3,107
views
2
recommends
+1 Recommend
1 collections
    4
    shares

      Acta Materia Medica now indexed by SCOPUS from May 2024. Interested in becoming an AMM published author?

      • Platinum Open Access with no APCs.
      • Fast peer review/Fast publication online after article acceptance.

      Check out the call for papers on our website https://amm-journal.org/index.php/2023/04/26/acta-materia-medica-call-for-papers-2/

      scite_
       
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Latent herpes simplex virus type 1 reactivation increases the susceptibility of neurodegenerative diseases: intervention with traditional Chinese medicine

      Published
      review-article
      Bookmark

            Abstract

            Owing to environmental and lifestyle changes in modern society, physical and mental stress is on the rise among the population in attempting to compensate for the changes. Stress, defined as a series of non-specific responses to abnormal internal and external stimuli, has long been recognized to increase susceptibility to various diseases, including neurodegenerative diseases (NDs). Herpes simplex virus type 1 (HSV-1), a neurotropic herpes virus, establishes latent infections in the trigeminal ganglia for the lifetime of the host. Upon reactivation, the latent HSV-1 enters the lytic cycle, which not only causes a substantial production of reactive oxygen species (ROS) in the neurons, but also induces glial cells to produce inflammatory mediators, eventually resulting in neuronal damage and occurrence of NDs. Remarkably, both physical and psychological stress are common stimuli leading to latent HSV-1 reactivation. Acyclovir is a classic antiviral drug for the treatment of HSV-1 infection and recurrence, but acyclovir cannot reverse neuronal damage caused by HSV-1 reactivation. Recent studies have shown that traditional Chinese medicine (TCM) has advantages in the treatment of NDs. In addition, TCM is thought to prevent latent HSV-1 reactivation and NDs, which may involve regulation of the neuro-endocrine-immune network, treatment of emotional disorders, and modulation of oxidative stress. Understanding the mechanism underlying stress-induced reactivation of latent HSV-1 and the potential consequences of NDs is needed. TCM may be a promising alternative therapy for related diseases.

            Main article text

            1. INTRODUCTION

            With the quickened pace of modern life, people are faced with environmental and lifestyle changes, social factors, emotional fluctuation, and other stimuli that cause physical and mental stress. Stress causes excitation of the sympathetic-adrenal-medullary (SAM) system and hypothalamic-pituitary-adrenal (HPA) axis, and is accompanied by changes in tissue and organ function [1]. The neuroendocrine system is an important endogenous regulator of immunity. Appropriate stress is evolutionarily beneficial to life, while excessive stress has adverse effects on immunity that may accelerate the progression of neurodegenerative diseases (NDs), such as Alzheimer’s disease (AD), Parkinson’s disease (PD), Huntington’s disease (HD), and amyotrophic lateral sclerosis (ALS). Currently, approximately 50 million people worldwide have NDs and the prevalence is expected to be 131 million by 2050 [2], which poses a heavy medical and public health burden worldwide; however, the mechanisms underlying NDs remain elusive.

            Herpes simplex virus type 1 (HSV-1) is an enveloped double-stranded DNA virus that belongs to the Herpesviridae family. The clinical manifestations of HSV-1 include ocular infections and keratitis [3], skin, oral, and genital infections [4], and severe central nervous system (CNS) infections [4, 5]. A comprehensive epidemiologic survey of HSV-1 in Asia revealed that 50% of children and 75% of adults are infected. In addition, seroprevalence increases with age and most infections are acquired in childhood [6]. HSV-1 infection consists of four phases: acute infection; establishment of latency; maintenance of latency; and reactivation. During acute infection, HSV-1 enters epithelial cells of the peripheral mucosal tissue, then replicates in a strictly cascade manner, which ultimately leads to lytic infections. HSV-1 is a common neurotropic virus, and in addition to acute infection, HSV-1 establishes latent infections. During primary infections, the immediate early (IE), early (E), and late (L) genes are sequentially expressed to support sufficient replication in epithelial cells [7]. Following primary infection, HSV-1 from epithelial cells travels to the corresponding sensory ganglion (usually the trigeminal ganglion) by retrograde axonal transport, wherein a life-long latency is established [8]. Under a range of stimuli, including restraint stress, ultraviolet ray (UV) exposure, physical trauma, hormone imbalance, immunosuppression, or hyperthermic stress, latent HSV-1 can be reactivated and undergo productive replication. Notably, given the high infection rate in the general population, neurotropism, and lifelong persistence in neuronal cells, HSV-1 has gradually become an important factor in susceptibility to NDs.

            Currently, the main drugs clinically used to treat HSV-1 infection are nucleoside antiviral analogs, such as acyclovir, which prevent the synthesis of viral DNA by inhibiting DNA polymerase [9]. Although the early efficacy of acyclovir is clear, acyclovir cannot remove the latent virus in the trigeminal ganglion or prevent the recurrence of HSV-1. Traditional Chinese medicine (TCM) has unique advantages in treating HSV-1 because of its comprehensive pharmacologic effects of multi-channel and multi-target regulation. Among the different hypotheses on the mechanism of NDs put forward in the past few years, viral infection has been suspected to be associated with pathogenesis in the progression of NDs. TCM may be used to suppress latent HSV-1 reactivation, and thus prevent and treat NDs, which may involve regulating the neuro-endocrine-immune network, treating emotional disorders, and relieving oxidative stress. This paper reviews the possible mechanisms underlying latent HSV-1 reactivation under stress and the potential effects on NDs and proposes TCM as an alternative therapy for related diseases.

