Viruses and psychiatric disorders: We have not crossed the borderline from hypothesis to proof yet (Review)
- Authors:
- Published online on: December 31, 2024 https://doi.org/10.3892/mmr.2024.13426
- Article Number: 61
-
Copyright: © Siafakas et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
Abstract
Introduction
Viruses are obligate intracellular entities that exist at the border between the living and non-living world. Viruses differ from their prokaryotic and eukaryotic counterparts in that they totally rely on their host and do not have their own metabolism (1,2). Notably, they do not grow or undergo any form of division, and they are produced by the assembly of pre-formed macromolecules.
Viruses do, however, possess genetic material, either DNA or RNA exclusively. In addition, their structural and functional traits are inherited, and they are subject to the same Darwinian evolutionary forces that drive the adaptation and survival of all living organisms (1). Viruses, especially those with an RNA genome, exhibit extensive genetic variability, far greater than that of all the living world, which confers upon them the evolutionary advantage of continuous adaptation to a multitude of different organs, systems and hosts (2). Notably, all viruses have been continuously shaped in both their structure and biological life cycle by this evolutionary interaction, seeking dynamic equilibrium with their host that will sustain their fitness and survival, as they passively follow the stochastic driving forces of natural selection.
Until a few decades ago, conventional and cell culture-based systems were relied upon for virus detection and identification; however, the limited abilities of such systems have hindered the complete understanding of the presence and role of viruses in all domains of life. Electron microscopy has provided the means to distinguish between different viral species, genera and families purely on the basis of their morphology and size, with no information about the driving force behind viral fitness and evolutionary success in the form of adaptation to these diverse domains of life. The implementation of novel, extremely sensitive, molecular detection and sequence-based identification methods, which are applicable to all types of organisms and environmental samples, has provided an impetus to transform the concept of viruses (2). A new generation of sequencing techniques has led to the more precise classification of viruses and has identified the degree of genetic heterogeneity that drives viral evolutionary success. Furthermore, this novel methodology has set the basis for the discovery of novel viruses, and the rapid detection and characterization of re-emergent viral strains with novel virulence and epidemic potential. This may markedly improve the efficiency with which we respond to viral diseases and outbreaks. Fig. 1 provides information on what is known so far about viruses and their relationship with their hosts.
First, it is now known that humans live alongside a considerable number of viruses and continuously interact with them. Furthermore, viruses are neither confined to a single type of host, nor a certain ecological niche; they thrive in all domains of life, affecting both prokaryotic and eukaryotic organisms (1). It is estimated that there are >1031 viruses on Earth in total (2). For example, >200,000 types of viruses have been identified in ocean water (3), revealing the universal evolutionary adaptation of viruses to all diverse ecosystems. A previously unsuspected multitude and variety of viruses have been discovered and remain to be discovered (4), primarily due to continuous advances in genome sequencing and bioinformatics, the two branches of the scientific discipline metagenomics.
Despite both the magnitude of virus heterogeneity and a parasitic life cycle, only a small proportion of viruses can cause disease (2). Their obligate reliance on the intracellular environment dictates that their survival and final evolutionary success depend on the establishment of a symbiotic relationship with their host, rather than a parasitic one (1). Notably, the most successful viruses are those that have reached an equilibrium with their host by evading the challenges that they face within the host, such as defensive host mechanisms and the immune system, which is generally sufficient to eliminate viruses, or to at least keep viral replication in check.
Nevertheless, there is still a significant number of viruses that cause disease, either mild or serious, when the pathogen-host equilibrium cannot be attained. Factors pertinent to both virus biology and/or human social activity may be responsible for pathogenicity, especially in the case of epidemics and pandemics. For example, the emergence of a novel viral variant due to genetic variation and natural selection in overcrowded societies with efficient transportation may lead to an epidemic and possibly to rapid pandemic spread (5,6). Past pandemics have shaped societies, or even whole human nations and empires, such as the Spanish flu in 1918 and the recent COVID-19 pandemic (7). The genetic novelty of viral variants may not only lead to a lack of pathogen control by the immune system, but also to new virulence properties with corresponding, short-term and/or long-term effects on human physiology and disease. The recent severeacuterespiratorysyndromecoronavirus-2 (SARS-CoV-2) pandemic with the subsequent long coronavirus disease (COVID) sequelae is an example of this (8,9).
Following entry into the host, an acute or a chronic state of infection will be established, depending on the result of the interaction between the virus and the host defense mechanisms. Following an acute infection that is not lethal, the host will usually recover with final clearance of the virus. However, long-term sequelae may ensue, either directly by long-term damage to organs and tissues, and/or indirectly via inflammatory cascades and other pathophysiological effects irrelevant to organ damage and dysfunction; this has been demonstrated with SARS-CoV-2 infection and long COVID syndrome (10). Chronic viral infections may be the direct result of an acute infection or may develop months, or even years, after primary entry of the virus into the host organism. In contrast to acute infections, chronic infections are characterized by a dynamic and metastable equilibrium between the host immune system and the virus (11). Most chronic infections described thus far are generally benign, where the virus enters a form of symbiosis with the host; however, there is some evidence that serious diseases and syndromes can be linked to chronic viral infections with a continuous inflammatory, or other pathophysiological, component, such as cancer, immune deficiency (AIDS), neurodegenerative disorders and cardiovascular disease (11).
Human endogenous retroviruses (HERVs) provide a significant example of mutualistic symbiosis between the host and a virus. These viruses are similar in their genetic constitution to their infectious counterparts. They actually constitute fossil retroviruses in the sense that they originate from ancient, exogenous retroviruses which, during the course of 100 million years of evolutionary history, integrated cDNA sequences of their RNA into the genome of human genetic cells through reverse transcription (12). Gradual integration and vertical transmission of these sequences led to ~8% of the human genome being of HERV origin. HERVs do not produce infectious particles, but partial expression of retroviral genes has been linked to human physiology and, if aberrantly expressed, to disease (13). A significant example of that is the role of the env gene of certain endogenous retroviruses in human placental morphogenesis and fetomaternal tolerance (14).
The recent advances in genetic sequence detection and analysis have resulted in the realization that humans are not only colonized by diverse prokaryotic and eukaryotic microorganisms, which constitute the microbiome, but also by a marked multitude and variety of viruses, collectively termed the virome. Research has been performed on the composition and dynamics of the virome, and its role in human health and disease. The virome consists of viruses that infect human host cells and phages that infect bacteria of the microbiome (15). Viruses affect the host directly by interfering with the defense mechanisms, whereas phages may exert an indirect effect on host physiology and disease by modulation of the composition and fitness of the bacterial microbiome (16).
The aforementioned information poses a challenge for the scientific community related to the understanding of the role of viruses in human health and disease. It is known that the concept of viruses is more extensive than their exclusive perception as mere agents of an acute infection, or some chronic debilitating diseases, such as AIDS or some forms of cancer. Notably, a causal connection between viruses and disease pathophysiology may exist; however, there are still a number of questions that need to be answered. A number of hypotheses have been put forward regarding viruses and diseases, the etiology of which was previously unknown, but it remains unclear as to how to establish a causal connection. In addition, it remains to be determined how far research has gone regarding the possible role of viruses in chronic, debilitating diseases such as psychiatric disorders; and how many, and which, viruses may contribute to the development of psychiatric disease. Considering the great diversity of the infectious properties of different viruses, research is also required regarding how a viral infection could trigger a mental disorder; whether some viruses have specific virulent properties that have not been described so far and dictate previously unrecognized pathophysiological pathways; and whether these new associations with disease are the result of direct damage and its sequelae in the long-term, or chronic infection. Furthermore, it remains to be ascertained as to whether detection is enough, and how to detect viruses in a chronic state of infection, where viral load is frequently imperceptible and symptoms are initially obscure, developing years after viral infection. The role of the virome and endogenous retroviruses in psychiatric and other diseases also requires further assessment.
Suggested mechanisms and tentative data about the possible association of viral infections with psychiatric disease
Most clinical changes in an individual's cognition and mental status are heterogeneous and attributed to the synergistic action of a multitude of different factors. Genetic predisposition and environmental perturbations, by either stress from the social surroundings or alterations in the physiological state of the organism, have been proposed as significant contributing factors in the development of psychiatric disease (Fig. 2) (17). Such environmental influences may include direct insults or indirect sequelae that follow infection from viruses such as influenza, enteroviruses, arboviruses and several herpesviruses, or the differential expression of HERVs.
Viruses may contribute to psychiatric diseases such as schizophrenia, autism, major depression, bipolar disorder and chronic fatigue syndrome through various mechanisms (18). Thus far, these tentative mechanisms generally fall into two categories (Fig. 2): Indirect immune, inflammatory, metabolic and degenerative processes in the central nervous system (CNS), and/or direct neurotoxic effects following viral invasion into the CNS (19).
Indirect effects of viral infection on the physiology of CNS
Virus-induced psychopathology linked to neurodevelopmentA number of different viruses have been implicated in a single psychiatric disease, such as schizophrenia, and this may constitute an immune-mediated disorder, rather than a pathogen-specific link between viral infection and a psychiatric disorder (20). The model of neurodevelopment disruption has been suggested as the most promising mechanism underlying the onset of schizophrenia (19). Both direct viral infection and indirect viral effects, via the activation of inflammatory pathways and the excessive release of cytokines, have been suggested as possible disrupting mechanisms in normal neurodevelopment (21). Moreover, the timing of disruption of normal neurological processes that occur at critical stages of neurodevelopmental processes has been proposed as being of utmost importance. Specifically, effects incurred by viruses appear to be stronger when infection occurs during the late 1st and the 2nd trimesters of gestation (19), childhood and pre-adolescence (22,23). Several studies have also shown that childhood viral infection of the CNS, especially under the age of 3 years or in pre-adolescence, may be significantly associated with an increased risk for later psychosis (22,24).
However, virus-induced disruption during both pre- and postnatal neurodevelopmental processes is not solely responsible for the possible development of a psychiatric disease. Psychological trauma during adolescence or early adulthood, in conjunction with previous maternal or childhood viral infection during critical stages of neurodevelopment, has been linked with greater possibilities for the development of schizophrenia (25,26).
Activation of inflammatory pathways following infection and their link with psychiatric disease
It has been suggested that systemic or direct CNS infections may activate neuroinflammatory events mediated by activated microglia and the release of cytokines in the brain (27,28). In the case of a systemic viral infection through, for instance, the circulatory, respiratory and the gastrointestinal routes, an initial inflammatory event may ensue that leads to the release of proinflammatory cytokines and chemokines that can disrupt the integrity of the blood-brain barrier (BBB), rendering it permeable to viral particles, toxins and infected immune cells (Fig. 3) (28). Other neurotropic viruses, such as α-herpesviruses and rhabdoviruses, invade the CNS directly; therefore, a correspondingly direct neuroinflammatory event may be elicited (Fig. 3). In both cases, oligodendrocytes, astrocytes and microglia constitute the main mediators of inflammation in the brain (28).
