; Galama, J. There is no evidence for persistent enterovirus infections in chronic medical conditions in humans. Rev. Med. Virol. 1994, 4, 235–243. [Google Scholar]
  34. Muir, P.; Archard, L.C. There is evidence for persistent enterovirus infections in chronic medical conditions in humans. Rev. Med. Virol. 1994, 4, 245–250. [Google Scholar] [CrossRef]
  35. Kim, K.S.; Tracy, S.; Tapprich, W.; Bailey, J.; Lee, C.K.; Kim, K.; Barry, W.H.; Chapman, N.M. 5′-Terminal deletions occur in coxsackievirus B3 during replication in murine hearts and cardic myocyte cultures and correlate with encapsidation of negative-stranded viral RNA. J. Virol. 2005, 79, 7024–7041. [Google Scholar] [CrossRef] [PubMed]
  36. Flynn, C.T.; Kimura, T.; Frimpong-Boateng, K.; Whitton, J.L. Immunological and pathological consequences of coxsackievirus RNA persistence in the heart. Virology 2017, 512, 104–112. [Google Scholar] [CrossRef]
  37. Chapman, N.M. Persistent enterovirus infection: Little deletions, long infections. Vaccines 2022, 10, 770. [Google Scholar] [CrossRef]
  38. Rasa, S.; Nora-Krukle, Z.; Henning, N.; Eliassen, E.; Shikova, E.; Harrer, T.; Schelibenbogen, C.; Murovska, M.; Prusty, B.K. Chronic viral infections in myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS). J. Transl. Med. 2018, 16, 268. [Google Scholar] [CrossRef]
  39. Chia, J.; Chia, A.; Voeller, M.; Lee, T.; Chang, R. Acute enterovirus infection followed by myalgic encephalomeyelitis/chronic fatigue syndrome. J. Clin. Pathol. 2010, 63, 165–168. [Google Scholar]
  40. Hwang, J.H.; Lee, J.S.; Oh, H.M.; Lee, E.J.; Lim, E.J.; Son, C.G. Evaluation of viral infection as an etiology of ME/CFS: A systemic review and meta-analysis. J. Transl. Med. 2023, 21, 763. [Google Scholar]
  41. Soto, N.E.; Straus, S.E. Chronic fatigue syndrome and herpesviruses: The fading evidence. Herpes 2000, 7, 46–50. [Google Scholar]
  42. Howley, P.M.; Knipe, D.M.; Cohen, J.L.; Damania, B.A. Fields Virology: DNA Viruses, 7th ed.; Wolters Kluwer Publishers: Alphen aan den Rijn, The Netherlands, 2021; ISBN 979-1-9751-1257-8. [Google Scholar]
  43. Maple, P.A.C.; Hosseini, A.A. Human alpha herpesviruses infections (HSV1, HSV2, and VZV), Alzheimer’s disease, and the potential benefits of targeted treatment or vaccination—A virological perspective. Vaccines 2025, 13, 572. [Google Scholar] [CrossRef]
  44. Shang, Z.; Li, X. Human cytomegalovirus: Pathogenesis, prevention, and treatment. Mol. Biomed. 2024, 5, 61. [Google Scholar] [CrossRef]
  45. Agut, H.; Bonnafous, P.; Gautheret-Dejean, A. Laboratory and clinical aspects of human herpesvirus 6 infections. Clin. Microbiol. Rev. 2015, 28, 313–334. [Google Scholar] [CrossRef]
  46. Verbeek, R.; Vandekerckhove, L.; Van Cleemput, J. Update on human herpesvirus 7 pathogenesis and clinical aspects as a roadmap for future research. J. Virol. 2024, 98, e0043724. [Google Scholar] [CrossRef]
  47. Yu, H.; Robertson, E.S. Epstein–Barr virus history and pathogenesis. Viruses 2023, 15, 714. [Google Scholar] [CrossRef]
  48. Rigney, J.; Zhang, K.; Greas, M.; Liu, Y. A review of KSHV/HHV8-associated neoplasms and related lymphoproliferative lesions. Lymphatics 2025, 3, 20. [Google Scholar] [CrossRef]
  49. Bai, G.H.; Lin, S.C.; Hsu, Y.H.; Chen, S.Y. The human virome: Viral metagenomics, relations with human diseases and therapeutic applications. Viruses 2022, 14, 278. [Google Scholar] [CrossRef] [PubMed]
  50. Gholamzad, A.; Khakpour, N.; Hashemi, S.M.A.; Goudarzi, Y.; Ahmadi, P.; Gholamazad, M.; Mohammadi, M.; Hashemi, M. Exploring the virome: An integral part of human health and disease. Pathol. Res. Pract. 2024, 260, 155466. [Google Scholar] [CrossRef] [PubMed]
  51. Miller, R.R.; Uyaguari-Diaz, M.; McCabe, M.N.; Montoya, V.; Gardy, J.L.; Parker, S.; Steiner, T.; Hsiao, W.; Nesbitt, M.J.; Tang, P.; et al. Metagenomic investigation of plasma in individuals with ME/CFS highlights the importance of technical controls to elucidate contamination and batch effects. PLoS ONE 2016, 11, e0165691. [Google Scholar] [CrossRef] [PubMed]
  52. Giloteaux, L.; Hanson, M.R.; Keller, B.A. A pair of identical twins discordant for Myalgic Encephalomyelitis/Chronic Fatigue Syndrome differ in physiological parameters and gut microbiome composition. Am. J. Case Rep. 2016, 10, 720–729. [Google Scholar] [CrossRef]
  53. Bouquet, J.; Gardy, J.L.; Brown, S.; Pfeil, J.; Miller, R.R.; Morshed, M.; Avina-Zubieta, A.; Shojania, K.; McCabe, M.; Parker, S.; et al. RNA-seq analysis of gene expression, viral pathogen and B-cell/T-cell receptor signatures in complex chronic disease. Clin. Infect. Dis. 2017, 64, 476–481. [Google Scholar] [CrossRef]
  54. Bouquet, J.; Li, T.; Gardy, J.L.; Kang, X.; Stevens, S.; Stevens, J.; VanNess, M.; Snell, C.; Potts, J.; Miller, R.R.; et al. Whole blood human transcriptome and virome analysis of ME/CFS patients experiencing post-exertional malaise following cardiopulmonary exercise testing. PLoS ONE 2019, 14, e0212193. [Google Scholar] [CrossRef]
  55. Hsieh, S.Y.; Savva, G.M.; Telatin, A.; Tiwari, S.K.; Tariq, M.A.; Newberry, F.; Seton, K.A.; Booth, C.; Bansal, A.S.; Wileman, T.; et al. Investigating the human intestinal DNA virome and predicting disease-associated virus-host interactions in severe Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS). Int. J. Mol. Sci. 2023, 24, 17267. [Google Scholar] [CrossRef] [PubMed]
  56. Briese, T.; Tokarz, R.; Bateman, L.; Che, X.; Guo, C.; Jain, K.; Kapoor, V.; Levine, S.; Hornig, M.; Oleynik, A.; et al. A multicenter virome analysis of blood, feces and saliva in myalgic encephalomyelitis/chronic fatigue syndrome. J. Med. Virol. 2023, 95, e28993. [Google Scholar] [CrossRef]
  57. Obraitis, D.; Li, D. Blood virome research in myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS): Challenges and opportunities. Curr. Opin. Virol. 2024, 68–69, 101437. [Google Scholar] [CrossRef]