            2. ESSENTIAL FACTORS INVOLVED IN THE ESTABLISHMENT, MAINTENANCE, AND REACTIVATION CYCLE OF LATENT HSV-1

            At present, although no natural animal host of HSV-1 has been identified, mice and rabbits are still used to study HSV-1 latent transcription, epigenetic modification, and latent reactivation [10]. After these animals are infected with HSV-1, the virus first replicates in the peripheral epithelial cells, then travels in a retrograde fashion along the axon to the sensory ganglia, after which a latent infection is established. During latency, a group of viral non-coding RNAs is expressed, known as latency-related transcripts (LATs). Although LATs promote the establishment of latency [11], LATs may not be necessary to establish latency [12]. Yu et al. [13] reported that HSV-1 can be reactivated from latency under stress conditions. HSV-1 reactivation and replication is accompanied by a decrease in LATs. Impaired IE gene expression is another prominent cause that favors the establishment of HSV-1 latency [14]. Viral protein 16 (VP16) is an essential viral envelope protein for virion morphogenesis [15]. The neuronal latency of HSV-1 may be related to the regulation of VP16 function, which may be due to its lack of or transformation from an activator to a repressor by changing the interaction of neuronal cofactors [16, 17]. The HSV-1 gene transcription cascade is initiated by VP16, which is expressed in the late stage of the virus replication cycle [18]. VP16 can promote the transcription of IE mRNAs by binding to the octamer-binding protein 1 (Oct1) in epithelial cells and host cell factor-1 (HCF-1) in epithelial and neuron cells [19, 20]. When HSV-1 is retrogradely transferred to the nucleus along the axon, VP16 enters the nucleus and combines with the Pit-Oct-Unc (POU) transcription factor family to form a triplet, which initiates the expression of IE genes [21]. Due to the absence of VP16/HCF1/Oct1 triplet, ICP0 expression is reduced, and neurons block expression of the E and L genes via the HDAC/LSD1/REST/CoREST host silencing complex, further promoting the establishment of latency.

            Although many scientists are trying to unveil the mechanism underlying HSV-1 latency establishment and maintenance, no hypothesis can fully explain these life cycle events. The balance between HSV-1 lytic and latent infections is regulated by various factors, including HSV-1 encoded latency-associated transcript (LAT) [22], microRNAs [23], nerve growth factor (NGF) [1], epigenetic regulation [24], and immune response [25]. It is well-established that LAT expression is important for the maintenance of HSV-1 latency infections [26], which is related to antisense mechanisms [23], anti-apoptotic effects [27], and possible regulation of functioning protein translation [28]. Notably, in addition to LAT, other factors from the virus and the host are involved in the dynamic process of latency and reactivation of HSV-1. For example, NGF, a nerve cell growth regulator with dual biological functions (neuron nutrition and neurite outgrowth promotion), has a crucial role in the maintenance of HSV-1 latency [29]. Abundant evidence indicates that gene expression of HSV-1 is epigenetically-regulated. The changes in histone modification status control the switch between latency and reactivation. During latency, the viral genome binds to histones in the nucleus, and lytic genes are in a transcriptionally-repressed heterochromatin state. In the study of Neumann et al., the intraperitoneal administration of sodium butyrate, an inhibitor of histone deacetylase, induced HSV-1 reactivation with a frequency of 75%-100% [30]. By comparison, specific inhibitors of H3K27me3 demethylation prevented reactivation of HSV-1 in latently-infected neurons [31]. These studies indicated that the balance between latency and reactivation in the HSV-1 life cycle is exquisitely regulated by various factors.

            3. ROLE OF STRESS-INDUCED ENDOCRINE ALTERATIONS AND OTHER STIMULI IN HSV-1 REACTIVATION

            According to the organism response, stress is divided into physiologic, psychological, and comprehensive stress ( Table 1 ). Restraint stress in animal models has been shown to produce an inevitable physical and mental stress for induction of latent HSV-1 reactivation [45]. Our previous studies have shown that restraint stress leads to significant oxidative damage of central neurons, disruption of the endocrine system, and dysfunction of immunity, ultimately resulting in susceptibility of the host to viral infectious diseases and tumors [34, 46, 47]. In the case of HSV-1 infection, Ortiz et al. studied the modulatory effects of restraint stress on the clinical pathophysiology of primary HSV-1 infection in the SKH-1 mouse. The results showed that restraint stress prolonged viral replication during primary infection [48]. The application of restraint stress to induce HSV-1 reactivation from latency provides an experimental model to explore the complex relationships between stress and latent HSV-1 reactivation [49].

            Table 1 |

            Classification of stress animal models.

            Stress typeBuild methodApplicationPathophysiologic features
            Physiologic stress model Cold Stress Model [32]Put animals at low temperature (4 °C) environmentEstablish acute and chronic stress models in a short timeIncrease the concentration of CORT in the serum of rats
            Heat Stress Model [33]Place the animal in a high temperature (39 °C) environmentResearch on depression and cognitive abilityStimulate the central nerve axis to produce a series of pathophysiologic responses
            Restraint Stress Model [34, 35]Fix limbs and headInvestigate the mechanism of stress damageInduce vascular oxidative stress
            Fatigue Stress Model [36]Force running and swimmingResearch on drug evaluation and mechanismCause the organism’s immune system to decline
            Plantar Electric Shock Stress Model [37]Shock plantar repeatedlyGenerate a high-intensity stress responseInduce conditional fear memory
            Psychological stress model Post-traumatic Stress Disorder Model [38]Give natural enemies stimulusResearch on clinical mental disordersDamage hippocampal function
            Light Stress Model [39]Change the light time to build a stressAssess the effects of chronic stress on animalsCause cognitive impairment
            Natural Enemy Stress Model [40]Encounter natural predators or change odorInduce acute stress responseCause a rapid rise in serum adrenal hormone levels
            Restraint Stress Model [34, 35]Fix limbs and headInvestigate the mechanism of stress damageInduce vascular oxidative stress
            Mother-infant Separation Stress Model [41]Isolate newborn mice from the motherAssess cognitive and memory skillsCause mental and behavioral disorders
            Noise Stress Model [42]Exposure of animals to intense noiseStudy diseases in susceptible peopleAffect brain tissue ability to scavenge free radicals and change amino acid levels
            Repeated Survival Crisis Stress Model [43]Place adventive male rats in cages of “resident” male ratsProvide a more natural stress modelProduce lasting behavioral and neurochemical changes
            Comprehensive stress model Combined Physiologic and Psychological Stress Model [44]Work together of multiple stressesFit the social environment comprehensivelyDecrease 5-HT content in the hypothalamus

            In response to psychological stress, latent HSV-1 is reactivated from the sensory neuron, then enters the lytic cycle, subsequently causing release of the infectious virus. Because numerous hormones are involved in the stress response, it is reasonable to embrace the notion that reactivation of HSV-1 may be regulated by hormones, such as glucocorticoids (GCs) and thyroid hormones (THs) [1]. Under psychological stress, the HPA axis is activated and the concentration of GCs is rapidly increased in the blood [50]. Corticosterone (CORT) is a main type of GCs in rodents and is equivalent to cortisol in humans. CORT can establish a “long-term” stress response by binding to the glucocorticoid receptor (GR) or mineralocorticoid receptor (MR) to regulate metabolism and suppress the immune system [51]. Importantly, the elevated GCs can provoke HSV-1 replication and drive reactivation from latency.