Extensive data from clinical and experimental models have shown the induction of proinflammatory cytokines following infection with several viruses, such as flaviviruses, including Dengue virus (DENV) and tick-borne encephalitis virus (29,30). Japanese encephalitis virus (JEV) is another flavivirus that infects microglial cells, which may serve as a reservoir for the virus (31). In addition, Zika virus (ZIKV), as well as directly damaging neuronal cells, may mediate an exaggerated neuroinflammatory response, especially in neonates; postnatal microcephaly has been linked with ZIKV-induced neuroinflammation in both animal models and fatal cases (32,33). Another example is the neuronal damage caused by the rabies lyssavirus (RABV), which has also been suggested to be the result of the high production of inflammatory cytokines in primary astrocytes and microglia (34).
Regarding the available evidence regarding the role of these neuroinflammatory events in influencing neurobiological processes, potentially contributing to the development of schizophrenia and related psychotic disorders, elevated proinflammatory cytokines have been found in both the blood and cerebrospinal fluid (CSF) of patients with cognitive deficits (35,36). Notably, serum interleukin (IL)-6 and C-reactive protein in both childhood and adolescence have been shown to be linked to both the presence and severity of disease during adulthood (37,38). IL-6 is also considered not only as an important, diagnostic inflammatory marker but as a predictive one as well, since its increased levels during both childhood and adolescence have been associated with a high risk for adult psychotic disorders in a dose-dependent manner (39). IL-8 in the CSF has also been described as a sensitive marker of inflammation for numerous neurological and psychiatric diseases, whereas higher IL-12p70 levels have been detected in the serum of patients with schizophrenia (40).
Complement factors in the brain are also known to contribute to the process of synaptic pruning, which involves enhanced activation of proinflammatory cytokine secretion by glial cells, leading to possible neuronal cell damage and death (41). Moreover, activated complement may contribute to alterations in the BBB function, which could subsequently influence the progress of a psychiatric disease (42). Notably, elevated levels of complement have been detected in patients following first-episode psychosis (43,44).
Systemic, or CNS-specific viral infection and the inflammatory cascades that ensue may not exclusively affect brain physiology, they may also influence hypothalamic-pituitary-adrenal (HPA) axis dysregulation, which has been linked to psychiatric diseases such as schizophrenia (45). HPA axis dysregulation, and brain neuropathology induced by proinflammatory cascades elicited by either direct or systemic infections, have a common trait; they mostly seem to render the organism more vulnerable to a possible psychiatric disorder but do not lead to the disease on their own. An environmental, psychosocial stimulus in later life may more readily affect an individual primed by such inflammatory complications (46).
Genetic predisposition in human leukocyte antigen (HLA) genes and immune response to infections that may prime a psychiatric disease
HLA genes located on chromosome 6 code for cell surface proteins that serve a fundamental role in immune responses against foreign antigens. Class I and II HLA molecules represent two major pathways that act in synergy for host protection (47). Genome-wide research on the genetic background of schizophrenia has provided evidence that genetic polymorphisms within the HLA genes located on chromosome 6are associated with the disease (47). Specifically, polymorphisms involving HLA-DRB1 alleles of Class II antigens have been significantly linked with schizophrenia (48). There are polymorphisms, however, which are negatively associated with schizophrenia (49) or even have a protective effect (50), at least amongst certain ethnic populations.
As a consequence of the pivotal role that HLAs have in foreign antigen presentation and final elimination by the immune system, an ineffective, polymorphic version of an HLA allele may lead to more pronounced, adverse effects on the brain and the persistence of viral antigens, with poor consequences regarding inflammation and/or autoimmunity with respect to the development of psychiatric disease. For example, in a recent study, it was suggested that HLA alleles with a higher affinity for herpesviruses, leading to a more effective elimination of the consequences induced by acute infection, may confer protection against schizophrenia (51).
Viral infection could impair CNS energy metabolism, which may lead to psychiatric disease
As obligate intracellular entities, viruses rely exclusively on host cell metabolic machinery to replicate and sustain their existence. Notably, neurotropic viruses may disrupt brain physiology by altering neuron and astrocyte metabolism; they have evolved to alter host cell metabolic activities so that they can optimally be used for their own benefit, such as glycolysis, the pentose phosphate pathway, the tricarboxylic acid cycle and oxidative phosphorylation (52). For example, there is increasing evidence that ZIKV infection suppresses neuronal stem cell proliferation and induces premature differentiation by intervening with glycolysis and oxidative phosphorylation properties of the host cells, leading to microcephaly of neonates during pregnancy (53–55). Moreover, a reduction in glutamine metabolism in astrocytes following a viral infection may disrupt their cooperation with neurons to maintain the glutamate-glutamine cycle in the brain, something that may shape brain activities and lead to neuropsychiatric disorders (56,57). For instance, the release of Tat protein by infected cells in HIV-associated neurocognitive disorders (HAND) has been linked with disruption of this metabolic cooperation between astrocytes and neurons (58,59). The same has also been proposed for SARS-CoV-2, which may also be found in the brain and mainly infects astrocytes (56,57). Finally, another means for impairment of neuronal metabolism may be elicited by a viral infection through alteration of mitophagy and mitochondrial dynamics, which has been reported for HIV-1 proteins that inhibit mitophagic flux in human primary neurons, leading to neuronal damage (60).
Direct impairment of the CNS by neurotropic virus infection
Evidence and suggestions were previously outlined above regarding the indirect effects of viral infections on inflammatory pathways and neurodevelopmental processes that may lead to severe neuropsychiatric disorders in the long-term and not immediately after infection. Moreover, these sequelae do not appear on their own; viral infections and their effects could prepare the organism to respond more readily to an appropriate environmental stimulus that would act in synergy to mount the emergence of a psychiatric disease, later on in the individual's life.
However, a number of viruses have evolved to invade the CNS directly via numerous pathways, including leukocyte-mediated transfer through the BBB, neuronal axon transport from peripheral nerves and invasion of CNS endothelial cells, or the olfactory nerve (61). Therefore, the possible role of direct damage inflicted upon the CNS by these neurotropic viruses in psychiatric disease should be assessed. So far, numerous neurotropic viruses that belong to a diverse number of families have generally been linked to psychiatric disease, and these include herpesviruses, enteroviruses, arboviruses and retroviruses, as well as respiratory viruses that also exhibit a degree of neurotropism, such as influenza and coronaviruses, (52,62–64). Clinical and in vitro studies have shown that these viruses can be directly neurotoxic via a number of diverse mechanisms, including manipulation of apoptosis and autophagy, production of reactive oxygen species (ROS) and disruption of the production of antioxidants, localized inflammation in the central and peripheral nervous system (neuroinflammation), and alterations in neurotransmission (52,65).
Apoptosis and autophagy during viral infection
Apoptosis and autophagy constitute two important defense mechanisms against viral spread. It has been reported that the apoptosis signaling pathway is activated in pediatric patients with HIV-1 encephalitis (66), as well as in infections caused by enterovirus A71(EV-A71) (67), RABV (68–72), West Nile virus (WNV) (73–77) and JEV (78,79). Some viruses may evade degradation and the subsequent elimination by the immune system through inhibition of autophagy, but others modulate autophagy by exploiting autophagosomes and their secretion pathway as sites for replication, viral particle maturation and release from the host cell (80). For example, the VP1 capsid protein of EV-A71 is not only a major modulator of viral antigenic properties and interaction with the immune system, but it also contains important neurovirulence properties that induce autophagy by regulating the mammalian target of rapamycin signaling pathway to promote viral replication (81–83). Modulation of autophagy by the HIV-1 proteins Nef and Tat has also been evaluated and proposed, which may contribute to HAND (84–86).
Oxidative, mitochondrial and endoplasmic reticulum (ER) stress following viral infection
Infection by neurotropic viruses has been associated with excessive production of ROS and deficient cellular antioxidant defenses (52). JEV is a significant example of such a neurotropic virus that increases ROS production; it has been suggested as an important contributor to psychiatric, or other neurologic sequelae, in ~50% of survivors (87,88). Other neurotropic viruses associated with excessive oxidative stress include RABV, EV-A71, which is prevalent in South East Asia, and ZKV (52). Moreover, oxidative stress is not exclusively seen in viruses causing mainly acute infections, but is also a characteristic of viruses causing latent, or persistent infections, such as herpes simplex virus (HSV) and HIV. HSV-1 infection and intracellular ROS generation have been observed in microglia (89) and neural cells (90). Glutamate-mediated excitotoxicity is also an important mechanism of neuronal injury by viral infection. The effect of HIV-1 proteins on neurotoxicity induced by glutamatergic system dysregulation has also been revealed (91).
Several in vitro studies have also shown that infection with certain viruses may induce mitochondrial and ER stress (52). For instance, as well as inducing neuronal damage via neuroinflammation, RABV has been reported to invoke significant alterations in different mitochondrial parameters (92). In vitro studies in human neural stem cells have also shown that JEV, ZIKV and WNV infection may promote the expression of ER stress-related proteins (93–95), whereas the enterovirus EV-A71 may induce oxidative stress both in the ER (96) and mitochondria (97), the latter leading to morphological changes and subsequent functional anomalies in glioblastoma cells.
Viral infection could interfere with neurotransmission
Neurotropic viruses may alter the levels of specific neurotransmitter levels, leading to dysregulation of synaptic nerve signal transmission and subsequent impairment of specific brain functions that may trigger psychotic disorders (52). Specifically, viruses affect pathways that involve dopamine metabolism (98,99) and glutamate transmission via molecular mimicry at N-methyl-D-aspartate receptors (100). JEV infection can significantly increase dopamine production and modulate the rate-limiting enzyme of dopamine biosynthesis, with an increase in phosphorylated tyrosine hydroxylase levels at the early stage of infection (101). JEV may also exploit dopamine-mediated neuronal communication to increase the susceptibility of dopamine D2 receptor-expressing cells to JEV infection, which causes damage to dopaminergic neuron-rich areas such as the thalamus and midbrain, leading to neuronal loss and increasing the fatality rate of JEV-infected mice (101).