  58. Walitt, B.; Singh, K.; LaMunion, S.R.; Hallett, M.; Jacobson, S.; Chen, K.; Yoshimi Enose-Akahata, Y.; Apps, R.; Barb, J.J.; Bedard, P.; et al. Deep phenotyping of post-infectious myalgic encephalomyelitis/chronic fatigue syndrome. Nat. Commun. 2024, 15, 907. [Google Scholar] [CrossRef]
  59. Bastos, V.C.; Greene, K.A.; Tabachnikova, A.; Bhattacharjee, B.; Sjogren, P.; Bertilson, B.; Reifert, J.; Zhang, M.; Kamath, K.; Shon, J.; et al. Cerebrospinal fluid immune phenotyping reveals distinct immunotypes of myalgic encephalomyelitis/chronic fatigue syndrome. J. Immunol. 2025, 15, vkaf087. [Google Scholar] [CrossRef]
  60. Vider-Shalit, T.; Fishbain, V.; Raffaeli, S.; Louzoun, Y. Phase-dependent immune evasion of herpesviruses. J. Virol. 2007, 81, 9536–9545. [Google Scholar] [CrossRef] [PubMed]
  61. Loebel, M.; Eckey, M.; Sotzny, F.; Hahn, E.; Bauer, S.; Grabowski, P.; Zerweck, J.; Holenya, P.; Hanitsch, L.G.; Wittke, K.; et al. Serological profiling of EBV immune response in chronic fatigue syndrome using a peptide microarray. PLoS ONE 2017, 12, e0179124. [Google Scholar] [CrossRef] [PubMed]
  62. Blomberg, J.; Rizwan, M.; Bohlin-Wiener, A.; Elfaitouri, A.; Julin, P.; Zachrisson, O.; Rosen, A.; Gottfries, C.G. Antibodies to human herpesviruses in myalgic encephalomyelitis/chronic fatigue syndrome patients. Front. Immunol. 2019, 10, 1946. [Google Scholar] [CrossRef]
  63. De Paschale, M.; Clerici, P. Serological diagnosis of Epstein-Barr virus infection: Problems and solutions. World J. Virol. 2012, 12, 31–43. [Google Scholar] [CrossRef]
  64. Saarinen, N.V.V.; Lehtonen, J.; Veijola, R.; Lempainen, J.; Knip, M.; Hyoty, H.; Laitin, O.H.; Hytonen, V.P. Multiplex high-throughput serological assay for human enteroviruses. Microorganisms 2020, 8, 963. [Google Scholar] [CrossRef] [PubMed]
  65. Mertens, T.; Pika, U.; Eggers, H.J. Cross antigenicity among enteroviruses as revealed by immunoblot technique. Virology 1983, 129, 431–442. [Google Scholar] [CrossRef] [PubMed]
  66. Samuelson, A.; Forsgren, M.; Johansson, B.; Wahren, B.; Sallberg, M. Molecular basis for serological cross-reactivity between enteroviruses. Clin. Diagn. Lab. Immunol. 1994, 1, 336–341. [Google Scholar] [CrossRef]
  67. Iwasaki, J.; Smith, W.A.; Stone, S.R.; Thomas, W.R.; Hales, B.J. Species-specific and cross-reactive IgG1 antibody binding to viral capsid protein 1 (VP1) antigens of human rhinovirus species A, B and C. PLoS ONE 2013, 8, e70552. [Google Scholar]
  68. Xu, G.J.; Kula, T.; Xu, Q.; Li, M.Z.; Vernon, S.D.; Ndung’u, T.; Ruxrungtham, K.; Sanchez, J.; Brander, C.; Chung, R.T.; et al. Comprehensive serological profiling of human populations using a synthetic human virome. Science 2015, 348, aaa0698. [Google Scholar] [CrossRef]
  69. Bopegamage, S. Enterovirus infections: Pivoting role of the adaptive immune response. Virulence 2016, 7, 495–497. [Google Scholar] [CrossRef]
  70. Saarinen, N.V.V.; Stone, V.M.; Hankaniemi, M.M.; Mazur, M.A.; Vuorinen, T.; Flodström-Tullberg, M.; Hyöty, H.; Hytönen, V.P.; Laitinen, O.H. Antibody responses against enterovirus proteases are potential markers for an acute infection. Viruses 2020, 12, 78. [Google Scholar] [CrossRef]
  71. Jouppila, N.V.V.; Lehtonen, J.; Seppälä, E.; Puustinen, L.; Oikarinen, S.; Laitinen, O.H.; Knip, M.; Hyöty, H.; Hytönen, V.P. Assessment of enterovirus antibodies during early childhood using a multiplex immunoassay. Microbiol. Spectr. 2023, 11, e0535222. [Google Scholar] [CrossRef]
  72. Cameron, B.; Flamand, L.; Juwana, H.; Middeldrop, J.; Naing, Z.; Rawlinson, W.; Ablashi, D.; Lloyd, A. Serological and virological investigation of the role of the herpesviruses EBV, CMV and HHV-6 in post-infective fatigue syndrome. J. Med. Virol. 2010, 82, 1684–1688. [Google Scholar]
  73. Swanink, C.M.; van der Meer, J.W.; Vancoulen, J.H.; Bleijenberg, G.; Fennis, J.F.; Galama, J.M. Epstein-Barr virus (EBV) and the chronic fatigue syndrome: Normal virus load in blood and normal immunological reactivity in the EBV regression assay. Clin. Infect. Dis. 1995, 20, 1390–1392. [Google Scholar] [CrossRef]
  74. Loebel, M.; Strohschein, K.; Giannini, C.; Koelsch, U.; Bauer, S.; Doebis, C.; Thomas, S.; Unterwalder, N.; von Baehr, V.; Reinke, P.; et al. Deficient EBV-specific B-and T-cell response in patients with Chronic Fatigue Syndrome. PLoS ONE 2014, 9, e85387. [Google Scholar] [CrossRef]
  75. Kristianse, M.S.; Stabursvik, J.; O’Leary, E.C.; Pedersen, M.; Asprusten, T.T.; Leegaard, T.; Osnes, L.T.; Tjade, T.; Skovlund, E.; Godang, K.; et al. Clinical symptoms and markers of disease mechanisms in adolescent chronic fatigue following Epstein-Barr virus infections: An exploratory cross-sectional study. Brain Behav. Immun. 2019, 80, 551–563. [Google Scholar] [CrossRef] [PubMed]
  76. Laichalk, L.L.; Thorley-Lawson, D.A. Terminal differentiation into plasma cells initiates the replicative cycle of Epstein-Barr virus in vivo. J. Virol. 2005, 79, 1296–1307. [Google Scholar] [CrossRef] [PubMed]
  77. Al Tabaa, Y.; Tuaillon, E.; Bollore, K.; Foulongne, V.; Petitjean, G.; Seigneurin, J.M.; Duperray, C.; Desgranges, C.; Vendrell, J.P. Functional Epstein-Barr virus reservoir in plasma cells derived from infected blood memory B cells. Blood 2009, 113, 604–611. [Google Scholar] [CrossRef]
  78. Al Tabaa, Y.; Tuaillon, E.; Jeziorski, E.; Ouedraogo, D.E.; Bollore, K.; Rubbo, P.A.; Foulongne, V.; Rodiere, M.; Vendrell, J.P. B-cell polyclonal activation and Epstein-Barr viral abortive lytic cycle are two key features in acute infectious mononucleosis. J. Clin. Virol. 2011, 52, 33–37. [Google Scholar] [CrossRef] [PubMed]
  79. Altmann, M.; Hammerschmidt, W. Epstein-Barr virus provides a new paradigm: A requirement for the immediate inhibition of apoptosis. PLoS Biol. 2005, 3, e404. [Google Scholar] [CrossRef]