            3.1 Stress-induced elevation of GCs

            Glial cells (e.g., microglia and astrocytes) have a key role in maintenance of the neuroinflammatory profile in the brain. Microglia are resident myeloid cells of the CNS that have an important role in immune surveillance [52]. Astrocytes also contribute to the immune response by releasing cytokines that influence activation of microglia and repair of neurons [53]. Overabundance of GRs in glia suggests distinct roles in inflammatory modulation. We previously reported that the stress hormone, CORT, enhances HSV-1 susceptibility in SH-SY5Y cells and a mouse model, which are largely dependent on GR. Our results further demonstrated that CORT suppresses the HSV-1-induced immune response by activating GRs, which inhibit phosphorylation of interferon regulatory factor 3 (IRF3) and expression of IFN-β, indicating the inhibitory effect of GR on anti-viral immunity [1]. There is still a lack of Reports involving the GR distribution in dopaminergic neurons or other types of neurons, however, are limited. Thus, further studies on the stress response and related diseases are warranted. The second factor involved in GR availability is transcription activity. The phosphorylation of GR has been proposed to contribute to GC resistance in multiple disease models [54]. The state of GC resistance, which may result from chronic stress [55], coincides with a decreased level of brain-derived neurotrophic factor (BDNF) in the cortex and hippocampus, and an increased level of BDNF in amygdala and dopaminergic neurons [56]. Although a majority of GR phosphorylation is glucocorticoid-dependent [57], some investigations have indicated that GR phosphorylation can be glucocorticoid-independent. For example, the BDNF-tyrosine receptor kinase B (TrkB) pathway has been reported to be involved in phosphorylation of human GRs [58, 59].

            The role of GCs in HSV-1 reactivation has been extensively studied. Specifically, it has been shown that GR and Krüppel-like transcription factor 15 (KLF15) cooperatively transactivate the promoter of infected cell protein 0 (ICP0) when cells are treated with the synthetic corticosteroid, dexamethasone [60]. Additionally, Treg cells are involved in GC-induced HSV-1 reactivation. Yu et al. proposed that GCs lead to HSV-1 reactivation via increased Treg cell control of cell cluster differentiation 8+ T cells (CD8+ T cells), thus promoting viral replication under diminished immune surveillance [13]. When the HSV-1 lytic genes are expressed at a low level, CD8+ T cells secrete interferon-γ (IFN-γ) to inhibit the expression of ICP0 and prevent reactivation of the virus. Nevertheless, this process does not occur in all latently-infected neurons [1]. Moreover, intrinsic immunity is closely related to the GCs. For example, stress-induced autophagy promotes degradation of nuclear intrinsic defense components, such as promyelocytic leukemia (PML), thus mediating intrinsic immunity [61]. Promyelocytic leukemia nuclear bodies (PML-NBs) are multiprotein complexes. There is growing evidence that PML-NBs play an important role in intrinsic cellular defense against many DNA viruses, including HSV-1. Due to alternative splicing, PML has seven isoforms (I-VII), and fully functional PML-NBs requires collaboration of different PML isoforms [62]. The viral protein, ICP0, localizes to PML-NBs in HSV-1 infections, and induces proteasome degradation of PML [63]. The absence of all PML isoforms decreases cellular resistance to ICP0-null mutant HSV-1 [64]. The interaction causes the auto-lysosome degradation of PML, resulting in enhanced HSV-1 replication in neuron cells [65]. This finding is consistent with previous work in our laboratory, in which restraint stress/CORT was shown to increase HSV-1 infection by promoting the interaction of PML with autophagy-related proteins (sequestosome-1 and microtubule-associated proteins 1A/1B light chain 3B). This study provided direct evidence linking intrinsic immunity and stress-induced virus reactivation.

            3.2 Stress-induced disturbance of THs

            In addition to GCs, another type of endocrine hormone, TH, also has a vital role in regulating HSV-1 reactivation. Generally, TH exerts an effect via thyroid hormone receptors (TRs). The mechanism underlying HSV-1 reactivation induced by TH is distinct from that of GCs. It has been shown that TH suppresses HSV-1 replication through histone modification in differentiated neuroendocrine cells. Specifically, TH activates the transcription of LAT via TR, inhibits ICP0 expression, and recruits the repressive histones, H3K9me3 and H3K9me2, to the HSV-1 thymidine kinase promoter, thus inhibiting viral reactivation [66]. Under stimuli, such as hyperthermia and surgical trauma, TH secretions are substantially reduced and cannot effectively silence the HSV-1 gene, thus causing HSV-1 reactivation [67]. In addition, some putative TH-responsive elements (TREs) are located within HSV-1 genes, including thymidine kinase (TK) and LAT [67]. Liganded TR represses the transcription of TK in neurons to inhibit viral gene expression and promote the establishment of latency, while unliganded TR interacts with TK TREs to increase viral replication. Taken together, these results suggest that TH-mediated regulation participates in HSV-1 reactivation control.

            3.3 Other stimuli

            Other than endocrine alterations, UV exposure and physical trauma have been reported to be stimuli for HSV-1 latency reactivation. Rooney et al. evaluated the effect of UV light on recurrent herpes labialis in 38 patients in a double-blind, placebo-controlled crossover trial. Interestingly, following UV irradiation, herpes labialis occurred in 27 of 38 patients (71%) and the mean time to recurrence was 2.9 days. In contrast, when protected against UV using sunscreen, only 1 of 35 patients developed virus at the site of exposure [68]. Similarly, Ludema et al. proposed that UV exposure is associated with HSV recurrence. Spending ≥ 8 hours per week outdoors under high UV exposure increases the risk of ocular HSV recurrence [69].

            Overall, reactivation of HSV-1 is a complex process. As a result of the host and viral interaction, it depends not only on the immune status of the host, but also on the characteristics of the virus. Therefore, the identification of regulatory factors involved in HSV-1 latency and reactivation is important for a better understanding of the underlying pathologic mechanism and clinical guidance for the treatment of related diseases. The roles of stress-induced endocrine alterations and other stimuli in HSV-1 reactivation are summarized in Figure 1 .

            Figure 1 |

            Role of stress-induced endocrine alterations and other stimuli in HSV-1 reactivation.