The quest for establishing an etiological link between viral infection and psychiatric diseases: Conjectures and evidence
The present review has thus far provided a summary of the possible mechanisms with which systemic and/or neurotropic viral infections may contribute to the development of psychiatric disease. Nevertheless, determination of the presence of a virus within the host during an acute, persistent or latent infection that evolved into an early or a delayed psychiatric abnormality is essential to establish the possible contribution of viruses to psychiatric abnormalities. Either seroepidemiological analysis or the direct detection of viral genetic material in appropriate clinical samples, such as the CSF, have been employed thus far in CNS and systemic infections that were subsequently linked to a psychiatric disease. Significant rates of neuropsychiatric manifestations (43%) have been found in HSV-2, varicella zoster virus (VZV) and enterovirus meningitis cases (102–104). Cognitive impairment in adults shortly after aseptic meningitis and encephalitis caused by HSV, VZV, enteroviruses, HIV, ZIKV and coronaviruses has also been reported (105,106).
Epidemiological and serological evidence about the role of specific viruses in psychiatric disease
Regarding viral infection and its role in the disruption of normal neurodevelopment, as long as 4–5 decades ago, there were initial reports on the greater incidence of patients with schizophrenia who were born during late winter and spring when the risk for respiratory tract infections, and most notably influenza virus infection, is significantly greater (107,108). Numerous studies have subsequently supported this observation (109,110). A significant risk for schizophrenia has also been reported for patients whose mothers were exposed to influenza epidemics during the 2nd trimester of gestation (111). Prenatal exposure to rubella is another suggestion for the increased risk of schizophrenia in the offspring of affected mothers (112). A high frequency of psychiatric disorders has also been reported for patients with a history of chronic hepatitis C virus (HCV) infection (113). Additionally, anti-viral treatment and final HCV clearance have been shown to positively affect the quality of life in these patients (114). Psychotic diseases, such as major depressive disorder, anxiety disorder and HAND, have commonly been reported amongst patients with HIV; however, not much information is currently available about a possible association between HIV infection and schizophrenia (46). Inconsistent results have been reported from such epidemiological studies and their role in identifying the viral connection in psychiatric diseases. For instance, in another large-scale study, no significant association was identified between maternal infection before, during or after the 1957 influenza epidemic, and the development of schizophrenia in offspring (115).
Further serological methods have been employed to decipher an association between prenatal exposure to influenza and an increased risk of schizophrenia. For example, one study based on serological investigation of the mothers of psychiatric patients indicated that the risk of schizophrenia was increased in people exposed to the influenza virus within the uterus during the first trimester of pregnancy (112).
Members of the Herpesviridae family are also very well known for their neurotropic properties and their persistence in the host, marked by alternating periods of latency and possible reactivation (116). Elevated IgG antibody levels against HSV-1 and/or cytomegalovirus (CMV) have also been associated with a first episode of schizophrenia, brain morphological changes, increased suicidal tendencies and cognitive impairment (117,118). In another, large-scale serological study of CMV IgG detection in plasma samples from psychiatric patients, high rates of antibody detection (61%) were associated with a variety of different psychiatric disorders (119). An aberrant response to infection by Epstein-Barr virus (EBV) has also been reported in patients with schizophrenia who have been shown to exhibit elevated levels of antibodies against the viral capsid antigen of the virus, but not against the EBV nuclear antigen-1 (120). However, other studies have shown inconsistent results regarding the potential association of a previous herpesvirus infection with psychiatric disease. In certain studies, no significant association between HSV-2, HSV-6 and VZV infections, and schizophrenia was found (121–123), whereas elevated antibody titers against Chlamydia trachomatis were more significantly correlated with schizophrenia than antibodies against other herpesviruses (124).
Arboviruses have also been implicated as etiological agents of psychiatric morbidity in patients with clinical and/or serological evidence of infection. For instance, cognitive difficulties have previously been reported amongst 49 patients with clinical and laboratory classification of a WNV infection (64). Persistence of these cognitive deficits for an extended period of time and their prevalence, irrespective of whether the infection was neuroinvasive or not, has been shown to advocate a systemic pattern of neuropsychiatric disorder manifestation, rather than the result of direct neurotoxicity of WNV. Chikungunya virus is another arbovirus that has been linked with depression, anxiety and somatoform disorders (60), although it has been suggested that prenatal and postnatal exposure to the virus may not be associated with impaired neurodevelopment (125). Nevertheless, such studies regarding the role of these and other arboviruses, such as ZIKV or DENV, in psychiatric disorders are still scarce and further studies must be conducted on larger sample sizes and the pathophysiology of arbovirus infection with psychiatric sequelae.
Direct evidence of the possible implication of viral infection in psychiatric disorders
Detection of viral genes and their products in clinical samples of psychiatric patientsMost of the viral infections that have been recognized so far as contributing factors in the etiology of psychiatric disease are based on epidemiological and serological analyses that provide speculative, and not definitive, evidence of an association between viruses and psychiatric disease. Psychiatric disorders often develop late, after viral infections during critical neurodevelopmental stages. This, combined with other environmental factors, makes it difficult to study the direct connection if viruses to psychiatric disease during the acute infection phase. Moreover, viral persistence through latency means that any viral load, which may be linked with direct neurotoxic, or indirect inflammatory and neurodevelopmental processes that could lead to a psychiatric disease, remains at almost imperceptible levels, posing a significant challenge for their detection, even when using the most modern, state-of-the-art molecular methodologies.
More compelling evidence for the role of viruses in psychiatric diseases comes from studies that detect and analyze viruses directly in the brain or in CNS-associated clinical samples, such as the CSF. These studies use highly sensitive molecular techniques, such as polymerase chain reaction (PCR) and next-generation sequencing (NGS), which can identify viral genes and products even at low levels. The potential conferred by these techniques in accurately detecting and characterizing viral genes and products, even when minimum viral loads persist during a dormant part of the biological cycle of the virus, is significant. A number of viruses, which are not obligate neurotropic viruses, may also be recognized, such as HCV. The psychopathology behind HCV infection is complex and poorly understood, involving activation of glial and local/systemic inflammatory pathways, as well as metabolic disruptions (126). However, HCV RNA has been detected in brain autopsy samples of patients at a 3–5 log lower load than in hepatocytes (127). Moreover, HCV RNA has been found in astrocytes and microglial cells (128), which may indicate viral spread within the CNS despite the fact that these cells do not express the appropriate receptors on their surface for HCV entry. In addition, HCV RNA has been detected in peripheral blood mononuclear cells (PBMCs), which may conditionally transport viral particles across the BBB (129).
Novel sequencing techniques, based on NGS and whole genome sequencing methodologies, provide a powerful means to delve into the heterogeneous viral populations of the virome and/or acutely infecting viruses within a host with psychiatric disorders, providing a fundamental basis for further analyses and studies of their possible relationship with the disease. However, few studies which either report inconsistent, varying results or are largely incomplete, have been performed to date, and these have reached controversial conclusions about the connection of viruses with psychiatric disease. For instance, in one study, whole-genome and RNA sequencing were employed for the investigation of viral infection directly in brain tissues and its association with schizophrenia, bipolar disorder and autism spectrum disorder (130). Samples from a large number of patients were analyzed; however, despite detection of diverse neurotropic DNA and RNA viruses in the brains of patients with major psychiatric disorders, such as herpesviruses, polyomaviruses, retroviruses, adenoviruses and others, neither significant qualitative (types of viruses detected) nor quantitative (viral load) differences were detected between patients and controls. Therefore, no association between viral infection of the brain and major psychiatric disorders could be inferred.
Another study based on novel sequencing technologies reported on the development of a sequence-capture method and an appropriate bioinformatics analysis pipeline that may perform detailed analysis of the human virome in a variety of clinical samples, with the aim of investigating its possible association with psychiatric disease (131). Detection of HIV-1, Torque teno virus, pegivirus, herpesvirus and human papillomavirus sequences were reported in PBMC, plasma and stool samples, but only Torque tenovirus was detected in psychiatric case samples and not in controls. A limitation of this previous study, however, was the fact that a limited number of samples and patients were examined and, notably, only peripheral samples were analyzed; since viruses present in the CNS may not be present in peripheral tissues, the study of CSF samples with the proposed method may enhance identification of viral activity in the CNS of psychiatric patients (131).
Evidence from research on models using experimental animals
Several studies on experimental animal models have been employed during the last two decades in an attempt to support the viral model of neuropsychiatric abnormalities. For instance, in such an experiment, transgenic mice that were genetically manipulated in order to express the phosphoprotein of Borna disease virus (BDV) in glial cells showed a significant reduction in synaptic density, and the expression of brain-derived neurotrophic factor and serotonin receptor, as well as a decrease in synaptic density (132). These mice then developed behavioral abnormalities, such as hyperactivity, increased aggressiveness between males and spatial reference memory deficit. In summary, the experiment showed that the expression of BDV phosphoprotein in glial cells may directly induce neuronal degeneration that could lead to disorders resembling those of psychiatric patients. In another model of schizophrenia, where mice were experimentally infected with influenza virus at embryonic days 7, 9, 16 and 18, the mice exhibited significant postnatal brain structural abnormalities that led to abnormal behavior (133). An experimental study carried out with conventional mice (adult male and female C57BL/6J, 129X1/SvJ and nude mice Foxn1nu/Foxn1n, all 8–12 weeks old), infected with chimeric EcoHIV, as a model to reproduce physiological conditions for the development of disease in people on antiretroviral therapy (ART), revealed that ART can prevent AIDS, but not HIV-associated neuropathogenesis (134). This study suggested that, although HIV replication is suppressed by ART, the persistence of integrated, replication-competent HIV in T cells and macrophages may serve an important role in neurocognitive and behavioral aberrations.
HERVs and their role in psychiatric disease
HERVs pose a different challenge from the classical problem of proving a possible association of viral infections with psychiatric disease. They are not the mediators of a viral infection, they are part of the human virome and, as retroviruses, they have permanently integrated cDNA sequences of their RNA into the human genome, comprising 8% of its total nucleotide composition. Therefore, the key point is not to prove any kind of infection by HERVs, but, since partial expression of retroviral genes has been linked to either human health or disease, to determine the association between HERVs and psychiatric disease, HERV transcripts and/or their products should be detected and characterized in appropriate clinical samples (135–138).
Aberrant expression of HERVs has recently been identified in schizophrenia (139). Upregulated transcripts of the polymerase gene of the HERV-W family have also been detected by quantitative PCR methods in the CSF samples from a significant proportion of patients with schizophrenia, or schizoaffective disorder, as well as in the frontal cortex of brains from individuals with schizophrenia (135). In addition, elevated levels of HERV-W transcripts and proteins have been reported in the blood, CSF and brains of patients with bipolar disorder (136,137). In another study, a more detailed association between HERVs and psychiatric disease was reported, where patients with schizophrenia and bipolar disorder could be differentiated into subgroups with differing inflammatory and clinical profiles (e.g. earlier disease onset), on the basis of HERV-W family env protein antigenemia and cytokines (140).