  80. Wen, W.; Iwakiri, D.; Yamamoto, K.; Maruo, S.; Kanda, T.; Takada, K. Epstein-Barr virus BZLF1 gene, a switch from latency to lytic replication, is expressed as an immediate-early gene after primary infection of B lymphocytes. J. Virol. 2007, 81, 1037–1042. [Google Scholar] [CrossRef]
  81. Shannon-Lowe, C.; Adland, E.; Bell, A.I.; Delecluse, H.J.; Rickman, A.B.; Rowe, M. Features distinguishing Epstein-Barr virus infections of epithelial cells and B cells: Viral genome expression, genome maintenance, and genome amplification. J. Virol. 2009, 83, 7749–7760. [Google Scholar] [CrossRef]
  82. Kalla, M.; Hammerschmidt, W. Human B cells on their route to latent infection-early but transient expression of lytic genes of Epstein-Barr virus. Eur. J. Cell Biol. 2012, 91, 65–69. [Google Scholar] [CrossRef]
  83. Perez, L. Characterizing cytomegalovirus infection one cell at a time. Nat. Microbiol. 2021, 6, 1477–1478. [Google Scholar] [CrossRef]
  84. Hein, M.Y.; Weissman, J.S. Functional single-cell genomics of human cytomegalovirus infection. Nat. Biotechnol. 2022, 40, 391–401. [Google Scholar] [PubMed]
  85. Prusty, B.K.; Gulve, N.; Chowdhury, S.R.; Schuster, M.; Strempel, S.; Descamps, V.; Rudel, T. HHV-6 encoded small non-coding RNAs define an immediate and early stage in viral replication. Genom. Med. 2018, 3, 25. [Google Scholar]
  86. Scholz, B.A.; Harth-Hertle, M.L.; Malterer, G.; Haas, J.; Ellwart, J.; Schulz, T.F.; Kempkes, B. Abortive lytic reactivation of KSHV in CBF1/CSL deficient human cell lines. PLoS Pathog. 2013, 9, e1003336. [Google Scholar]
  87. Campbell, D.M.; Rappocciolo, G.; Jenkins, F.J.; Rinaldo, C.R. Dendritic cells: Key players in human herpesvirus 8 infection and pathogenesis. Front. Microbiol. 2014, 5, 452. [Google Scholar] [CrossRef]
  88. Aneja, K.K.; Yuan, Y. Reactivation and lytic replication of Kaposi’s sarcoma-associated herpesvirus: An update. Front. Microbiol. 2017, 8, 613. [Google Scholar] [CrossRef]
  89. Ma, J.Z.; Russell, T.A.; Spelman, T.; Carbone, F.R.; Tscharke, D.C. Lytic replication is frequent in HSV-1 latent infection and correlates with the engagement of a cell-intrinsic transcriptional response. PLoS Pathog. 2014, 10, e1004361. [Google Scholar] [CrossRef]
  90. Drayman, N.; Patel, P.; Vistain, L.; Tay, S. HSV-1 single cell analysis reveals the activtion of anti-viral and developmental programs in distinct sub-populations. eLife 2019, 8, e46339. [Google Scholar]
  91. Kobiler, O.; Afriat, A. The fate of incoming HSV-1 genomes entering the nucleus. Curr. Issues Mol. Biol. 2021, 41, 221–266. [Google Scholar] [CrossRef]
  92. Fredrikson, J.P.; Domanico, L.F.; Pratt, S.L.; Loveday, E.K.; Taylor, M.P.; Chang, C.B. Single-cell herpes simplex virus type 1 infection of neyrons usingdrop-based microfluidics reveals heterogeneous replication kinetics. Sci. Adv. 2024, 10, eadk9185. [Google Scholar] [CrossRef] [PubMed]
  93. Laemmie, L.; Goldstein, R.S.; Kinchington, P.R. Modeling Varicella zoster virus persistence and reactivation-closer to resolving a perplexing persistent state. Front. Microbiol. 2019, 10, 1634. [Google Scholar]
  94. Warner, B.E.; Yee, M.B.; Zhang, M.; Hornung, R.S.; Kaufer, B.B.; Visalli, R.J.; Kramer, P.R.; Goins, W.F.; Kinchington, P.R. Varicella-zoster virus early infection but not complete replication is required for the induction of chronic hypersensitivity in rat models of postherpatic neuralgia. PLoS Pathog. 2021, 17, e1009689. [Google Scholar]
  95. Wang, J.; Bruckner, N.; Weissmann, S.; Gunther, T.; Zhu, S.; Vogt, C.; Sun, G.; Guo, R.; Bruno, R.; Ritter, B.; et al. Repression of varicella zoster gene express during quiescent infection in the absence of detectable histone deposition. PLoS Pathog. 2025, 21, e1012367. [Google Scholar]
  96. Halpin, P.; Williams, M.V.; Klimas, N.G.; Fletcher, M.A.; Barnes, Z.; Ariza, M.E. Myalgic encephalomyelitis/chronic fatigue syndrome and gulf war illness patients exhibit increase humoral responses to the herpesviruses-encoded dUTPase: Implications in disease pathophysiology. J. Med. Virol. 2017, 89, 1637–1645. [Google Scholar]
  97. Mena Paloma, I.; Cox, B.; Williams, M.V.; Ariza, M.E. Chronic reactivation of persistent human herpesviruses and hightened anti-dUTPase IgG antibodies is a recurrent hallmark in infectious ME/CFS and is associated with fatigue. J. Med. Virol. 2025. accepted. [Google Scholar]
  98. Kasimir, F.; Toomey, D.; Liu, Z.; Kaiping, A.C.; Ariza, M.E.; Prusty, B.P. Tissue specific signature of HHV-6 infection in ME/CFS. Front. Mol. Biosci. 2022, 9, 1044964. [Google Scholar] [CrossRef]
  99. Lerner, A.M.; Zervos, M.; Dworkin, H.J.; Chang, C.H.; Fitzgerald, J.T.; Goldstein, J.; Lawrie-Hoppen, C.; Franklin, B.; Krotkin, S.M.; Brodsky, M.; et al. New cardiomyopathy: Pilot study of intravenous ganciclovir in a subset of the chronic fatigue syndrome. Infect. Dis. Clin. Pract. 1997, 6, 110–117. [Google Scholar]
  100. Lerner, A.M.; Zervos, M.; Dworkin, H.J.; Chang, C.H.; O’Neill, W. A unified theory of the cause of chronic fatigue syndrome. Infect. Dis. Clin. Pract. 1997, 6, 239–243. [Google Scholar] [CrossRef]
  101. Manian, F.A. Simultaneous measurement of antibodies to Epstein-Barr virus, human herpesvirus 6, herpes simplex virus types 1 and 2 ad 14 enteroviruses in chronic fatigue syndrome: Is there evidence of activation of a nonspecific polyclonal immune response? Clin. Infect. Dis. 1994, 19, 448–453. [Google Scholar] [CrossRef]
  102. Chapenko, S.; Krumina, A.; Logina, I.; Rasa, S.; Chistjakovs, M.; Sultanova, A.; Viksna, L.; Murovska, M. Association of active human herpesvirus-6,-7 and parvovirus B19 infection with clinical outcomes in patients with myalgic encephalomyelitis/chronic fatigue syndrome. Adv. Virol. 2012, 2012, 205085. [Google Scholar] [CrossRef] [PubMed]