            An important regulator of stress-induced HSV-1 reactivation is hormone levels, such as GCs and TH. Generally, restraint stress stimulates the HPA axis to increase the corticotropin-releasing hormone (CRH) released from the hypothalamus, which subsequently enhances the adrenocorticotrophic hormone (ACTH) release from the pituitary. With the increase in ACTH levels, the GCs secreted by the adrenal glands are augmented considerably. The mounting levels of GCs disrupt the host factors that are necessary for maintaining viral latency, ultimately leading to viral gene transcription and HSV-1 reactivation. The host factors impacted by GCs can be summarized as follows: 1). GCs inhibit IFN-γ secretion from CD8+ T cells in response to latently-infected neurons [1]. 2). GCs led to HSV-1 reactivation via increased Treg cell control of CD8+ T cells, thus promoting viral replication under diminished immune surveillance [13]. Other than endocrine alterations, UV exposure and physical trauma are also stimuli for reactivation of HSV-1.

            4. REACTIVATION OF HSV-1 PROVOKES NEUROINFLAMMATION

            Neuro-inflammation is believed to have a key role in the progression of NDs, such as AD and PD [70]. Glial cells function to support and nourish neurons cells, mainly astrocytes, microglia, and oligodendrocytes [71]. Numerous studies have shown that neuroinflammation caused by HSV-1 infection is involved in the pathogenesis of NDs [5, 72, 73]. Moreover, chronic HSV-1 infection leads to the continuous activation of microglia and triggers the production of neurotoxic and neuroprotective factors [74]. Both stress and exogenous CORT contribute to the activation and proliferation of microglia [7577]. Once activated, microglia provide the first line of immune defense by releasing immune mediators that orchestrate innate and acquired immune responses within the CNS, including the release of pro-inflammatory cytokines [78, 79]. A study showed that activated astrocytes produce two phenotypes (A1- and A2-type) under different stimuli. A1-type astrocytes cause substantial neurotoxicity induced by three inflammatory factors (interleukin-1α [IL-1α], tumor necrosis factor [TNF], and complement component 1, q subcomponent [C1q]) secreted from microglia [80]. In contrast, A2-type astrocytes secrete various neuroprotective substances, such as glial cell-derived neurotrophic factor (GDNF), BDNF, cerebral dopamine neurotrophic factor (CDNF), and an anti-inflammatory factor (tumor growth factor-beta [TGF-β]), to maintain neuron survival and repair tissue damage during cerebral ischemia [80, 81]. Depending on the secreted cytokines, activated microglia are also classified into M1 and M2 types [82]. M1 microglia mainly secrete pro-inflammatory factors such, as tumor necrosis factor-alpha (TNF-α), interleukin-1 beta (IL-1β), nitric oxide (NO), superoxide, and fatty acid metabolites [83]. In contrast, M2 microglia mainly secrete some anti-inflammatory factors, such as interleukin 10 (IL-10). These anti-inflammatory factors are inhibit the activation of M1 microglia, and promote the repair of tissues and remodeling of the extracellular matrix [84]. Exposure to stress or HSV-1 infection can disrupt the balance between pro- and anti-inflammatory responses, resulting in inflammatory mediator dominance [78, 79]. Therefore, how to maintain homeostasis between microglia and astrocytes provides new research opportunities for the treatment of neuro-inflammation.

            The host recognizes HSV-1 infection through a series of germline gene-encoded pathogenic pattern receptors, such as toll-like receptors (TLRs), then produce type I interferon (IFN-I), which induces the interferon-stimulated gene (ISG) to exert an antiviral effect with the release of pro-inflammatory cytokines. A study demonstrated that HSV-1-infected microglia confer a STING-dependent antiviral state to neurons and prime IFN-I production in astrocytes via the TLR3 pathway [85]. In contrast, TLR3 is necessary to induce innate immune responses against HSV-1 in neurons and astrocytes, but not in microglia [86]. It has also been reported that Aβ-protein aggregation mimics the damage-related molecular pattern (DAMP) and promotes inflammation in microglia by activating TLR4 [87]. Although this reaction is responsible for the clearance of Aβ, the persistent pro-inflammatory condition may be harmful to the host [88, 89]. HSV-1 induces an intracranial inflammatory response through TLR2 in mice, while TLR2 knockout significantly improves the survival rate in mice [90]. It has been reported that TLR3 recognizes dsRNA [91] and induces activation of the nuclear factor-kappa B (NF-κB) signaling pathway and the subsequent release of inflammatory cytokines. In addition, inflammatory factors provoke astrocytes to produce IFN-I [92], which mediates antiviral innate immunity to fight against HSV-1 infection in the CNS.

            Furthermore, studies have reported that cytokines, including IFN-γ, IL-17, and IL-6, are elevated in multiple sclerosis lesions, and C-C chemokines, including CCL2, CCL3, CCL4, CCL5, CCL7, and CXCL12, are also increased [93, 94]. Glial-derived IL-1 and IL-6 have been shown to be critical inflammatory cytokines in the brain of patients with AD [95]. In addition, cytokines, including TNF-α, TGF-β1, IL-1β, IL-2, and INF-γ, as well as reactive oxygen species (ROS)/NO are increased in the brains of patients with PD [96]. Compared with healthy individuals, the plasma levels of IL-6, IL-8, IL-4, IL-10, TNF-α, and IL-5 are significantly elevated in patients with HD. Similarly, the levels of IL-6 and IL-8 in the cerebrospinal fluid and striatum are also elevated [97]. In addition, astrocytes from patients with familial and sporadic amyotrophic lateral sclerosis are toxic to motor neurons. The toxicity is related to inflammatory molecules produced by astrocytes, including several C-C and C-X-C chemokines, TNF, and IL-8 [98, 99]. Therefore, we speculate that the expression of inflammatory factors induced by HSV-1 and immune activation state in the brain may account for increased ND susceptibility.

            5. REACTIVATION OF HSV-1 INDUCES OXIDATIVE DAMAGE TO NEURONS

            It has been suggested that inflammation triggered by HSV-1 reactivation may be an important cause of NDs; however, the incidence of NDs is not mitigated by suppressing neuroinflammation. Hence, there may be other mechanisms for increasing ND susceptibility. Notably, because the underlying cause of NDs is the degenerative death of neurons, it is worth determining whether HSV-1 reactivation directly damages neurons.