Most studies carried out thus far on the potential association of HERVs with psychiatric disease have implicated HERV-W as the most frequently associated family (138). However, insufficient evidence is available due to the currently limited number of studies and the small sample sizes that have been employed in these studies. Further studies are required in order to obtain more unambiguous data regarding the possible role of HERVs.
The paradigm of SARS-CoV-2: An interplay of neuroinvasive, inflammatory and environmental processes
It has been suggested that the neuroinvasive properties of SARS-CoV-2 in conjunction with hyperactivity of inflammatory responses may have an impact on the emergence of neuropsychiatric disorders during both the acute and post-infectious phases of COVID-19 (141). Specifically, in long COVID syndrome, it has been proposed that neuropsychiatric sequelae may be mainly associated with inflammatory, metabolic and degenerative processes in brain areas where the presence of the virus receptor ACE2 is significant, extending from the somatosensory cortex to the rectal/orbital gyrus, the temporal lobe, the thalamus and hypothalamus, and further, to the brainstem and cerebellar regions (142). Moreover, the socioeconomic impact of the pandemic has imposed a significant strain on human societies and individual lives due to strict lockdowns, social distancing or isolation measures, incomplete physical health recovery and a worldwide economic crisis. In addition, it has been emphasized that individuals with intellectual and developmental disabilities may be at a greater risk of mental health deterioration during COVID-19 due to the direct effect of anxiety caused by the pandemic itself and the media, as well as to restrictions brought about by lockdown regulations (143–145). These showcase the indirect effects of SARS-CoV-2 on the increase in the prevalence of neuropsychiatric disorders, which differ from the purely biological, neuroinvasive properties of the virus (146–148). Therefore, it is crucial to broaden the current knowledge about the persistence of psychiatric disorders in the post-acute phase of infection, in order to distinguish between the social and the pathophysiological origin of emergent cases, and to develop an appropriate, targeted approach for patients.
In a previous study, it was estimated that the incidence of neurological or psychiatric diagnosis within 6 months after a SARS-CoV-2 infection was 33.62% in ~250,000 patients (149). Depression and anxiety have been reported as the most common psychiatric symptoms in patients with long COVID (150,151). Notably, post-traumatic stress disorder (PTSD) has been recorded following recovery from serious epidemic events in the past and the same applies to the most recent COVID-19 pandemic (152). Sleep disturbances, fatigue, memory impairment, concentration, disorientation, confusion and other serious disorders have been associated with long COVID with debilitating effects on everyday life.
Conclusion
The association of viruses with psychiatric diseases still remains controversial, since we have not crossed the borderline from hypothesis to proof yet. In most cases of viral disease and epidemiology, establishing a specific virus as the causative agent depends fundamentally on confirmation in the laboratory. However, most evidence available thus far regarding the possible role of viruses in the development of psychiatric disorders comes mainly from observational, epidemiological and serological studies. A number of older diagnostic approaches, such as serological demonstration of elevated antibody titers against known viruses, are of limited value. They frequently lack high specificity, constitute indirect proof of viral infection, and are ineffective in characterizing novel viruses as potential causes of psychiatric disease.
Accurate diagnosis is complex with a number of parameters, including the patient's medical history and immune status, appropriate sample type and timing of collection, and the methods used. Despite significant advances in diagnostic laboratory capabilities due to technological advancements in molecular methodologies, challenges remain. These advancements have enabled the high throughput of prompt and more accurate diagnostic results, reinforcing the liaison between laboratory and clinical staff. Quantitative results of viral loads and genotypic characterization of strains relevant to psychiatric or other diseases can now be obtained, further enhancing the clinical utility of diagnostic results. Moreover, they have enabled the recognition of emergent, re-emergent or known viruses that have never been associated with psychopathology before. However, studies focusing on direct virus detection in appropriate clinical samples are still limited, hindering the direct causal connection with disease onset.
Due to notable and inherent difficulties in both virus detection and the establishment of an etiological connection, satisfactory results are yet to be obtained. Firstly, detection of neurotropic viruses is hampered by their ability to establish chronic infections, either latent or with a late-onset. It is frequently difficult to determine the insidious persistence of such neurotropic viruses for months or years within the CNS, something which is attributed to their very low viral loads, particularly during dormancy. Moreover, irrespective of whether there is a direct neuroinvasive effect, or indirect activation of microglial cells and astrocytes through systemic inflammatory and neuroinflammatory processes, there is currently a lack of appropriate methodological approaches to sufficiently understand the progress of these neurotropic infections and the ensuing pathophysiology.
Secondly, the neurodevelopmental model of psychiatric disease suggests that transient viral infections at critical stages during early childhood or the prenatal period may lead to psychiatric disease. However, obtaining and preserving appropriate samples from these critical neurodevelopmental periods is difficult, especially when such viral infections are asymptomatic or subclinical.
Thirdly, the complexity of the virome and the existence of thousands of viral species, a number of which have not yet been fully described, adds to the challenge. The knowledge of viral diversity remains incomplete and numerous novel viruses are yet to be discovered. Additionally, RNA viruses evolve rapidly and novel strains with potential neuropsychiatric virulence, such asSARS-CoV-2, may emerge.
Finally, psychiatric diseases are multifactorial disorders involving a complex mixture of genetic and environmental influences. Viral agents may prime an organism for the development of psychiatric disease rather than causing it directly. Establishing the viral connection is challenging, deviating significantly from a simplistic, direct connection between infectious cause and disease, as described by Koch's postulates (153).
Further experimental research, including larger population sizes, and modern molecular methodologies accompanied by appropriate bioinformatics analysis, is essential for elucidating the complex interactions among genes, pathogens and the immune system in the etiology of psychiatric disease. The advent and further development of multi-omics technology, which collectively includes genomics, transcriptomics, proteomics, metabolomics, interactomics, epigenomics and pharmacogenomics, may enhance understanding of virus-host interactions; decipher the pathophysiological mechanisms that underlie the role of viruses in various diseases, such as psychiatric disease; identify effective antiviral treatments and vaccines; and identify reliable biomarkers of viral infections (154). Single-cell analysis by multi-omics is particularly promising in the identification of molecular characteristics of pathophysiological processes within specific cell types, which, for example, may be infected by a virus, and how these characteristics are associated with a specific phenotype, such as a psychiatric disease. However, there is still the challenge of analyzing the large amount of biological information collected by single-cell multi-omics, especially when comparison of data from different cell types is made, something which greatly relies on the development of appropriate bioinformatics tools. Moreover, these elaborate molecular methods need to be supplemented with accurate in vitro models to obtain a better understanding of the pathophysiology of viral infections in association with neuropsychiatric disorders.
Most of the currently available information about the course of viral pathogenesis in the CNS is limited since it originates only from lesions analyzed by MRI data and postmortem histology (155). Despite the fact that experiments with organoids have been promising for the evaluation of viral dissemination between neuronal cells and the activation of astrocytes, their low level of complexity cannot simulate all cellular factors involved in neuroinvasion and inflammation (156). Moreover, the currently available experimental animal models may not fully replicate the course of neuropathologic conditions in humans for a very important reason. These experiments are carried out under carefully controlled laboratory conditions and may not accurately represent the complexity of those interactions between the human host, the environment and the viruses that may lead to psychiatric disease. More elaborate models are needed that will take into account as many experimental parameters as possible and will more accurately represent these complex interactions. The continuous emergence of novel viral variants with unprecedented virulence potential, as was evident with the recent COVID-19 pandemic, may also add more complexity to the design of satisfactory experimental models. Further studies are needed on the study of viral tropism, and progress before the onset of symptoms and during the course of infection and, secondly, on the identification of genetic and epigenetic host factors that may lead to CNS impairment and subsequent neuropsychiatric disease (91). More extensive, prospective studies are also needed to follow the sequelae of acute viral infections of the CNS in conjunction with other environmental stimuli that may influence the development of psychiatric diseases. By addressing these challenges, research may move closer to understanding the role of viruses in psychiatric disorders, potentially developing targeted interventions that could mitigate their impact.
Acknowledgements
Not applicable.
Funding
Funding: No funding was received.
Availability of data and materials
Not applicable.
Authors' contributions
NS and CA were major contributors in writing the manuscript. SP, AT, EA and SK were contributors in writing and critical revision of the manuscript. DAS was a contributor in critical revision of the manuscript. ER was a major contributor in providing the subject of this review manuscript, as well as in writing and critical revision of the manuscript. Data authentication is not applicable. All authors have read and approved the final version of the manuscript.
Ethics approval and consent to participate
Not applicable.
Patient consent for publication
Not applicable.