  103. Rasa-Dzelzkaleja, S.; Krumina, A.; Capenko, S.; Nora-Krukle, Z.; Gravelsina, S.; Vilmane, A.; Ievina, L.; Shoenfeld, Y.; Murovska, M.; on behalf of the VirA project. The persistent viral infections in the development and severity of myalgic encephalomyelitis/chronic fatigue syndrome. J. Transl. Med. 2023, 21, 33. [Google Scholar] [CrossRef]
  104. Cliff, J.M.; King, E.C.; Lee, J.S.; Sepulveda, N.; Wolf, A.S.; Kingdon, C.; Bowman, E.; Dockrell, H.M.; Nacul, L.; Lacerda, E.; et al. Cellular immune function in myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS). Front. Immunol. 2019, 10, 796. [Google Scholar] [CrossRef]
  105. Domingues, T.D.; Grabowska, A.D.; Lee, J.S.; Ameijerias-Alonso, J.; Westermeier, F.; Scheibenbogen, C.; Cliff, J.M.; Nacul, L.; Lacerda, E.M.; Mourino, H.; et al. Herpesvirus serology distinguishes different subgroups of patients from the United Kingdom myalgic encephalomyelitis/chronic fatigue syndrome biobank. Front. Med. 2021, 8, 686736. [Google Scholar]
  106. Lerner, A.M.; Beqaj, S.H.; Deeter, R.G.; Dworkin, H.J.; Zervos, M.; Chang, C.H.; Fitzgerald, J.T.; Goldstein, J.; O’Neill, W. A six-month trial of valacyclovir in the Epstein-Barr virus subset of myalgic encephalomyelitis/chronic fatigue syndrome: Improvement in left ventricular function. Drugs Today 2002, 38, 549–561. [Google Scholar]
  107. Lerner, A.M.; Beqaj, S.H.; Deeter, R.G.; Fitzgerald, J.T. Valacyclovir treatment in Epstein-Barr virus subset chronic fatigue syndrome; thirty-six months follow-up. In Vivo 2007, 21, 707–714. [Google Scholar]
  108. Lerner, A.M.; Beqaj, S.; Fitzgerald, J.T.; Gill, K.; Gill, C.; Edington, J. Subset-directed antiviral treatment of 142 herpesvirus patients with chronic fatigue syndrome. Virus Adapt. Treat. 2010, 2, 47–57. [Google Scholar]
  109. Kogelnik, A.M.; Loomis, K.; Hoegh-Petersen, M.; Rosso, F.; Hischier, C.; Montoya, J.G. Use of valganciclovir in patients with elevated antibody titers against human herpesvirus-6 (HHV-6) and Epstein-Barr virus (EBV) who were experiencing central nervous system dysfunction including long-standing fatigue. J. Clin. Virol. 2006, 73, S33–S38. [Google Scholar] [CrossRef] [PubMed]
  110. Watt, T.; Oberfoell, S.; Balise, R.; Lunn, M.R.; Kar, A.K.; Merrihew, L.; Bhangoo, M.S.; Montoya, J.G. Response to valganciclovir in chronic fatigue syndrome patients with human herpesvirus 6 and Epstein-Barr virus IgG antibody titers. J. Med. Virol. 2012, 84, 1967–1974. [Google Scholar] [PubMed]
  111. Montoya, J.G.; Kogelnik, A.M.; Bhangoo, M.; Lunn, M.R.; Flamand, L.; Merrihew, L.E.; Watt, T.; Kubo, J.T.; Paik, J.; Desai, M. Randomized clinical trial to evaluate the efficacy and safety of valganciclovir in a subset of patients with chronic fatigue syndrome. J. Med. Virol. 2013, 85, 2101–2109. [Google Scholar] [CrossRef] [PubMed]
  112. Blake, J.M.; Edwards, J.M.; Fletcher, W.; McSwiggan, D.A.; Pereira, M.S. Measurement of heterophile antibody and antibodies to EB viral capsid antigen IgG and IgM in suspected cases of infectious mononucleosis. J. Clin. Pathol. 1976, 29, 841–847. [Google Scholar]
  113. Katz, B.Z.; Shiraishi, Y.; Mears, C.J.; Binns, H.J.; Taylor, R. Chronic fatigue syndrome after infectious mononucleosis in adolescents. Pediatrics 2009, 124, 189–193. [Google Scholar] [CrossRef]
  114. Katz, B.Z.; Jason, L.A. Chronic fatigue syndrome following infections in adolescents. Curr. Opin. Pediatr. 2013, 25, 95–102. [Google Scholar] [CrossRef]
  115. Jason, L.A.; Katz, B.; Gleason, K.; McManimen, S.; Sunnquist, M.; Thrope, T. A prospective study of infectious mononucleosis in college students. Int. J. Psychiatry 2017, 2, 1–21. [Google Scholar] [PubMed Central]
  116. Katz, B.Z.; Reuter, C.; Lupovitch, Y.; Gleason, K.; McClellan, D.; Cotler, J.; Jason, L.A. A validated scale for assessing the severity of acute infectious mononucleosis. J. Pediatr. 2019, 209, 130–133. [Google Scholar] [CrossRef]
  117. Jason, L.A.; Cotler, J.; Islam, M.F.; Sunnquist, M.; Katz, B.Z. Risk for developing Myalgic encephalomyelitis/chronic fatigue syndrome in college students following infectious mononucleosis: A prospective cohort study. Clin. Infect. Dis. 2021, 73, e3740–e3746. [Google Scholar] [CrossRef]
  118. Jason, L.A.; Katz, B.Z. Predisposing and precipitating factors in Epstein-Barr. Virus-caused Myalgic encephalomyelitis/chronic fatigue syndrome. Microorganisms 2025, 13, 702. [Google Scholar] [CrossRef] [PubMed]
  119. Lemon, S.M.; Hutt, L.M.; Blum, J.; Pagano, J.S. Simultaneous infection with multiple herpesviruses. Am. J. Med. 1979, 66, 270–276. [Google Scholar] [CrossRef]
  120. Bertram, G.; Dreiner, N.; Krueger, G.R.; Ramon, A.; Ablashi, D.V.; Salahuddin, S.Z.; Balachandram, N. Frequent double infection with Epstein-Barr virus and human herpesvirus-6 in patients with acute infectious mononucleosis. In Vivo 1991, 5, 271–279. [Google Scholar]
  121. Olson, D.; Huntington, M.K. Co-infection with cytomegalovirus and Epstein-Barr virus in mononucleosis: Case report and review of literature. S. D. Med. 2009, 62, 351–353. [Google Scholar]
  122. Wang, X.; Yang, K.; Wei, C.; Huang, Y.; Zhao, D. Coinfection with EBV/CMV and other respiratory agents in children with suspected infectious mononucleosis. Virol. J. 2010, 7, 247. [Google Scholar] [CrossRef]
  123. Al Ameen, O.; Mohammed, A.; Faisal, M.; Kohla, S.; Abdulhadi, A. Co-infection of cytomegalovirus and Epstein-Barr virus in an immunocompetent patient: A case series and literature review. Cureus 2023, 15, e47599. [Google Scholar]
  124. McArdle, A.J.; Turkova, A.; Cunnington, A.J. When do co-infections matter? Curr. Opin. Infect. Dis. 2018, 31, 209–215. [Google Scholar] [CrossRef]
  125. Devi, P.; Khan, A.; Chattopathy, P.; Mehta, P.; Sahni, S.; Sharma, S.; Pandey, R. Co-infections as modulators of disease outcome: Minor players or major players? Front. Microbiol. 2021, 12, 664386. [Google Scholar] [CrossRef] [PubMed]