            Oxidative stress plays a significant role in the pathogenesis of NDs because dopaminergic neurons are regularly subjected to oxidative stress, which triggers the death of neurons. Patients with PD have been shown to have reduced antioxidant capacity and lower glutathione levels [100]. Additionally, depletion of anti-oxidative substances in neurons is also related to HSV-1 reactivation. Interactions between HSV-1 and oxidative stress promote neurodegenerative processes that occur in AD. De et al. reported that antioxidant compounds prevent HSV-1 from inducing the formation of Aβ oligomers in SH-SY5Y cells, therefore suggesting that oxidative stress contributes to the pathogenesis of AD [101]. ROS induced by viral infection may defeat antioxidant defenses and cause oxidative damage to host tissues [102].

            Neurons in the CNS are particularly vulnerable to oxidative stress damage compared to other organs. Exposure to oxidative stress increases the risk of neuronal damage in NDs manifested by neuronal function deficiency and neuron loss. Wang et al. showed that carbon disulfide exposure causes CNS toxicity manifested by learning and memory function impairment, which might be attributed to the extensive oxidative stress-induced apoptosis of hippocampal neurons [103]. Long-term oxidative stress in the CNS causes irreversible oxidative damage to neuron macromolecules, which leads to neuronal cell death. In fact, apoptosis, autophagy, and ferroptosis are the main pathways mediating oxidative stress-induced neuronal cell loss.

            Apoptosis is activated in response to external or internal stimuli [104]. In fact, induction of host cell apoptosis is one of the consequences of HSV-1 infection, and HSV-1 induces apoptosis-related pathways at multiple stages after neuronal infection [105]. HSV-1-infected microglia produce high levels of ROS through stimulation of microglia TLR2 and the subsequent redox imbalance leads to neuronal damage, which is characterized by lipid peroxidation in murine mixed microglia neuronal cultures [106]. ROS may cause the peroxidation of cardiolipin, a mitochondrial inner membrane-specific phospholipid, and subsequent products formed by lipid peroxidation may activate intrinsic apoptosis [107]. LATs promote the survival of neuronal cells by inhibiting cell apoptosis. The anti-apoptotic function of LATs is mediated in many ways. For example, the anti-apoptotic function of LATs protects cells from cold shock-induced apoptosis by inhibiting the dephosphorylation of protein kinase B [108]. Alternatively, LATs inhibit granzyme B-induced cleavage of caspase-3 and neuronal apoptosis. In addition, LATs also prevent infected neurons from being killed by CD8+ T cells [109].

            As a conserved physiologic process, autophagy has also been linked to the anti-HSV-1 effect of host. For example, autophagy removes old or damaged organelles to the cell during starvation and eliminates aggregated proteins caused by misfolding disorders to maintain cell homeostasis [110]. Under normal circumstances, ROS-induced autophagy reduces the damage caused by oxidative stress [111]; however, excessive autophagy caused by ROS leads to autophagic cell death [112]. Autophagy has a dual role in the HSV-1 infectious process. HSV-1 causes CNS system damage via autophagic regulation, which enhances neurodegeneration [113].

            In addition to apoptosis and autophagy, ferroptosis, a novel iron-dependent cell death process, also participates in oxidative stress-induced neuron damage [114]. Ferroptosis is characterized by the cellular accumulation of lipid-ROS, which ultimately leads to cell death [115]. Vigorous dopamine (DA) metabolism in midbrain dopaminergic neurons is more likely to cause the release of oxidized free radicals [116]. Neurons containing various polyunsaturated fatty acids (PUFAs) are susceptible to attack by oxygen-free radicals. In addition, excessive activation of ferritinophagy induces free iron overload in cells, thus participating in the development of NDs [117]. It has been suggested that HSV-1 encephalitis in mice is associated with nucleic acid damage owing to hydroxyl radical attack, lipid peroxidation, and protein damage mediated by 4-hydroxy-2-nonenal (4-HNE) [118]. Hence, it can be inferred that ferroptosis might be involved in the oxidative damage to neurons triggered by HSV-1 reactivation, which requires further in-depth investigation ( Figure 2 ).

            Figure 2 |

            Stress-induced HSV-1 reactivation triggers neuron damage or death via ROS accumulation.

            Under a series of stressors, latent HSV-1 can be reactivated. In neurons latently-infected with HSV-1, the reactivated virus induces ROS accumulation to activate the apoptosis, autophagy, and ferroptosis pathways via caspase 3 activation, oxidative stress, and lipid peroxidation, respectively, which directly leads to neural damage or death. Moreover, the reactivated virus infects microglia and astrocytes, the primary immune cells in CNS, triggering repeated pro-inflammatory cytokine secretion. Finally, the accumulating pro-inflammatory cytokines indirectly cause deleterious effects on normal or HSV-1 latent neurons.

            In summary, brain tissue is susceptible to oxidative stress due to its composition and functional characteristics [119]. Oxidative stress increases with age and promotes neurodegenerative events associated with HSV-1 infection [112]. Achieving a balance between the oxidant and antioxidant systems in the brain are essential for maintaining neuronal activity and preventing neuronal degeneration [120, 121]. Based on existing research, the factors involved in maintaining a balance between oxidative and anti-oxidative stress are worthy of in-depth study and clarification. Further research will help develop neuroprotective strategies based on antioxidants, which can then be applied to the clinical prevention and treatment of NDs. Taken together, the interaction between HSV-1 and oxidative stress is important because oxidative damage occurs early in the pathogenesis of NDs.

            6. REACTIVATION OF LATENT HSV-1 IS INVOLVED IN THE OCCURRENCE AND DEVELOPMENT OF NDs

            NDs are characterized by progressive deterioration of CNS structure, which leads to impaired motor or cognitive function. The etiology of NDs is complex and includes genetic and environmental factors. Increased neuroinflammation and oxidative stress are key factors contributing to NDs, and in turn, oxidative damage and neuroinflammation induced by reactivation of HSV-1 may be intrinsic molecular mechanisms in the occurrence and development of NDs. In this section, we discuss the molecular signals and mechanisms in which reactivation of HSV-1 might be involved in NDs to provide new perspectives for developing therapy strategies.