Competing interests
DAS is the Editor-in-Chief for the journal but had no personal involvement in the reviewing process, or any influence in terms of adjudicating on the final decision, for this article. The authors declare that they have no competing interests
References
Nasir A, Forterre P, Kim KM and Caetano-Anollés G: The distribution and impact of viral lineages in domains of life. Front Microbiol. 5:1942014. View Article : Google Scholar : PubMed/NCBI | |
Call L, Nayfach S and Kyrpides NC: Illuminating the virosphere through global metagenomics. Annu Rev Biomed Data Sci. 4:369–391. 2021. View Article : Google Scholar : PubMed/NCBI | |
Gregory AC, Zayed AA, Conceição-Neto N, Temperton B, Bolduc B, Alberti A, Ardyna M, Arkhipova K, Carmichael M, Cruaud C, et al: Marine DNA viral macro- and microdiversity from pole to pole. Cell. 177:1109–1123.e14. 2019. View Article : Google Scholar : PubMed/NCBI | |
Neri U, Wolf YI, Roux S, Camargo AP, Lee B, Kazlauskas D, Chen IM, Ivanova N, Zeigler Allen L, Paez-Espino D, et al: Expansion of the global RNA virome reveals diverse clades of bacteriophages. Cell. 185:4023–4037.e18. 2022. View Article : Google Scholar : PubMed/NCBI | |
Mohapatra S and Menon NG: Factors responsible for the emergence of novel viruses: An emphasis on SARS-CoV-2. Curr Opin Environ Sci Health. 27:1003582022. View Article : Google Scholar : PubMed/NCBI | |
Marie V and Gordon ML: The (Re-)emergence and spread of viral zoonotic disease: A perfect storm of human ingenuity and stupidity. Viruses. 15:16382023. View Article : Google Scholar : PubMed/NCBI | |
Piret J and Boivin G: Pandemics throughout history. Front Microbiol. 11:6317362021. View Article : Google Scholar : PubMed/NCBI | |
Davis HE, McCorkell L, Vogel JM and Topol EJ: Long COVID: Major findings, mechanisms and recommendations. Nat Rev Microbiol. 21:133–146. 2023. View Article : Google Scholar : PubMed/NCBI | |
Boufidou F, Medić S, Lampropoulou V, Siafakas N, Tsakris A and Anastassopoulou C: SARS-CoV-2 reinfections and long COVID in the post-omicron phase of the pandemic. Int J Mol Sci. 24:129622023. View Article : Google Scholar : PubMed/NCBI | |
Castanares-Zapatero D, Chalon P, Kohn L, Dauvrin M, Detollenaere J, Maertens de Noordhout C, Primus-de Jong C, Cleemput I and Van den Heede K: Pathophysiology and mechanism of long COVID: A comprehensive review. Ann Med. 54:1473–1487. 2022. View Article : Google Scholar : PubMed/NCBI | |
Virgin HW, Wherry EJ and Ahmed R: Redefining chronic viral infection. Cell. 138:30–50. 2009. View Article : Google Scholar : PubMed/NCBI | |
Grandi N and Tramontano E: HERV envelope proteins: Physiological role and pathogenic potential in cancer and autoimmunity. Front Microbiol. 9:4622018. View Article : Google Scholar : PubMed/NCBI | |
Zhang M, Liang JQ and Zheng S: Expressional activation and functional roles of human endogenous retroviruses in cancers. Rev Med Virol. 29:e20252019. View Article : Google Scholar : PubMed/NCBI | |
Luganini A and Gribaudo G: Retroviruses of the human virobiota: The recycling of viral genes and the resulting advantages for human hosts during evolution. Front Microbiol. 11:11402020. View Article : Google Scholar : PubMed/NCBI | |
Liang G and Bushman FD: The human virome: Assembly, composition and host interactions. Nat Rev Microbiol. 19:514–527. 2021. View Article : Google Scholar : PubMed/NCBI | |
Seo SU and Kweon MN: Virome-host interactions in intestinal health and disease. Curr Opin Virol. 37:63–71. 2019. View Article : Google Scholar : PubMed/NCBI | |
van Os J, Rutten BP and Poulton R: Gene-environment interactions in schizophrenia: Review of epidemiological findings and future directions. Schizophr Bull. 34:1066–1082. 2008. View Article : Google Scholar : PubMed/NCBI | |
Pearce BD: Viruses and psychiatric disorders. Siegel A and Zalcman SS: The Neuroimmunological Basis of Behavior and Mental Disorders. Springer; Boston, MA: pp. 383–410. 2009, View Article : Google Scholar | |
Hobbs JA: The virus connection: How viruses affect psychiatric pathologies. Psychiatr Times. 33:2016.PubMed/NCBI | |
Müller N and Schwarz MJ: Immune system and schizophrenia. Curr Immunol Rev. 6:213–220. 2010. View Article : Google Scholar : PubMed/NCBI | |
Fatemi SH and Folsom TD: The neurodevelopmental hypothesis of schizophrenia, revisited. Schizophr Bull. 35:528–548. 2009. View Article : Google Scholar : PubMed/NCBI | |
Blomström Å, Karlsson H, Svensson A, Frisell T, Lee BK, Dal H, Magnusson C and Dalman C: Hospital admission with infection during childhood and risk for psychotic illness-a population-based cohort study. Schizophr Bull. 40:1518–1525. 2014. View Article : Google Scholar : PubMed/NCBI | |
Hickie IB, Banati R, Stewart CH, Stewart CH and Lloyd AR: Are common childhood or adolescent infections risk factors for schizophrenia and other psychotic disorders? Med J Aust. 190 (S4):S17–S21. 2009. View Article : Google Scholar : PubMed/NCBI | |
Liang W and Chikritzhs T: Early childhood infections and risk of schizophrenia. Psychiatry Res. 200:214–217. 2012. View Article : Google Scholar : PubMed/NCBI | |
Maynard TM, Sikich L, Lieberman JA and LaMantia AS: Neural development, cell-cell signaling, and the ‘two-hit’ hypothesis of schizophrenia. Schizophr Bull. 27:457–476. 2001. View Article : Google Scholar : PubMed/NCBI | |
Debost JCPG, Larsen JT, Munk-Olsen T, Mortensen PB, Meyer U and Petersen L: Joint effects of exposure to prenatal infection and peripubertal psychological trauma in schizophrenia. Schizophr Bull. 43:171–179. 2017. View Article : Google Scholar : PubMed/NCBI | |
Raison CL, Borisov AS, Majer M, Drake DF, Pagnoni G, Woolwine BJ, Vogt GJ, Massung B and Miller AH: Activation of central nervous system inflammatory pathways by interferon-alpha: Relationship to monoamines and depression. Biol Psychiatry. 65:296–303. 2009. View Article : Google Scholar : PubMed/NCBI | |
Rocamonde B, Hasan U, Mathieu C and Dutartre H: Viral-induced neuroinflammation: Different mechanisms converging to similar exacerbated glial responses. Front Neurosci. 17:11082122023. View Article : Google Scholar : PubMed/NCBI | |
Niranjan R, Muthukumaravel S and Jambulingam P: The involvement of neuroinfammation in dengue viral disease: Importance of innate and adaptive immunity. Neuroimmunomodulation. 26:111–118. 2019. View Article : Google Scholar : PubMed/NCBI | |
Bogovic P and Strle F: Tick-borne encephalitis: A review of epidemiology, clinical characteristics, and management. World J Clin Cases. 3:430–441. 2015. View Article : Google Scholar : PubMed/NCBI | |
Lannes N, Neuhaus V, Scolari B, Kharoubi-Hess S, Walch M, Summerfield A and Filgueira L: Interactions of human microglia cells with Japanese encephalitis virus. Virol J. 14:82017. View Article : Google Scholar : PubMed/NCBI | |
Shao Q, Herrlinger S, Yang SL, Lai F, Moore JM, Brindley MA and Chen JF: Zika virus infection disrupts neurovascular development and results in postnatal microcephaly with brain damage. Development. 143:4127–4136. 2016. View Article : Google Scholar : PubMed/NCBI | |
de Sousa JR, Azevedo RSS, Martins Filho AJ, Araujo MTF, Moutinho ERC, Baldez Vasconcelos BC, Cruz ACR, Oliveira CS, Martins LC, Baldez Vasconcelos BH, et al: Correlation between apoptosis and in situ immune response in fatal cases of microcephaly caused by Zika virus. Am J Pathol. 188:2644–2652. 2018. View Article : Google Scholar : PubMed/NCBI | |
Li Y, Zhao L, Luo Z, Zhang Y, Lv L, Zhao J, Sui B, Huang F, Cui M, Fu ZF and Zhou M: Interferon-λ attenuates rabies virus infection by inducing interferon-stimulated genes and alleviating neurological inflammation. Viruses. 12:4052020. View Article : Google Scholar : PubMed/NCBI | |
Marsland AL, Petersen KL, Sathanoori R, Muldoon MF, Neumann SA, Ryan C, Flory JD and Manuck SB: Interleukin-6 covaries inversely with cognitive performance among middle-aged community volunteers. Psychosom Med. 68:895–903. 2006. View Article : Google Scholar : PubMed/NCBI | |
Dickerson SS, Gable SL, Irwin MR, Aziz N and Kemeny ME: Social-evaluative threat and proinflammatory cytokine regulation: An experimental laboratory investigation. Psychol Sci. 20:1237–1244. 2009. View Article : Google Scholar : PubMed/NCBI | |
Metcalf SA, Jones PB, Nordstrom T, Timonen M, Mäki P, Miettunen J, Jääskeläinen E, Järvelin MR, Stochl J, Murray GK, et al: Serum C-reactive protein in adolescence and risk of schizophrenia in adulthood: A prospective birth cohort study. Brain BehavImmun. 59:253–259. 2017. | |
Müller N: Inflammation in schizophrenia: Pathogenetic aspects and therapeutic considerations. Schizophr Bull. 44:973–982. 2018. View Article : Google Scholar : PubMed/NCBI | |
Khandaker GM, Pearson RM, Zammit S, Lewis G and Jones PB: Association of serum interleukin 6 and C-reactive protein in childhood with depression and psychosis in young adult life: A population-based longitudinal study. JAMA Psychiatry. 71:1121–1128. 2014. View Article : Google Scholar : PubMed/NCBI | |
Maxeiner HG, Marion Schneider E, Kurfiss ST, Brettschneider J, Tumani H and Bechter K: Cerebrospinal fluid and serum cytokine profiling to detect immune control of infectious and inflammatory neurological and psychiatric diseases. Cytokine. 69:62–67. 2014. View Article : Google Scholar : PubMed/NCBI | |
Sarma JV and Ward PA: The complement system. Cell Tissue Res. 343:227–235. 2011. View Article : Google Scholar : PubMed/NCBI | |