  126. Du, Y.; Wang, C.; Zhang, Y. Viral coinfections. Viruses 2022, 14, 2645. [Google Scholar] [CrossRef]
  127. Wu, H.; Zhou, H.Y.; Zheng, H.; Wu, A. Towards understanding and identification of human viral co-infections. Viruses 2024, 16, 673. [Google Scholar] [CrossRef]
  128. Flamand, L.; Stefanescu, I.; Ablashi, D.V.; Menezes, J. Activation of the Epstein-Barr Virus replicative cycle by human herpesvirus 6. J Virol. 1993, 67, 6768–6777. [Google Scholar] [CrossRef]
  129. Karner, W.; Bauer, G. Activation of a Varicella-zoster virus specific IgA response during acute Epstein-Barr virus infection. J. Med. Virol. 1994, 44, 258–262. [Google Scholar] [CrossRef] [PubMed]
  130. Pitksrsnts, A.; Piiparinen, H.; Mannonen, L.; Vesaluoma, M.; Vaheri, A. Detection of human herpesvirus 6 and varicella-zoster virus in tear fluid of patients with Bell’s palsy by PCR. J. Clin. Microbiol. 2000, 38, 2753–2755. [Google Scholar]
  131. Wu, H.; Li, T.; Zeng, M.; Peng, T. Herpes simplex virus type 1 infection activates the Epstein-Barr virus replication cycle via a CREB-dependent mechanism. Cell. Microbiol. 2012, 14, 546–559. [Google Scholar]
  132. Hama, N.; Abe, R.; Gibson, A.; Phillips, E.J. Drug-induced hypersensitivity syndrome (DIHS)/drug reaction with eosinophilia and systemic symptoms (DRESS): Clinical features and pathogenesis. J. Allergy Clin. Immunol. Pract. 2022, 10, 1155–1167. [Google Scholar] [CrossRef] [PubMed]
  133. Mizukawa, Y.; Shiohara, T. Recent advances in the diagnosis and treatment of DIHS/DRESS in 2025. Allergol. Int. 2025, 74, 372–379. [Google Scholar] [CrossRef] [PubMed]
  134. Kano, Y.; Hiraharas, K.; Sakuma, K.; Shiohara, T. Several herpesviruses can reactivate in severe drug-induced multiorgan reaction in the same sequential order as in graft-versus-host disease. Br. J. Dermatol. 2006, 155, 301–306. [Google Scholar]
  135. Seishima, M.; Yamanaka, S.; Fuijsawa, T.; Tohyama, M.; Hashimoto, K. Reactivation of human herpesvirus (HHV) family members other than HHV-6 in drug-induced hypersensitivity syndrome. Br. J. Dermatol. 2006, 155, 344–349. [Google Scholar]
  136. Ramirez, G.A.; Ripa, M.; Burastero, S.; Benanti, G.; Bagnasco, D.; Nannipieri, S.; Monardo, R.; Ponta, G.; Asperti, C.; Cilona, M.B.; et al. Drug reaction with eosinophilia and systemic symptoms (DRESS): Focus on the pathophysiological and diagnostic role of viruses. Microorganisms 2023, 11, 346. [Google Scholar] [CrossRef]
  137. Mizukawa, Y.; Aoyama, Y.; Takahashi, H.; Takahashi, R.; Shiohara, T. Risk of progression to autoimmune disease in severe drug eruption: Risk factors and the factor-guided stratification. J. Investig. Dermatol 2022, 142, 960–968. [Google Scholar] [PubMed]
  138. Hu, Z.; Usherwood, E.J. Immune escape of γ-herpesviruses from adaptive immunity. Rev. Med. Virol. 2014, 24, 365–378. [Google Scholar] [CrossRef] [PubMed]
  139. Yu, W.; Geng, S.; Suo, Y.; Wei, X.; Cal, Q.; Wu, B.; Zhou, X.; Shi, Y. Critical role of regulatory T cells in the latency and stress-induced reactivation of HSV-1. Cell Rep. 2018, 25, 2379–2389. [Google Scholar] [CrossRef] [PubMed]
  140. Peng, Q.; Cuo, X.; Luo, Y.; Wang, G.; Zhong, L.; Zhu, J.; Li, Y.; Zeng, X.; Feng, Z. Dynamic immune landscape and VZV-specific T cell responses in patients with herpes zoster and postherpetic neuralgia. Front. Immunol. 2022, 13, 887892. [Google Scholar] [CrossRef]
  141. Kakh, M.K.; Doroudchi, M.; Talepoor, V.A. Induction of regulatory T cells after virus infection and vaccination. Immunology 2025. Online ahead of print. [Google Scholar] [CrossRef]
  142. Ramos, S.; Brenu, E.; Broadley, S.; Kwiatek, R.; Ng, J.; Nguyen, T.; Freeman, S.; Staines, D.; Marshall-Gradisnik, S. Regulatory T, natural killer T and γδ T cells in multiple sclerosis and chronic fatigue syndrome/Myalgic encephalomyelitis: A comparison. Asian Pac. J. Allergy Immunol. 2016, 34, 300–305. [Google Scholar] [CrossRef]
  143. Rivas, J.L.; Palencia, T.; Fernandez, G.; Garcia, M. Association of T and NK cell phenotype with the diagnosis of myalgic encephalomyelitis/chronic fatigue syndrome. Front. Immunol. 2018, 9, 1028. [Google Scholar] [CrossRef]
  144. Ono, H.; Sato, W.; Yamamura, T. Dysregulation of T and B cells in myalgic encephalomyelitis/chronic fatigue syndrome. J. Neurol. Sci. 2017, 381, 899–900. [Google Scholar] [CrossRef]
  145. Gil, A.; Hoag, G.E.; Salerno, J.P.; Horning, M.; Klimas, N.; Selin, L.K. Identification of CD8 T-cell dysfunction associated with symptoms in myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) and Long COVID and treatment with a nebulized antioxidant/anti-pathogen agent in a retrospective case series. Brain Behav. Immun. 2024, 36, 100720. [Google Scholar] [CrossRef]
  146. Eaton-Fitch, N.; Rudd, P.; Er, T.; Hool, L.; Herrero, L.; Marshall-Gradisnik, S. Immune ehaustion in ME/CFS and Long COVID. JCI Insight 2024, 9, e183810. [Google Scholar] [CrossRef]
  147. Iu, D.S.; Maya, J.; Vu, L.T.; Fogarty, E.A.; McNairn, A.J.; Ahmed, F.; Franconi, C.J.; Munn, P.R.; Grenier, J.K.; Hanson, M.R.; et al. Transcriptional reprogramming primes CD8+ T cells toward exhaustion in Myalgic encephalomyelitis/chronic fatigue syndrome. Proc. Natl. Acad. Sci. USA 2024, 121, e241511912. [Google Scholar] [CrossRef]
  148. Williams, M.V.; Cox, B.; Ariza, M.E. Herpesviruses-encoded dUTPases: A new family of Pathogen-Associated Molecular Pattern (PAMP) proteins with Implications in human disease. Pathogens 2016, 6, 2. [Google Scholar] [CrossRef] [PubMed]
  149. Ariza, M.E.; Williams, M.V. EBV-dUTPase modulates host immune responses potentially altering the tumor microenvironment in EBV-associated maligancies. J. Curr. Res. HIV/AIDS 2016, 2016, 1. [Google Scholar]
  150. Young, N.A.; Williams, M.V.; Jarjour, W.N.; Bruss, M.S.; Bolon, B.; Parikh, S.; Satoskar, A.; Ariza, M.E. Epstein-Barr virus (EBV) encoded dUTPase exacerbates the immune pathology of lupus nephritis in vivo. Int. J. Immunol. Immunother. 2016, 3, 023. [Google Scholar] [CrossRef]