            6.1 Reactivation of latent HSV-1 increases susceptibility to AD

            AD is a complex ND with an insidious onset and slow progression. The clinical manifestations of memory loss and progressive cognitive decline significantly reduces the quality of life. A possible link between the reactivation of latent HSV-1 and NDs has been proposed by recent studies. Wozniak et al. reported that a high frequency of HSV-1 DNA is detected in the brains of AD patients, but only a small proportion is detected in healthy control brains [122]. More recently, De Chiara et al. observed an accumulation of AD hallmarks, including amyloid-β protein, tau hyperphosphorylation, and neuroinflammation, in a murine model of serial recurrent HSV-1 infections [123]. Irreversible cognitive deficits occurred after seven cycles of reactivation [123]. Protto et al. concluded that the level of oxidative stress is increased in murine brains, and expression of 4- and 13-HNE-modified proteins were noted in the cortex after multiple stress-induced HSV-1 reactivations [124]. Taken together, HSV-1 infection is probably an important etiologic agent for AD, and the periodic reactivation of HSV-1 in the brain during episodes of stress can lead to cumulative neural cell damage [125].

            6.2 Reactivation of latent HSV-1 increases susceptibility to PD

            PD is a progressive ND with motor and non-motor symptoms, pathologic manifestations of dopaminergic neuron loss, and the presence of Lewy bodies in the midbrain [126]. It has been shown that apoptosis, autophagy, and ferroptosis are all involved in the degenerative loss of dopaminergic neurons. In addition, inflammatory responses and oxidative damage are also found in brains of PD patients. Primary HSV-1 infection causes neurotoxicity with increased ROS levels in the brain and neural cells [106]. It has been demonstrated that activated astrocytes are involved in central neuroinflammation, oxidative stress, and mitochondrial function abnormalities of DA neurons, which initiates death of neurons [120]. In addition, DA is oxidized by ferric ion to generate numerous reductive ions, which initiates a Fenton reaction with hydrogen peroxide. Through the Fenton reaction, hydroxyl radicals with higher cytotoxicity are generated and trigger severe lipid peroxidation, leading to apoptosis or death of substantia nigra neurons [120]. Increased ions can also cause mitochondrial dysfunction of neurons, thereby leading to dopaminergic neuron death [127]. Lipid peroxidation products, such as 4-HNE and malondialdehyde, may distort the physiologic function of neurons by crosslinking proteins and DNA, and even cause neuronal death [128]. In conclusion, oxidative stress and neuroinflammation mediated by latent HSV-1 reactivation has an important role in the pathogenesis of PD.

            6.3 Reactivation of latent HSV-1 increases susceptibility to HD

            HD is a hereditary autosomal dominant ND that is clinically characterized by chorea, loss of coordination, and cognitive decline with destruction of neurons in the subcortical parts of the cerebral hemispheres [129]. Oxidative stress and chronic neuroinflammation are likely complicit in driving progression of HD. It has been shown that selective activation of nuclear factor erythroid 2-related factor 2 (NRF2) signaling effectively inhibits the release of pro-inflammatory cytokines, such as IL-6, in murine microglia and astrocytes. In addition, NRF2 suppresses pro-inflammatory cytokines (IL-1, IL-6, IL-8, and TNF-α) in blood monocytes from HD patients and healthy subjects [130]. The levels of neuroinflammation-related mediator expression in the striatum, cortex, and cerebellum from post-mortem HD patient samples and controls were compared. CCL2 and IL-10 were shown to be increased in the striatum of HD patients and IL-6, IL-8, and matrix metalloproteinase 9 (MMP9) was upregulated in the cerebral cortex and cerebellum [131]. Therefore, oxidative stress and neuroinflammation mediated by latent HSV-1 reactivation have a significant role in the pathogenesis of HD.

            6.4 Reactivation of latent HSV-1 increases susceptibility to ALS

            ALS is the most common motor neuron disease and is characterized by the progressive degeneration of motor neurons in the brain and spinal cord [132]. To date, numerous mechanisms have been reported to be associated with ALS, including oxidative stress, neuroinflammation, excitotoxicity, impaired neurotrophic support, and mitochondrial dysfunction [133, 134]. Microglia and astrocyte proliferation were found in the motor neuron degeneration region in patients with ALS [135]. Frakes et al. reported that NF-κB is upregulated in the spinal cords of ALS patients and SOD1-G93A mice, which serves as a therapeutic target for familial and sporadic ALS patients [136]. Our previous research showed that the antioxidant enzyme, glutathione peroxidase 4 (GPX4), is significantly decreased in the lumbar spinal cord of a murine ALS model compared with the control group; however, a supplemental intrathecal injection of Gpx4-AAV significantly alleviates the symptoms and progression of ALS. In addition, Wang et al. found that GPX4 is depleted in the spinal cords of post-mortem ALS patients, which is consistent with the spinal cord and early brain findings in transgenic animal models with ALS [137]. Oxidative stress and neuroinflammatory responses may be responsible for the ALS pathologic process. Therefore, we reasoned that latent HSV-1 reactivation exacerbates disease progression of ALS by inducing neuroinflammation and oxidative damage of motor neurons.

            7. POTENTIAL THERAPEUTIC EFFECT OF TCM ON HSV-1-INDUCED NDS

            TCM is an effective clinical resource for the treatment of NDs, but little evidence indicates the relationship between the neuroprotective effect of TCM and inhibition of latent HSV-1 reactivation. Currently, no treatment eliminates latent HSV-1 virus in neurons, while acyclovir and other purine nucleoside analogues only inhibit the replication of reactivated virus with increasing drug resistance. An advantage of TCM is prevention of latent HSV-1 reactivation and increasing the difficulty of virus reactivation by regulating the host NEI network. In addition, the antioxidative and antiviral effects of TCM also controls viral replication and oxidative stress in host cells, thus reducing damage to central and peripheral tissues ( Figure 3 ). Hence, we discuss the possible neuroprotective mechanism underlying TCM through inhibition of HSV-1 reactivation and replication in four ways.

            Figure 3 |

            Potential therapeutic effects of TCM on HSV-1-induced NDs.

            TCM alleviates HSV-1-induced NDs by controlling viral replication and reactivation and reducing damage to neurons. The inhibitory mechanism underlying TCM against HSV-1 activation and replication can be summarized as follows: 1) anti-stress effects; 2) immunomodulatory effects; 3) anti-oxidant effects; and 4) anti-viral effects.