Keshavan M, Lizano P and Prasad K: The synaptic pruning hypothesis of schizophrenia: Promises and challenges. World Psychiatry. 19:110–111. 2020. View Article : Google Scholar : PubMed/NCBI | |
Druart M and Le Magueresse C: Emerging roles of complement in psychiatric disorders. Front Psychiatry. 10:5732019. View Article : Google Scholar : PubMed/NCBI | |
Mondelli V, Di Forti M, Morgan BP, Murray RM, Pariante CM and Dazzan P: Baseline high levels of complement component 4 predict worse clinical outcome at 1-year follow-up in first-episode psychosis. Brain BehavImmun. 88:913–915. 2020.PubMed/NCBI | |
Tomonaga K: Virus-induced neurobehavioral disorders: Mechanisms and implications. Trends Mol Med. 10:71–77. 2004. View Article : Google Scholar : PubMed/NCBI | |
Kotsiri I, Resta P, Spyrantis A, Panotopoulos C, Chaniotis D, Beloukas A and Magiorkinis E: Viral infections and schizophrenia: A comprehensive review. Viruses. 15:13452023. View Article : Google Scholar : PubMed/NCBI | |
Sayeh A, Cheikh CB, Mrad M, Lakhal N, Gritli N, Galelli S, Oumaya A and Fekih-Mrissa N: Association of HLA-DR/DQ polymorphisms with schizophrenia in Tunisian patients. Ann Saudi Med. 34:503–507. 2014. View Article : Google Scholar : PubMed/NCBI | |
Prasard S, Semwal P, Deshpande S, Bhatia T, Nimgaonkar VL and Thelma BK: Molecular genetics of schizophrenia: Past, present and future. J Biosci. 27 (Suppl 1):S35–S52. 2002. View Article : Google Scholar | |
Wright P, Donaldson PT, Underhill JA, Choudhuri K, Doherty DG and Murray RM: Genetic association of the HLA DRB1 gene locus on chromosome 6p21.3 with schizophrenia. Am J Psychiatry. 153:1530–1533. 1996. View Article : Google Scholar : PubMed/NCBI | |
Nimgaonkar VL, Rudert WA, Zhang X, Trucco M and Ganguli R: Negative association of schizophrenia with HLA DQB1*0602: Evidence from a second African-American cohort. Schizophr Res. 23:81–86. 1997. View Article : Google Scholar : PubMed/NCBI | |
James LM, Charonis SA and Georgopoulos AP: Schizophrenia, human leukocyte antigen (HLA), and herpes viruses: Immunogenetic associations at the population level. Neurosci Insights. 18:263310552311664112023. View Article : Google Scholar : PubMed/NCBI | |
Wongchitrat P, Chanmee T and Govitrapong P: Molecular mechanisms associated with neurodegeneration of neurotropic viral infection. Mol Neurobiol. 61:2881–2903. 2024. View Article : Google Scholar : PubMed/NCBI | |
Li C, Xu D, Ye Q, Hong S, Jiang Y, Liu X, Zhang N, Shi L, Qin CF and Xu Z: Zika virus disrupts neural progenitor development and leads to microcephaly in mice. Cell Stem Cell. 19:120–126. 2016. View Article : Google Scholar : PubMed/NCBI | |
Tiwari SK, Dang JW, Lin N, Qin Y, Wang S and Rana TM: Zika virus depletes neural stem cells and evades selective autophagy by suppressing the Fanconi anemia protein FANCC. EMBO Rep. 21:e491832020. View Article : Google Scholar : PubMed/NCBI | |
Gabriel E, Ramani A, Karow U, Gottardo M, Natarajan K, Gooi LM, Goranci-Buzhala G, Krut O, Peters F, Nikolic M, et al: Recent Zika virus isolates induce premature differentiation of neural progenitors in human brain organoids. Cell Stem Cell. 20:397–406.e5. 2017. View Article : Google Scholar : PubMed/NCBI | |
Crunfi F, Carregari VC, Veras FP, Silva LS, Nogueira MH, Antunes ASLM, Vendramini PH, Valença AGF, Brandão-Teles C, Zuccoli GDS, et al: Morphological, cellular, and molecular basis of brain infection in COVID-19 patients. Proc Natl Acad Sci USA. 119:e22009601192022. View Article : Google Scholar | |
de Oliveira LG, de Souza Angelo Y, Yamamoto P, Carregari VC, Crunfli F, Reis-de-Oliveira G, Costa L, Vendramini PH, Duque ÉA, Dos Santos NB, et al: SARS-CoV-2 infection impacts carbon metabolism and depends on glutamine for replication in Syrian hamster astrocytes. J Neurochem. 163:113–132. 2022. View Article : Google Scholar : PubMed/NCBI | |
Fan Y and He JJ: HIV-1 Tat induces unfolded protein response and endoplasmic reticulum stress in astrocytes and causes neurotoxicity through glial fbrillary acidic protein (GFAP) activation and aggregation. J Biol Chem. 291:22819–22829. 2016. View Article : Google Scholar : PubMed/NCBI | |
Zhou BY, Liu Y, Kim BO, Xiao Y and He JJ: Astrocyte activation and dysfunction and neuron death by HIV-1 Tat expression in astrocytes. Mol Cell Neurosci. 27:296–305. 2004. View Article : Google Scholar : PubMed/NCBI | |
Teodorof-Diedrich C and Spector SA: Human immunodeficiency virus type 1 gp120 and Tat induce mitochondrial fragmentation and incomplete mitophagy in human neurons. J Virol. 92:e00993–18. 2018. View Article : Google Scholar : PubMed/NCBI | |
McGavern DB and Kang SS: Illuminating viral infections in the nervous system. Nat Rev Immunol. 11:318–329. 2011. View Article : Google Scholar : PubMed/NCBI | |
Bhatia MS, Gautam P and Jhanjee A: Psychiatric morbidity in patients with chikungunya Fever: First report from India. J Clin Diagn Res. 9:VC01–VC03. 2015.PubMed/NCBI | |
Figueiredo T, Dias da Costa M and Segenreich D: Manic episode after a chikungunya virus infection in a bipolar patient previously stabilized with valproic acid. J Clin Psychopharmacol. 38:395–397. 2018. View Article : Google Scholar : PubMed/NCBI | |
Samaan Z, McDermid Vaz S, Bawor M, Potter TH, Eskandarian S and Loeb M: Neuropsychological impact of west nile virus infection: An extensive neuropsychiatric assessment of 49 cases in Canada. PLoS One. 11:e01583642016. View Article : Google Scholar : PubMed/NCBI | |
Srivastava R, Kalita J, Khan MY and Misra UK: Free radical generation by neurons in rat model of Japanese encephalitis. Neurochem Res. 34:2141–2146. 2009. View Article : Google Scholar : PubMed/NCBI | |
James HJ, Sharer LR, Zhang Q, Wang HG, Epstein LG, Reed JC and Gelbard HA: Expression of caspase-3 in brains from paediatric patients with HIV-1 encephalitis. Neuropathol Appl Neurobiol. 25:380–386. 1999. View Article : Google Scholar : PubMed/NCBI | |
Du X, Wang H, Xu F, Huang Y, Liu Z and Liu T: Enterovirus 71 induces apoptosis of SH-SY5Y human neuroblastoma cells through stimulation of endogenous microRNA let-7b expression. Mol Med Rep. 12:953–959. 2015. View Article : Google Scholar : PubMed/NCBI | |
Jackson AC and Rossiter JP: Apoptosis plays an important role in experimental rabies virus infection. J Virol. 71:5603–5607. 1997. View Article : Google Scholar : PubMed/NCBI | |
Rutherford M and Jackson AC: Neuronal apoptosis in immunodeficient mice infected with the challenge virus standard strain of rabies virus by intracerebral inoculation. J Neurovirol. 10:409–413. 2004. View Article : Google Scholar : PubMed/NCBI | |
Jackson AC: Apoptosis in experimental rabies in bax-deficient mice. Acta Neuropathol. 98:288–294. 1999. View Article : Google Scholar : PubMed/NCBI | |
Fu ZF and Jackson AC: Neuronal dysfunction and death in rabies virus infection. J Neurovirol. 11:101–106. 2005. View Article : Google Scholar : PubMed/NCBI | |
Kojima D, Park CH, Tsujikawa S, Kohara K, Hatai H, Oyamad T, Noguchi A and Inoue S: Lesions of the central nervous system induced by intracerebral inoculation of BALB/c mice with rabies virus (CVS-11). J Vet Med Sci. 72:1011–1016. 2010. View Article : Google Scholar : PubMed/NCBI | |
Parquet MC, Kumatori A, Hasebe F, Morita K and Igarashi A: West Nile virus-induced bax-dependent apoptosis. FEBS Lett. 500:17–24. 2001. View Article : Google Scholar : PubMed/NCBI | |
Kleinschmidt MC, Michaelis M, Ogbomo H, Doerr HW and Cinatl J Jr: Inhibition of apoptosis prevents West Nile virus induced cell death. BMC Microbiol. 7:492007. View Article : Google Scholar : PubMed/NCBI | |
van Marle G, Antony J, Ostermann H, Dunham C, Hunt T, Halliday W, Maingat F, Urbanowski MD, Hobman T, Peeling J and Power C: West Nile virus-induced neuroinflammation: Glial infection and capsid protein-mediated neurovirulence. J Virol. 81:10933–10949. 2007. View Article : Google Scholar : PubMed/NCBI | |
Yang JS, Ramanathan MP, Muthumani K, Choo AY, Jin SH, Yu QC, Hwang DS, Choo DK, Lee MD, Dang K, et al: Induction of inflammation by West Nile virus capsid through the caspase-9 apoptotic pathway. Emerg Infect Dis. 8:1379–1384. 2002. View Article : Google Scholar : PubMed/NCBI | |
Ramanathan MP, Chambers JA, Pankhong P, Chattergoon M, Attatippaholkun W, Dang K, Shah N and Weiner DB: Host cell killing by the West Nile virus NS2B-NS3 proteolytic complex: NS3 alone is sufficient to recruit caspase-8-based apoptotic pathway. Virology. 345:56–72. 2006. View Article : Google Scholar : PubMed/NCBI | |
Swarup V, Das S, Ghosh S and Basu A: Tumor necrosis factor receptor-1-induced neuronal death by TRADD contributes to the pathogenesis of Japanese encephalitis. J Neurochem. 103:771–783. 2007. View Article : Google Scholar : PubMed/NCBI | |
Mishra MK and Basu A: Minocycline neuroprotects, reduces microglial activation, inhibits caspase 3 induction, and viral replication following Japanese encephalitis. J Neurochem. 105:1582–1595. 2008. View Article : Google Scholar : PubMed/NCBI | |
Chen T, Tu S, Ding L, Jin M, Chen H and Zhou H: The role of autophagy in viral infections. J Biomed Sci. 30:52023. View Article : Google Scholar : PubMed/NCBI | |
Huang SC, Chang CL, Wang PS, Tsai Y and Liu HS: Enterovirus 71-induced autophagy detected in vitro and in vivo promotes viral replication. J Med Virol. 81:1241–1252. 2009. View Article : Google Scholar : PubMed/NCBI | |
Liu ZW, Zhuang ZC, Chen R, Wang XR, Zhang HL, Li SH, Wang ZY and Wen HL: Enterovirus 71 VP1 protein regulates viral replication in SH-SY5Y cells via the mTOR autophagy signaling pathway. Viruses. 12:112019. View Article : Google Scholar : PubMed/NCBI | |