  151. Chiu, C.; McCausland, M.; Sidney, J.; Duh, F.M.; Rouphael, N.; Mehta, A.; Mulligan, M.; Carrington, M.; Wieland, A.; Sullivan, N.L.; et al. Broadly reactive human CD8 T cells that recognize an epitope conserved between VZV, HSV and EBV. PLoS Pathog. 2014, 10, e1004008. [Google Scholar]
  152. Jing, L.; Laing, K.J.; Dong, L.; Russel, R.M.; Barlow, R.S.; Haas, J.G.; Ramchandani, M.S.; Johnston, C.; Buus, S.; Redwood, A.J.; et al. Extensive CD4 and CD8 T-cell cross-reactivity between alphaherpesviruses. J. Immunol. 2016, 195, 2205–2218. [Google Scholar] [CrossRef]
  153. Almulla, A.F.; Normatov, M.G.; Supasitthumrong, T.; Maes, M. Molecular mimicry between Epstein-Barr virus and human herpesvirus-6 proteins and central nervous system proteins: Implications for T and B cell immunogenicity in an in silico study. bioRxiv 2025. [Google Scholar] [CrossRef]
  154. Fukuda, K.; Nisenbaum, R.; Stewart, G.; Thompson, W.W.; Robin, L.; Washko, R.M.; Noah, D.L.; Barrett, D.H.; Randall, B.; Herwaldt, B.L.; et al. Chronic multisymptom illness affecting Air Force veterans of the Gulf War. J. Am. Med. Soc. 1998, 280, 981–998. [Google Scholar]
  155. Steele, L. Prevalence and patterns of Gulf War Illness in Kansas veterans: Association of symptoms with characteristics of person, place and time of military service. Am. J. Epidemiol. 2000, 152, 992–1002. [Google Scholar]
  156. Vojdani, A.; Thrasher, J.D. Cellular and humoral immune antibodies in Gulf War veterans. Environ. Health Perspect. 2004, 112, 840–846. [Google Scholar] [PubMed]
  157. Maloney, S.R.; Jensen, S.; Gil-Rivas, V.; Goolkasian, P. Latent viral immune inflammatory response model for chronic multisystem illness. Med. Hypothesis 2013, 80, 220–229. [Google Scholar]
  158. Bast, E.; Jester, D.J.; Palacio, A.; Krengel, M.; Reinhard, M.; Ashford, J.W. Gulf War illness: A historical review and considerations of a post-viral syndrome. Mil. Med. 2025, in press. [Google Scholar] [CrossRef] [PubMed]
  159. Cox, B.; Goolkasian, P.; Mena-Palomo, I.; Williams, M.V.; Maloney, S.R.; Ariza, M.E. Reactivation of latent herpesviruses and a faulty antiviral response may contribute to chronic multi-system and multi-system illnesses in U.S. military veterans. J. Med. Virol. 2025, 97, e70400. [Google Scholar]
  160. Padgett, D.A.; Hotchkiss, A.K.; Pyter, L.M.; Nelson, R.J.; Yang, E.; Yeh, P.E.; Litsky, M.; Williams, M.; Glaser, R. Epstein-Barr Virus-Encoded dUTPase Modulates Immune Function and Induces Sickness Behavior in Mice. J. Med. Virol. 2004, 74, 442–448. [Google Scholar]
  161. Glaser, R.; Litsky, M.L.; Padgett, D.A.; Baiocchi, R.A.; Yang, E.V.; Chen, M.; Yeh, P.E.; Green-Church, K.B.; Caligiuri, M.A.; Williams, M. EBV-encoded dUTPase induces immune dysregulation: Implications for the pathophysiology of EBV-associated malignant disease. Virology 2006, 346, 205–218. [Google Scholar] [CrossRef]
  162. Waldman, W.J.; Williams, M.V.; Lemeshow, S.A.; Binkley, P.; Guttridge, D.; Kiecolt-Glaser, J.K.; Knight, D.A.; Guttridge, K.; Glaser, R. Epstein-Barr virus encoded dUTPase enhances proinflammatory cytokine production by macrophages in contact with endothelial cells: Evidence for depression-induced atherosclerotic risk. Brain Behav. Immun. 2008, 22, 215–223. [Google Scholar]
  163. Ariza, M.E.; Glaser, R.; Kaumaya, P.T.P.; Jones, C.; Williams, M. The Epstein-Barr Virus (EBV)-Encoded dUTPase Activates NF-kappa B through the TLR2 and MyD88-dependent Signaling Pathway. J. Immunol. 2009, 182, 851–859. [Google Scholar] [CrossRef]
  164. Lerner, A.M.; Ariza, M.E.; Williams, M.; Jason, L.; Beqaj, S.; Fitzgerald, J.T.; Lemeshow, S.; Glaser, R. Antibody to Epstein-Barr Virus deoxyuridine triphosphate nucleotidohydrolase and deoxyribonucleotide polymerase in a Chronic Fatigue Syndrome subset. PLoS ONE 2012, 7, e47891. [Google Scholar]
  165. Ariza, M.E.; Glaser, R.; Williams, M.V. Human herpesviruses encoded dUTPases: A family of proteins that modulate dendritic cells function and innate immunity. Front. Microbiol. 2014, 5, 504. [Google Scholar] [CrossRef]
  166. Aubrecht, T.G.; Salloum, B.A.; Ariza, M.E.; Williams, M.; Reader, B.; Glaser, R.; Sheridan, J.; Nelson, R.J. Restraint induces sickness responses independent of injection with Epstein-Barr virus-encoded dUTPase. J. Behav. Brain Sci. 2014, 4, 491–505. [Google Scholar] [CrossRef]
  167. Aubrecht, T.G.; Weil, Z.M.; Salloum, B.A.; Ariza, M.E.; Williams, M.; Reader, B.; Glaser, R.; Sheridan, J.; Nelson, R.J. Chronic physical stress does not interact with Epstein-Barr virus (EBV) encoded dUTPase to alter the sickness response. J. Behav. Brain Sci. 2015, 5, 513–523. [Google Scholar] [CrossRef]
  168. Williams, M.V.; Cox, B.; Alharshawi, K.; Lafuse, W.P.; Ariza, M.E. Epstein-Barr virus dUTPase contributes to neuroinflammation in a cohort of patients with Encephalomyelitis/Chronic Fatigue Syndrome. Clin. Ther. 2019, 41, 848–863. [Google Scholar] [CrossRef]
  169. Ariza, M.E. Myalgic encephalomyelitis/chronic fatigue syndrome: The human herpesviruses are back! Biomolecules 2021, 11, 185. [Google Scholar] [CrossRef]
  170. Ariza, M.E.; Cox, B.; Martinez, B.; Mena-Palomo, I.; Zarate, G.J.; Williams, M.V. Viral dUTPases: Modulators of Innate Immunity. Biomolecules 2022, 12, 227. [Google Scholar] [CrossRef]
  171. Alharshawi, K.; Cox, B.; Ariza, M.E. Examination of control asymptomatic cohorts reveals heightened anti-EBV and HHV-6A/B dUTPase antibodies in the aging population. J. Med. Virol. 2022, 94, 3464–3468. [Google Scholar] [CrossRef]
  172. Liu, Z.; Hollmann, C.; Kalanidhi, S.; Grothey, A.; Keating, S.; Mena-Palomo, I.; Schlosser, L.S.; Kaiping, A.; Scheller, C.; Sotzny FHorn, A.; et al. Increased circulating fibronectin, depletion of natural IgM and heightened EBV, HSV-1 reactivation in ME/CFS and long COVID. MedRxiv 2023. preprint. [Google Scholar]