            7.1 Anti-stress TCM activity

            TCM is widely used for the treatment of a variety of physical and psychological stress-related disorders in China. The anti-stress TCM activity is related to a regulatory effect on the neuroendocrine system. Collective evidence supports the inhibitory effect of TCM on the overactive HPA axis and abnormal GC release under stress conditions [138, 139]. Our previous studies showed that excessive GCs impair host innate immunity and render the host susceptible to influenza viruses and HSV-1 [65, 140]. As mentioned above, GCs inhibit host immune surveillance of latent HSV-1. TCM may control GC levels in stressed hosts, and hence reduce latent HSV-1 reactivation and the risk of non-hereditary NDs. In addition to Chinese medicine, TCM includes non-drug therapies, such as acupuncture, that has been shown to modulate the NEI network according to recent studies [141]. Similarly, electroacupuncture, an improved form of traditional Chinese acupuncture, inhibits hyperactivity of the HPA axis for the treatment of depression or depressive disorder [142]. Electroacupuncture has been shown to improve HSV-1 infection-induced facial nerve palsy with a decrease in HSV-1 DNA content in mice, indicating the efficacy of electroacupuncture to promote the recovery of nerves from HSV-1 infection-related injury [143]. Harmine, isolated from the seeds of Peganum harmala, is a natural tricyclic β-carboline alkaloid with a core indole structure and a pyridine ring [144]. Harmine is a selective inhibitor of tau phosphorylation at multiple AD-related sites. Frost et al. showed that β-carboline compounds, including harmine, not only effectively reduces the expression of phosphorylated forms of tau protein, but also inhibits the direct phosphorylation of tau protein on serine 396 catalyzed by the DYRK1A gene [145]. Harmine blocks HSV-1 infection by downregulating the cellular NF-κB and MAPK pathways [146]. In 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD mice, baicalin treatment effectively prevents the loss of TH-positive neurons in the substantia nigra and decreases the DA content in the striatum [147]. Our previous results indicated that the anti-HSV-1 effect of baicalin involves two pathways, including inactivation of viral particles and inhibition of phosphorylation of IκB kinase-β (IKK-β) [148]. Baicalin is a promising candidate for the treatment of HSV-1 infection and reactivation.

            7.2 Immunomodulatory effect of TCM

            TCM regulates host immune function to cope with exogenous pathogenic microbe infections. Three TCM decoctions and three formulated Chinese medicines have been widely applied in the treatment of COVID-19 in the absence of effective anti-SARS-CoV-2 drugs in China [149, 150]. It has been reported that TCM and its active ingredients modulate innate and acquired immunity. In addition to exerting an effect on the neuroendocrine pathway, TCM also has a direct regulatory effect on immune cells [151, 152]. Several studies have demonstrated that TCM enhances immune defenses of the host against HSV-1. Shi et al. discovered that astragalus polysaccharide promotes TLR3/NF-κB-mediated immunologic response and hence protects astrocytes from HSV-1 infection-induced cell damage [153]. Total flavonoids from Ixeris Sonchifolia markedly increase the levels of IL-2 and INF-γ in serum of HSV-1 infected mice, and alleviates pathologic damage to the cornea [154]. Moreover, moxibustion, another non-pharmaceutical technique in TCM, markedly increases the survival rate of mice infected with lethal doses of HSV-1, and is correlated with the augmentation of natural killer cell activity and cytokine production, such as IFN [155]. Therefore, TCM might reduce damage to the CNS through inhibiting latent HSV-1 reactivation or improving the clearance of HSV-1 by immunity enhancement of people under stress or with low immunity.

            More recently it has been shown that gut microbiota and its metabolites act as an important regulator of the neuroimmune system, and a dysregulated gut microbiome-neuroimmune axis contributes to neurologic disorders [156]. A recent study showed that an intestinal neomycin-sensitive bacteria metabolite, nicotinamide N-oxide, inhibits microglia activation and neuroinflammation of the CNS by promoting mitophagy to prevent herpes simplex encephalitis [156]. Polysaccharides are common active ingredients in TCM, which can be degraded and absorbed by intestinal bacteria for production of short-chain fatty acids (SCFAs), which are beneficial to host hemostasis [157]. Importantly, SCFAs are recognized as a mediator in gut-brain axis communication to help the recovery of various neurologic diseases, including AD and PD [158]. In addition, TCM polysaccharides modulate the composition of the beneficial and pathogenic bacteria to relieve intestinal dysbiosis due to an unbalanced diet, antibiotic treatment, stress stimuli, or some disease conditions [159].

            7.3 TCM antioxidative effect

            Chinese medicinal herbs are a source of natural antioxidants against oxidative stress or oxidative damage and show great potential for neuroprotection of dopaminergic neurons in the treatment of PD [160, 161]. In fact, counteracting oxidative stress is an important strategy to prevent or treat a wide variety of NDs. Free radical scavenging can reduce oxidation of protein, DNA, and lipids, and inhibit downstream signaling pathways activated by ROS to protect neurons from inflammatory autophagy and cell death [162]. Furthermore, Kim et al. found that a Mori ramulus ethanol extract and its component (morusin) block HSV-1-induced production of ROS in Vero cells, which may explain the antiviral activity [163]. The relationship between oxidative stress and HSV-1 replication, however, needs to be further elucidated. The neuroprotective effect of TCM based on mechanisms against oxidative damage is an important approach to prevent and treat neurodegenerative diseases.

            7.4 TCM antiviral effect

            The damage of host cells induced by latent HSV-1 reactivation also depends on the outcome of viral replication. Hence, TCM functions as an antiviral reagent to inhibit viral replication in neurons, reducing activation of microglia and subsequent neuroinflammation. We have thoroughly reviewed the anti-HSV-1 effects of TCM and its active ingredients in vitro and in vivo [164]. In addition, our laboratory found that baicalein, a flavonoid from Scutellaria baicalensis Georgi, demonstrates a strong inhibitory effect on the replication of HSV-1/F and acyclovir-resistant HSV-1/Blue through dual mechanisms of inactivating viral particles and inhibiting the phosphorylation of IKK-β [148]. Hence, TCM not only directly inhibits the activity of virus, but also regulates the function of host cells to reduce their susceptibility, which may explain the decreased resistance that has emerged in the treatment of viral infectious diseases with TCM.