Too IH, Yeo H, Sessions OM, Yan B, Libau EA, Howe JLC, Lim ZQ, Suku-Maran S, Ong WY, Chua KB, et al: Enterovirus 71 infection of motor neuron-like NSC-34 cells undergoes a non-lytic exit pathway. Sci Rep. 6:369832016. View Article : Google Scholar : PubMed/NCBI | |
Fields JA, Metcalf J, Overk C, Adame A, Spencer B, Wrasidlo W, Florio J, Rockenstein E, He JJ and Masliah E: The anticancer drug sunitinib promotes autophagyand protects from neurotoxicity in an HIV-1 Tat model of neurodegeneration. J Neurovirol. 23:290–303. 2017. View Article : Google Scholar : PubMed/NCBI | |
Cheney L, Guzik H, Macaluso FP, Macian F, Cuervo AM and Berman JW: HIV Nef and antiretroviral therapy have an inhibitory effect on autophagy in human astrocytes that may contribute to HIV-associated neurocognitive disorders. Cells. 9:14262020. View Article : Google Scholar : PubMed/NCBI | |
Fields J, Dumaop W, Elueteri S, Campos S, Serger E, Trejo M, Kosberg K, Adame A, Spencer B, Rockenstein E, et al: HIV-1 Tat alters neuronal autophagy by modulating autophagosome fusion to the lysosome: Implications for HIV-associated neurocognitive disorders. J Neurosci. 35:1921–1938. 2015. View Article : Google Scholar : PubMed/NCBI | |
Fischer M, Lindsey N, Staples JE and Hills S; Centers for Disease Control and Prevention (CDC), : Japanese encephalitis vaccines: Recommendations of the advisory committee on immunization practices (ACIP). MMWR Recomm Rep. 59:1–27. 2010. | |
Unni SK, Růžek D, Chhatbar C, Mishra R, Johri MK and Singh SK: Japanese encephalitis virus: From genome to infectome. Microbes Infect. 13:312–321. 2011. View Article : Google Scholar : PubMed/NCBI | |
Hu S, Sheng WS, Schachtele SJ and Lokensgard JR: Reactive oxygen species drive herpes simplex virus (HSV)-1-induced proinflammatory cytokine production by murine microglia. J Neuroinflammation. 8:1232011. View Article : Google Scholar : PubMed/NCBI | |
Kavouras JH, Prandovszky E, Valyi-Nagy K, Kovacs SK, Tiwari V, Kovacs M, Shukla D and Valyi-Nagy T: Herpes simplex virus type 1 infection induces oxidative stress and the release of bioactive lipid peroxidation by-products in mouse P19N neural cell cultures. J Neurovirol. 13:416–425. 2007. View Article : Google Scholar : PubMed/NCBI | |
Gorska AM and Eugenin EA: The glutamate system as a crucial regulator of CNS toxicity and survival of HIV reservoirs. Front Cell Infect Microbiol. 10:2612020. View Article : Google Scholar : PubMed/NCBI | |
Alandijany T, Kammouni W, Roy Chowdhury SK, Fernyhough P and Jackson AC: Mitochondrial dysfunction in rabies virus infection of neurons. J Neurovirol. 19:537–549. 2013. View Article : Google Scholar : PubMed/NCBI | |
Ghosh Roy S, Sadigh B, Datan E, Lockshin RA and Zakeri Z: Regulation of cell survival and death during flavivirus infections. World J Biol Chem. 5:93–105. 2014. View Article : Google Scholar : PubMed/NCBI | |
Mukherjee S, Singh N, Sengupta N, Fatima M, Seth P, Mahadevan A, Shankar SK, Bhattacharyya A and Basu A: Japanese encephalitis virus induces human neural stem/progenitor cell death by elevating GRP78, PHB and hnRNPC through ER stress. Cell Death Dis. 8:e25562017. View Article : Google Scholar : PubMed/NCBI | |
Tan Z, Zhang W, Sun J, Fu Z, Ke X, Zheng C, Zhang Y, Li P, Liu Y, Hu Q, et al: ZIKV infection activates the IRE1-XBP1 and ATF6 pathways of unfolded protein response in neural cells. J Neuroinflammation. 15:2752018. View Article : Google Scholar : PubMed/NCBI | |
Hu DD, Mai JN, He LY, Li PQ, Chen WX, Yan JJ, Zhu WD, Deng L, Wei D, Liu DH, et al: Glucocorticoids prevent enterovirus 71 capsid protein VP1 induced calreticulin surface exposure by alleviating neuronal ER stress. Neurotox Res. 31:204–217. 2017. View Article : Google Scholar : PubMed/NCBI | |
Cheng ML, Weng SF, Kuo CH and Ho HY: Enterovirus 71 induces mitochondrial reactive oxygen species generation that is required for efficient replication. PLoS One. 9:e1132342014. View Article : Google Scholar : PubMed/NCBI | |
Guo L, Xing Y, Pan R, Jiang M, Gong Z, Lin L, Wang J, Xiong G and Dong J: Curcumin protects microglia and primary rat cortical neurons against HIV-1 gp120-mediated inflammation and apoptosis. PLoS One. 8:e705652013. View Article : Google Scholar : PubMed/NCBI | |
Shah A, Kumar S, Simon SD, Singh DP and Kumar A: HIV gp120- and methamphetamine-mediated oxidative stress induces astrocyte apoptosis via cytochrome P450 2E1. Cell Death Dis. 4:e8502013. View Article : Google Scholar : PubMed/NCBI | |
Ivanov AV, Valuev-Elliston VT, Ivanova ON, Kochetkov SN, Starodubova ES, Bartosch B and Isaguliants MG: Oxidative stress during HIV infection: Mechanisms and consequences. Oxid Med Cell Longev. 2016:89103962016. View Article : Google Scholar : PubMed/NCBI | |
Simanjuntak Y, Liang JJ, Lee YL and Lin YL: Japanese encephalitis virus exploits dopamine D2 receptor-phospholipase C to target dopaminergic human neuronal cells. Front Microbiol. 8:6512017. View Article : Google Scholar : PubMed/NCBI | |
Damsgaard J, Hjerrild S, Andersen H and Leutscher PDC: Long-term neuropsychiatric consequences of aseptic meningitis in adult patients. Infect Dis (Lond). 47:357–363. 2015. View Article : Google Scholar : PubMed/NCBI | |
Omland LH, Vestergaard BF and Wandall JH: Herpes simplex virus type 2 infections of the central nervous system: A retrospective study of 49 patients. Scand J Infect Dis. 40:59–62. 2008. View Article : Google Scholar : PubMed/NCBI | |
Persson A, Bergström T, Lindh M, Namvar L and Studahl M: Varicella-zoster virus CNS disease-viral load, clinical manifestations and sequels. J Clin Virol. 46:249–253. 2009. View Article : Google Scholar : PubMed/NCBI | |
Sittinger H, Müller M, Schweizer I and Merkelbach S: Mild cognitive impairment after viral meningitis in adults. J Neurol. 249:554–560. 2002. View Article : Google Scholar : PubMed/NCBI | |
Du C, Li G and Han G: Biosafety and mental health: Virus induced cognitive decline. Biosaf Health. 5:159–167. 2023. View Article : Google Scholar : PubMed/NCBI | |
Hare EH, Price JS and Slater E: Schizophrenia and season of birth. Br J Psychiatry. 120:124–125. 1972. View Article : Google Scholar : PubMed/NCBI | |
Machón RA, Mednick SA and Schulsinger F: The interaction of seasonality, place of birth, genetic risk and subsequent schizophrenia in a high risk sample. Br J Psychiatry. 143:383–388. 1983. View Article : Google Scholar : PubMed/NCBI | |
Pallast EG, Jongbloet PH, Straatman HM and Zielhuis GA: Excess seasonality of births among patients with schizophrenia and seasonal ovopathy. Schizophr Bull. 20:269–276. 1994. View Article : Google Scholar : PubMed/NCBI | |
Susser ES, Brown AS and Gorman JM: Prenatal exposures in schizophrenia. American Psychiatric Association; 1999 | |
Mednick SA, Machon RA, Huttunen MO and Bonett D: Adult schizophrenia following prenatal exposure to an influenza epidemic. Arch Gen Psychiatry. 45:189–192. 1988. View Article : Google Scholar : PubMed/NCBI | |
Brown AS, Begg MD, Gravenstein S, Schaefer CA, Wyatt RJ, Bresnahan M, Babulas VP and Susser ES: Serologic evidence of prenatal influenza in the etiology of schizophrenia. Arch Gen Psychiatry. 61:774–780. 2004. View Article : Google Scholar : PubMed/NCBI | |
Bonkovsky HL, Snow KK, Malet PF, Back-Madruga C, Fontana RJ, Sterling RK, Kulig CC, Di Bisceglie AM, Morgan TR, Dienstag JL, et al: Health-related quality of life in patients with chronic hepatitis C and advanced fibrosis. J Hepatol. 46:420–431. 2007. View Article : Google Scholar : PubMed/NCBI | |
Gragnani L, Cerretelli G, Lorini S, Steidi C, Giovannelli A, Monti M, Petraccia L, Sadalla S, Urraro T, Caini P, et al: Interferon-free therapy in hepatitis C virus mixed cryoglobulinaemia: A prospective, controlled, clinical and quality of live analysis. Aliment Pharmacol Ther. 48:440–450. 2018. View Article : Google Scholar : PubMed/NCBI | |
Torrey EF, Bowler AE and Rawlings R: An influenza epidemic and the seasonality of schizophrenic births. Kurstak: Psychiatry and Biological Factors. Springer; Boston, MA: pp. 109–116. 1991, View Article : Google Scholar | |
Nicoll MP, Proença JT and Efstathiou S: The molecular basis of herpes simplex virus latency. FEMS Microbiol Rev. 36:684–705. 2012. View Article : Google Scholar : PubMed/NCBI | |
Torrey EF, Leweke MF, Schwarz MJ, Mueller N, Bachmann S, Schroeder J, Dickerson F and Yolken RH: Cytomegalovirus and schizophrenia. CNS Drugs. 20:879–885. 2006. View Article : Google Scholar : PubMed/NCBI | |
Prasad KMR, Shirts BH, Yolken RH, Keshavan MS and Nimgaonkar VL: Brain morphological changes associated with exposure to HSV1 in first-episode schizophrenia. Mol Psychiatry. 12:105–113. 12007. View Article : Google Scholar : PubMed/NCBI | |
Burgdorf KS, Trabjerg BB, Pedersen MG, Nissen J, Banasik K, Pedersen OB, Sørensen E, Nielsen KR, Larsen MH, Erikstrup C, et al: Large-scale study of toxoplasma and cytomegalovirus shows an association between infection and serious psychiatric disorders. Brain BehavImmun. 79:152–158. 2019. | |
Dickerson F, Jones-Brando L, Ford G, Genovese G, Stallings C, Origoni A, O'Dushlaine C, Katsafanas E, Sweeney K, Khushalani S and Yolken R: Schizophrenia is associated with an aberrant immune response to epstein-barr virus. Schizophr Bull. 45:1112–1119. 2019. View Article : Google Scholar : PubMed/NCBI | |