  173. Gentilotti, E.; Gorska, A.; Tami, A.; Gusinow, R.; Miramdola, M.; Bario, J.R.; Baena, Z.R.P.; Rossi, E.; Hasenauer, J.; Lopes-Rafegas, I.; et al. Clinical phenotypes and quality of life to define post-COVID-19 syndrome: A cluster analysis of the multinational, prospective ORCHESTRA cohort. eClinicalMedicine 2023, 62, 102107. [Google Scholar] [CrossRef] [PubMed]
  174. Komaroff, A.L.; Lipkin, W.I. ME/CFS and Long COVID share similar symptoms and biological abnormalities: Road map to the literature. Front. Med. 2023, 10, 1187163. [Google Scholar] [CrossRef]
  175. Haider, S.; Janowski, A.J.; Lesnak, J.B.; Hayashi, K.; Dailey, D.L.; Chimenti, R.; Frey-Law, L.A.; Sluka, K.A.; Berardi, G. A comparison of pain, fatigue, and function between post–COVID-19 condition, fibromyalgia, and chronic fatigue syndrome: A survey study. Pain 2023, 164, 385–401. [Google Scholar] [CrossRef]
  176. Peppercorn, K.; Edgar, C.D.; Kleffmann, T.; Tate, W.O. A pilot study on the immune cell proteome of long COVID patients shows changes to physiological pathways similar to those in myalgic encephalomyelitis/chronic fatigue syndrome. Sci. Rep. 2023, 12, 22068. [Google Scholar] [CrossRef] [PubMed]
  177. Appleman, B.; Charlton, B.T.; Goulding, R.P.; Kerkhoff, T.J.; Breedveld, E.A.; Noort, W.; Offringa, C.; Bloemers, F.W.; van Weeghel, M.; Schomakers, B.V.; et al. Muscle abnormalities worsen after post-exertional malaise in long COVID. Nat. Commun. 2024, 15, 17. [Google Scholar] [CrossRef] [PubMed]
  178. Wong, T.L.; Weitzer, D.J. Long COVID and Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS)—A systemic review and comparison of clinical presentation and symptomatology. Medicina 2021, 57, 418. [Google Scholar] [CrossRef] [PubMed]
  179. Jason, L.A.; Dorri, J.A. ME/CFS and post-exertional malaise among patients with Long COVID. Neurol. Int. 2022, 15, 1–11. [Google Scholar] [CrossRef]
  180. Bonilla, H.; Quach, T.C.; Tiwari, A.; Bonilla, A.E.; Miglis, M.; Yang, P.C.; Eggert, L.E.; Sharifi, H.; Horomanski, A.; Subramanian, A.; et al. Myalgic Encephalomyelitis/Chronic Fatigue Syndrome is common in post-acute sequelae of SARS-CoV-2 infection (PASC): Results from a post-COVID-19 multidisciplinary clinic. Front. Neurol. 2023, 14, 1090747. [Google Scholar] [CrossRef]
  181. Gold, J.E.; Okyay, R.A.; Licht, W.E.; Hurley, D.J. Investigation of long COVID prevalence and its relationship to Epstein-Barr virus reactivation. Pathogens 2021, 10, 763. [Google Scholar] [CrossRef]
  182. Xie, Y.; Cao, S.; Dong, H.; Lv, H.; Teng, X.; Zhang, J.; Wang, T.; Zhang, X.; Qin, Y.; Chai, Y.; et al. Clinical characteristics and outcomes of critically ill patients with acute COVID-19 with Epstein-Barr virus reactivation. BMC Infect. Dis. 2021, 21, 955. [Google Scholar] [CrossRef]
  183. Paolucci, S.; Cassaniti, I.; Novazzi, F.; Fiorina, L.; Piralla, A.; Comolli, G.; Bruno, R.; Maserati, R.; Gulminetti, R.; Novati, S.; et al. COVID-19 Task Force. EBV DNA increase in COVID-19 patients with impaired lymphocyte subpopulation count. Int. J. Infect. Dis. 2021, 104, 315–319. [Google Scholar] [CrossRef]
  184. Bernal, K.D.E.; Whitehurst, C.B. Incidence of Epstein-Barr virus reactivation is elevated in COVID-19 patients. Virus Res. 2023, 334, 199157. [Google Scholar] [CrossRef] [PubMed]
  185. Kim, J.Y.H.; Ragusa, M.; Tortosa, F.; Torres, A.; Gresh, L.; Mendez-Rico, J.A.; Alvarez-Moreno, C.A.; Lisboa, T.C.; Valderrama-Beltran, S.L.; Aldighieri, S.; et al. Viral reactivations and co-infections in COVID-19 patients: A systematic review. BMC Infect. Dis. 2023, 23, 259. [Google Scholar] [CrossRef]
  186. Su, S.; Yuan, D.; Chen, D.G.; Ng, R.H.; Wang, K.; Choi, J.; Li, S.; Hong, S.; Zhang, R.; Xie, J.; et al. Multiple early factors anticipate post- acute COVID-19 sequelae. Cell 2022, 185, 881–895. [Google Scholar] [CrossRef] [PubMed]
  187. Peluso, M.J.; Deveau, T.M.; Munter, S.E.; Ryder, D.; Buck, A.; Beck-Engeser, G.; Chan, F.; Lu, S.; Goldberg, S.A.; Hoh, R.; et al. Chronic viral coinfections differentially affect the likelihood of developing long COVID. Clin. Investig. 2023, 133, e163669. [Google Scholar] [CrossRef] [PubMed]
  188. Verma, D.; Church, T.M.; Swaminathan, S. Epstein-Barr virus lytic replication induces ACE2 expression and enhances SARS-CoV-2 pseudotyped virus entry in epithelial cells. J. Virol. 2021, 95, e0019221. [Google Scholar] [CrossRef]
  189. Klein, J.; Wood, J.; Jaycox, J.R.; Dhodapkar, R.M.; Lu, P.; Gehlhausen, J.R.; Tabachnikova, A.; Greene, K.; Tabacof, L.; Malik, A.A.; et al. Distinguishing features of long COVID identified through immune profiling. Nature 2023, 623, 139–147. [Google Scholar] [CrossRef]
  190. Karachaliou, M.; Ranzani, O.; Espinosa, A.; Iraola-Guzman, S.; Castano-Vinyals, G.; Vidal, M.; Jimenez, A.; Banus, M.; Nogues, E.A.; Aguilar, R.; et al. Antibody responses to common viruses according to COVID-19 severity and postacute sequelae of COVID-19. J. Med. Virol. 2024, 96, e29862. [Google Scholar] [CrossRef]
  191. Liew, F.; Efstahiou, C.; Fontanella, S.; Richardson, M.; Saunders, R.; Swieboda, D.; Sidhu, J.K.; Ascough, S.; Moore, S.C.; Mohamed, N.; et al. Large-scale phenotyping of patients with long COVID post-hospitalization reveals mechanistic subtypes of disease. Nat. Immunol. 2024, 25, 607–621. [Google Scholar] [CrossRef]
  192. Shafiee, A.; Athar, M.M.T.; Amini, M.J.; Hajishah, H.; Siahvoshi, S.; Jalali, M.; Jahanbakhshi, B.; Mozhagani, S.H. Reactivation of herpesviruses during COVID-19: A systematic review and meta-analysis. Rev. Med. Virol. 2023, 33, e2437. [Google Scholar]
  193. Banko, A.; Miljanovic, D.; Cirkovic, A. Systematic review with meta-analysis of active herpesvirus infections in patients with COVID-19: Old players on the new field. Int. J. Infect. Dis. 2023, 130, 108–125. [Google Scholar] [CrossRef]
  194. Talukder, S.; Deb, P.; Parveen, M.; Zannat, K.E.; Bhuiyan, A.H.; Yeasmin, M.; Molla, M.M.A.; Saif-Ur-Rahman, K.M. Clinical features and outcomes of COVID-19 patients with concomitant herpesvirus co-infection or reactivation: A systematic review. New Microbes New Infect. 2024, 58, 101223. [Google Scholar] [CrossRef]
  195. Schneiderova, P.; Mizera, J.; Gharibian, A.; Manukyan, G.; Raska, M.; Gajdos, P.; Kosztyu, P.; Genzor, S.; Kudelka, M.; Sova, M.; et al. The SARS-Co-V-2 trigger highlights host interleukin 1 genetics in Epstein-Barr virus reactivation. Cell Rep. 2025, 44, 115859. [Google Scholar]
  196. Manoharan, S.; Ying, L.Y. Epstein Barr virus reactivation during COVID-19 hospitalization significantly increased mortality/death in SARS-CoV-2(+)/ EBV(+) than SARS-CoV-2(+)/EBV (−) patients: A comparative meta-analysis. Int. J. Clin. Pract. 2023, 2023, 1068000. [Google Scholar] [CrossRef] [PubMed]
  197. Mattei, A.; Schiavoni, L.; Riva, E.; Ciccozzi, M.; Veralli, R.; Urselli, A.; Citriniti, V.; Nenna, A.; Pascarella, G.; Costa, F.; et al. Epstein–Barr virus, Cytomegalovirus, and Herpes Simplex-1/2 reactivations in critically ill patients with COVID-19. Intensive Care Med. Exp. 2024, 12, 40. [Google Scholar] [CrossRef] [PubMed]
  198. Maguire, C.; Chen, J.; Rouphael, N.; Pickering, H.; Hoang Van Phan, H.; Glascock, A.; Chu, V.; Dandekar, R.; Corry, D.; Kheradmand, F.; et al. Chronic Viral Reactivation and Associated Host Immune Response and Clinical Outcomes in Acute COVID-19 and Post-Acute Sequelae of COVID-19. bioRxiv 2024. [Google Scholar] [CrossRef] [PubMed]
  199. Hoeggerl, A.D.; Nunhofer, V.; Lauth, W.; Badstuber, N.; Held, N.; Zimmermann, G.; Grabmer, C.; Weidner, L.; Jungbauer, C.; Lindbauer, N.; et al. Epstein-Barr virus reactivation is not causative for post-COVID-19-syndrome in individuals with asymptomatic or mild SARS-CoV-2 disease course. BMC Infect. Dis. 2023, 23, 800. [Google Scholar] [CrossRef]
  200. Peter, R.S.; Nieters, A.; Göpel, S.; Merle, U.; Steinacker, J.M.; Deibrert, P.; Friedmann-Bette, B.; Nieß, A.; Müller, B.; Schilling, C.; et al. Persistent symptoms and clinical findings in adults with post-acute sequelae of COVID-19/post-COVID-19 syndrome in the second year after acute infection: A population-based, nested case-control study. PLoS Med. 2025, 22, e100451. [Google Scholar]
  201. Cox, B.S.; Alharshawi, K.; Mena-Palomo, I.; Lafuse, W.P.; Ariza, M.E. EBV/HHV-6A dUTPases contribute to myalgic encephalomyelitis/chronic fatigue syndrome pathophysiology by enhancing TFH cell differentiation and extrafollicular activities. JCI Insight 2022, 7, e158193. [Google Scholar] [CrossRef]
Figure 1. Hypothetical interactions between sequential reactivation of several herpesviruses undergoing abortive lytic activation and the immune system and how these interactions contribute to symptomology observed in a cohort of patients diagnosed with ME/CFS.
Figure 1. Hypothetical interactions between sequential reactivation of several herpesviruses undergoing abortive lytic activation and the immune system and how these interactions contribute to symptomology observed in a cohort of patients diagnosed with ME/CFS.
Viruses 17 01624 g001
Figure 2. Flow diagram depicting protocols that need to be developed and followed for the accurate diagnosis and treatment of patients with ME/CFS caused by herpesvirus reactivation.
Figure 2. Flow diagram depicting protocols that need to be developed and followed for the accurate diagnosis and treatment of patients with ME/CFS caused by herpesvirus reactivation.
Viruses 17 01624 g002
Table 1. Human herpesviruses.
Table 1. Human herpesviruses.
VirusPrevalence
(%)
Age of Primary Infection Site of Productive InfectionSite of Latency Common Disease
HSV-1 [42,43]700.5–5 yrsEpithelial cellsTrigeminal GangliaOral herpes
HSV-2 [42,43]20–2515–49 yearsEpithelial cellsSacral GangliaGenital herpes
VZV [42,43]>955–9 yearsEpithelial cellsDorsal Root GangliaChickenpox and zoster
HCMV [42,44]50–>905–40 yearsEpithelial and Endothelial cellsMyeloid Cell
Linage
Immunosuppressed
Transplant
HHV-6A
[42,45]
>950.5–2 yearsCD4+ T cellsMonocyte/Macrophage Cell LinageImmunosuppressed
Transplant
HHV-6B
[42,45]
>950.5–2 years CD4+ T cellsMonocyte/Macrophage Cell LinageInfantum roseola (sixth disease)
HHV-7 [42,46]>951.5–3 yearsCD4+ T cellsMonocyte/Macrophage Cell LinageNeurological ?
EBV [42,47]>953–4 years
10–30 years
Epithelial and Plasma cellsMemory B cellInfectious mononucleosis, various lymphomas, and gastric carcinoma
HHV-8 [42,48]5–504 years in endemic areasEpithelial, Epithelial, and Plasma cellsEpithelial, Endothelial, and B cellsKaposi lymphoma, primary effusion lymphoma, and multicentric Castleman’s Disease
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Ariza, M.E.; Mena Palomo, I.; Williams, M.V. Does Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) Represent a Poly-Herpesvirus Post-Virus Infectious Disease? Viruses 2025, 17, 1624. https://doi.org/10.3390/v17121624

AMA Style

Ariza ME, Mena Palomo I, Williams MV. Does Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) Represent a Poly-Herpesvirus Post-Virus Infectious Disease? Viruses. 2025; 17(12):1624. https://doi.org/10.3390/v17121624

Chicago/Turabian Style

Ariza, Maria Eugenia, Irene Mena Palomo, and Marshall V. Williams. 2025. "Does Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) Represent a Poly-Herpesvirus Post-Virus Infectious Disease?" Viruses 17, no. 12: 1624. https://doi.org/10.3390/v17121624

APA Style

Ariza, M. E., Mena Palomo, I., & Williams, M. V. (2025). Does Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) Represent a Poly-Herpesvirus Post-Virus Infectious Disease? Viruses, 17(12), 1624. https://doi.org/10.3390/v17121624

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

Article Metrics

Back to TopTop