Dickerson FB, Boronow JJ, Stallings C, Origoni AE, Ruslanova I and Yolken RH: Association of serum antibodies to herpes simplex virus 1 with cognitive deficits in individuals with schizophrenia. Arch Gen Psychiatry. 60:466–472. 2003. View Article : Google Scholar : PubMed/NCBI | |
Thomas P, Bhatia T, Gauba D, Wood J, Long C, Prasad K, Dickerson FB, Gur RE, Gur RC, Yolken RH, et al: Exposure to herpes simplex virus, type 1 and reduced cognitive function. J Psychiatr Res. 47:1680–1685. 2013. View Article : Google Scholar : PubMed/NCBI | |
Yolken RH, Torrey EF, Lieberman JA, Yang S and Dickerson FB: Serological evidence of exposure to herpes simplex virus type 1 is associated with cognitive deficits in the CATIE schizophrenia sample. Schizophr Res. 128:61–65. 2011. View Article : Google Scholar : PubMed/NCBI | |
Krause D, Matz J, Weidinger E, Wagner J, Wildenauer A, Obermeier M, Riedel M and Müller N: The association of infectious agents and schizophrenia. World J Biol Psychiatry. 11:739–743. 2010. View Article : Google Scholar : PubMed/NCBI | |
Waechter R, Ingraham E, Evans R, Cudjoe N, Krystosik A, Isaac R, Watts A, Noël T, Landon B, Fernandes M, et al: Pre and postnatal exposure to chikungunya virus does not affect child neurodevelopmental outcomes at two years of age. PLoS Negl Trop Dis. 14:e00085462020. View Article : Google Scholar : PubMed/NCBI | |
Mazzaro C, Quartuccio L, Adinolfi LE, Roccatello D, Pozzato G, Nevola R, Tonizzo M, Gitto S, Andreone P and Gattei V: A review on extrahepatic manifestations of chronic hepatitis C virus infection and the impact of direct-acting antiviral therapy. Viruses. 13:22492021. View Article : Google Scholar : PubMed/NCBI | |
Fishman SL, Murray JM, Eng FJ, Walewski JL, Morgello S and Branch AD: Molecular and bioinformatic evidence of hepatitis C virus evolution in brain. J Infect Dis. 197:597–607. 2008. View Article : Google Scholar : PubMed/NCBI | |
Wilkinson J, Radkowski M and Laskus T: Hepatitis C virus neuroinvasion: Identification of infected cells. J Virol. 83:1312–1319. 2009. View Article : Google Scholar : PubMed/NCBI | |
Laskus T, Radkowski M, Bednarska A, Wilkinson J, Adair D, Nowicki M, Nikolopoulou GB, Vargas HE and Rakela J: Detection and analysis of hepatitis C virus sequences in cerebrospinal fluid. J Virol. 76:10064–10068. 2002. View Article : Google Scholar : PubMed/NCBI | |
Min S, Gandal MJ, Kopp RF, Liu C and Chen C: No increased detection of nucleic acids of CNS-related viruses in the brains of patients with schizophrenia, bipolar disorder, and autism spectrum disorder. Schizophr Bull. 49:551–558. 2023. View Article : Google Scholar : PubMed/NCBI | |
Yolken RH, Kinnunen PM, Vapalahti O, Dickerson F, Suvisaari J, Chen O and Sabunciyan S: Studying the virome in psychiatric disease. Schizophr Res. 234:78–86. 2021. View Article : Google Scholar : PubMed/NCBI | |
Kneeland RE and Fatemi SH: Viral infection, inflammation and schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry. 42:35–48. 2013. View Article : Google Scholar : PubMed/NCBI | |
Gu CJ, Borjabad A, Hadas E, Kelschenbach J, Kim BH, Chao W, Arancio O, Suh J, Polsky B, McMillan J, et al: EcoHIV infection of mice establishes latent viral reservoirs in T cells and active viral reservoirs in macrophages that are sufficient for induction of neurocognitive impairment. PLoS Pathog. 14:e10070612018. View Article : Google Scholar : PubMed/NCBI | |
Gruchot J, Herrero F, Weber-Stadlbauer U, Meyer U and Küry P: Interplay between activation of endogenous retroviruses and inflammation as common pathogenic mechanism in neurological and psychiatric disorders. Brain BehavImmun. 107:242–252. 2023. | |
Kamitani W, Ono E, Yoshino S, Kobayashi T, Taharaguchi S, Lee BJ, Yamashita M, Kobayashi T, Okamoto M, Taniyama H, et al: Glial expression of Borna disease virus phosphoprotein induces behavioral and neurological abnormalities in transgenic mice. Proc Natl Acad Sci USA. 100:8969–8974. 2003. View Article : Google Scholar : PubMed/NCBI | |
Li F, Sabunciyan S, Yolken RH, Lee D, Kim S and Karlsson H: Transcription of human endogenous retroviruses in human brain by RNA-seq analysis. PLoS One. 14:e02073532019. View Article : Google Scholar : PubMed/NCBI | |
Karlsson H, Bachmann S, Schröder J, McArthur J, Torrey EF and Yolken RH: Retroviral RNA identified in the cerebrospinal fluids and brains of individuals with schizophrenia. Proc Natl Acad Sci USA. 98:4634–4639. 2001. View Article : Google Scholar : PubMed/NCBI | |
Perron H, Hamdani N, Faucard R, Lajnef M, Jamain S, Daban-Huard C, Sarrazin S, LeGuen E, Houenou J, Delavest M, et al: Molecular characteristics of human endogenous retrovirus type-W in schizophrenia and bipolar disorder. Transl Psychiatry. 2:e2012012. View Article : Google Scholar : PubMed/NCBI | |
Tamouza R, Meyer U, Foiselle M, Richard JR, Wu CL, Boukouaci W, Le Corvoisier P, Barrau C, Lucas A, Perron H and Leboyer M: Identification of inflammatory subgroups of schizophrenia and bipolar disorder patients with HERV-W ENV antigenemia by unsupervised cluster analysis. Transl Psychiatry. 11:3772021. View Article : Google Scholar : PubMed/NCBI | |
Slokar G and Hasler G: Human endogenous retroviruses as pathogenic factors in the development of schizophrenia. Front Psychiatry. 6:1832016. View Article : Google Scholar : PubMed/NCBI | |
Jesuthasan A, Massey F, Manji H, Zandi MS and Wiethoff S: Emerging potential mechanisms and predispositions to the neurological manifestations of COVID-19. J Neurol Sci. 428:1176082021. View Article : Google Scholar : PubMed/NCBI | |
Guedj E, Campion JY, Dudouet P, Kaphan E, Bregeon F, Tissot-Dupont H, Guis S, Barthelemy F, Habert P, Ceccaldi M, et al: 18F-FDG brain PET hypometabolism in patients with long COVID. Eur J Nucl Med Mol Imaging. 48:2823–2833. 2021. View Article : Google Scholar : PubMed/NCBI | |
Tromans S, Kinney M, Chester V, Alexander R, Roy A, Sander JW, Dudson H and Shankar R: Priority concerns for people with intellectual and developmental disabilities during the COVID-19 pandemic. BJPsych Open. 6:e1282020. View Article : Google Scholar : PubMed/NCBI | |
Holmes EA, O'Connor RC, Perry VH, Tracey I, Wessely S, Arsenault L, Ballard C, Christensen H, Cohen Silver R, Everall I, et al: Multidisciplinary research priorities for the COVID-19 pandemic: A call for action for mental health science. Lancet Psychiatry. 7:547–560. 2020. View Article : Google Scholar : PubMed/NCBI | |
Ho CS, Chee CY and Ho RC: Mental health strategies to combat the psychological impact of coronavirus disease 2019 (COVID-19) beyond paranoia and panic. Ann Acad Med Singap. 49:155–160. 2020. View Article : Google Scholar : PubMed/NCBI | |
Tsamakis K, Tsiptsios D, Ouranidis A, Mueller C, Schizas D, Terniotis C, Nikolakakis N, Tyros G, Kympouropoulos S, Lazaris A, et al: COVID-19 and its consequences on mental health (Review). Exp Ther Med. 21:2442021. View Article : Google Scholar : PubMed/NCBI | |
Efstathiou V, Stefanou MI, Demetriou M, Siafakas N, Makris M, Tsivgoulis G, Zoumpourlis V, Kympouropoulos SP, Tsoporis JN, Spandidos DA, et al: Long COVID and neuropsychiatric manifestations (Review). Exp Ther Med. 23:3632022. View Article : Google Scholar : PubMed/NCBI | |
Efstathiou V, Stefanou MI, Demetriou M, Siafakas N, Katsantoni E, Makris M, Tsivgoulis G, Zoumpourlis V, Kympouropoulos SP, Tsoporis JN, et al: New-onset neuropsychiatric sequelae and ‘long-COVID’ syndrome (Review). Exp Ther Med. 24:7052022. View Article : Google Scholar : PubMed/NCBI | |
Yuan K, Gong YM, Liu L, Sun YK, Tian SS, Wang YJ, Zhong Y, Zhang AY, Su SZ, Liu XX, et al: Prevalence of posttraumatic stress disorder after infectious disease pandemics in the twenty-first century, including COVID-19: A meta-analysis and systematic review. Mol Psychiatry. 26:4982–4998. 2021. View Article : Google Scholar : PubMed/NCBI | |
Antonelli G and Cutler S: Evolution of the Koch postulates: Towards a 21st-century understanding of microbial infection. Clin Microbiol Infect. 22:583–584. 2016. View Article : Google Scholar : PubMed/NCBI | |
Taquet M, Geddes JR, Husain M, Luciano S and Harrison PJ: 6-Month neurological and psychiatric outcomes in 236 379 survivors of COVID-19: A retrospective cohort study using electronic health records. Lancet Psychiatry. 8:416–427. 2021. View Article : Google Scholar : PubMed/NCBI | |
Schou TM, Joca S, Wegener G and Bay-Richter C: Psychiatric and neuropsychiatric sequelae of COVID-19-A systematic review. Brain Behav Immun. 97:328–348. 2021. View Article : Google Scholar : PubMed/NCBI | |
Shanbehzadeh S, Tavahomi M, Zanjari N, Ebrahimi-Takamjani I and Amiri-Arimi S: Physical and mental health complications post-COVID-19: Scoping review. J Psychosom Res. 147:1105252021. View Article : Google Scholar : PubMed/NCBI | |
Chen C, Wang J, Pan D, Wang X, Xu Y, Yan J, Wang L, Yang X, Yang M and Liu GP: Applications of multi-omics analysis in human diseases. MedComm (2020). 4:e3152023. View Article : Google Scholar : PubMed/NCBI | |
Ong K, Ng K, Ng C, Tan S, Teo W, Karim N, Kumar S and Wong KT: Neuronal infection is a major pathogenetic mechanism and cause of fatalities in human acute nipah virus encephalitis. Neuropathol Appl Neurobiol. 48:e128282022. View Article : Google Scholar : PubMed/NCBI | |
Mathieu C, Bovier FT, Ferren M, Lieberman NAP, Predella C, Lalande A, Peddu V, Lin MJ, Addetia A, Patel A, et al: Molecular features of the measles virus viral fusion complex that favor infection and spread in the brain. mBio. 12:e00799212021. View Article : Google Scholar : PubMed/NCBI |