            8. CONCLUSIONS AND PERSPECTIVES

            With acceleration of the life rhythm, the increasing work pressure and environment deterioration has become an extremely important factor in the occurrence, development, and recovery of diseases. TCM has proposed the theory of emotion changes inducing diseases, and it is gaining increasing value and significance in the etiology of modern diseases. At present, HSV-1 is one of the most widespread viruses known to cause acute and recurrent infections in humans, with an adult infection rate of 60%–95% [165]. The stress of the organism is the direct cause of HSV-1 recurrence, and the occurrence and development of NDs are closely related to latent pathogens in the brain. The incidence of dementia among individuals infected with HSV-1 is 2.564 times the control group, and the use of anti-herpes drugs is associated with a reduced risk of dementia [166]. When exposed to stress events, latent HSV-1 is repeatedly reactivated, leading to nerve cell damage and accelerating the progression of NDs [123, 167]. Therefore, improving an organism’s ability to resist stress or reduce susceptibility to recurrence caused by stress will help prevent HSV-1 reactivation. In this review we discussed stress-induced reactivation of latent HSV-1, which not only induces an organism’s neuroinflammatory reaction, but also leads to an oxidative stress response. The mechanism by which stress causes HSV-1 reactivation needs further exploration. In addition, we also discussed the anti-HSV-1 mechanism underlying TCM from multiple components, targets, and perspectives. It has been demonstrated that TCM and its active ingredients have broad prospects in the treatment of HSV-1.

            ABBREVIATIONS

            ACTH, adrenocorticotrophic hormone; AD, Alzheimer’s disease; CD8+, cluster differentiation 8+ T cells; CNS, central nervous system; CORT, corticosterone; CRH, corticotropin-releasing hormone; DA, dopamine; E, early; GCs, glucocorticoids; GR, glucocorticoid receptor; HPA, hypothalamus-pituitary-adrenal; HSV-1, herpes simplex virus type 1; ICP0, the infected cell protein 0; IE, immediate early; IFN, interferon; IFN-I, type I interferon; IFN-gamma, interferon-γ; L, late; LAT, latency-associated transcript; NDs, neurodegenerative diseases; NGF, nerve growth factor; PD, Parkinson’s disease; PML, Promyelocytic leukemia; ROS, reactive oxygen species; TH, thyroid hormones; TLRs, toll-like receptors; TNF-alpha, tumor necrosis factor-α; TCM, Traditional Chinese medicine; TRH, thyrotropin-releasing hormone; TSH, thyroid-stimulating hormone; UV, ultraviolet ray; 4-HNE, 4-hydroxy-2-nonenal; GPX4, glutathione peroxidase 4.

            ACKNOWLEDGEMENTS

            This study was partly supported by Natural Science Foundation of China (grant numbers 82125038, 81873209, U1801284, and 81673709), the Local Innovative and Research Teams Project of Guangdong Pearl River Talents Program (grant number 2017BT01Y036), GDUPS (2019), the Guangdong Science and Technology Foundation for Distinguished Young Scholars (grant number 2017A030306004), and the Science and Technology Program of Guangzhou (grant number 201903010062). The authors (R.R. He and Y.F. Li) gratefully acknowledge the support of the K. C. Wong Education Foundation.

            CONFLICTS OF INTEREST

            The authors declare no conflicts of interest regarding the publication of this paper.

            REFERENCES

            1. Yan C, Luo Z, Li W, Li X, Dallmann R, Kurihara H, et al.. Disturbed Yin-Yang Balance: Stress Increases the Susceptibility to Primary and Recurrent Infections of Herpes Simplex Virus Type 1. Acta Pharmaceutica Sinica. B. 2020. Vol. 10:383–398

            2. Arvanitakis Z, Shah RC, Bennett DA. Diagnosis and Management of Dementia: Review. JAMA. 2019. Vol. 322:1589–1599

            3. Rowe AM, St Leger AJ, Jeon S, Dhaliwal DK, Knickelbein JE, Hendricks RL. Herpes Keratitis. Progress in Retinal Eye Research. 2013. Vol. 32:88–101

            4. Pires de Mello CP, Bloom DC, Paixão IC. Herpes Simplex Virus Type-1: Replication, Latency, Reactivation and its Antiviral Targets. Antiviral Therapy. 2016. Vol. 21:277–286

            5. Duarte LF, Farías MA, Álvarez DM, Bueno SM, Riedel CA, González PA. Herpes Simplex Virus Type 1 Infection of the Central Nervous System: Insights into Proposed Interrelationships with Neurodegenerative Disorders. Frontiers in Cellular Neuroscience. 2019. Vol. 13:46

            6. Khadr L, Harfouche M, Omori R, Schwarzer G, Chemaitelly H, Abu-Raddad LJ. The Epidemiology of Herpes Simplex Virus Type 1 in Asia: Systematic Review, Meta-Analyses, and Meta-Regressions. Clinical Infectious Diseases. 2019. Vol. 68:757–772

            7. Knipe DM, Cliffe A. Chromatin Control of Herpes Simplex Virus Lytic and Latent Infection. Nature Reviews Microbiology. 2008. Vol. 6:211–221

            8. D’Aiuto L, Bloom DC, Naciri JN, Smith A, Edwards TG, McClain L, et al.. Modeling Herpes Simplex Virus 1 Infections in Human Central Nervous System Neuronal Cells using Two- and Three-dimeansional Cultures Derived from Induced Pluripotent Stem Cells. Journal of Virology. 2019. Vol. 93:e00111–e00119

            9. James SH, Prichard MN. Current and Future Therapies for Herpes Simplex Virus Infections: Mechanism of Action and Drug Resistance. Current Opinion in Virology. 2014. Vol. 8:54–61

            10. Kollias CM, Huneke RB, Wigdahl B, Jennings SR. Animal Models of Herpes Simplex Virus Immunity and Pathogenesis. Journal of Neurovirology. 2015. Vol. 21:8–23

            11. Perng GC, Slanina SM, Yukht A, Ghiasi H, Nesburn AB, Wechsler SL. The Latency-Associated Transcript Gene Enhances Establishment of Herpes Simplex Virus Type 1 Latency in Rabbits. Journal of Virology. 2000. Vol. 74:1885–1891

            12. Zheng X, Marquart ME, Loustch JM, Shah P, Sainz B, Ray A, et al.. HSV-1 Migration in Latently Infected and Naive Rabbits after Penetrating Keratoplasty. Investigative Ophthalmology & Visual Science. 1999. Vol. 40:2490–2497

            13. Yu W, Geng S, Suo Y, Wei X, Cai Q, Wu B, et al.. Critical Role of Regulatory T Cells in the Latency and Stress-Induced Reactivation of HSV-1. Cell Reports. 2018. Vol. 25:2379–2389.e3

            14. Bigley NJ. Complexity of Interferon-γ Interactions with HSV-1. Frontiers in Immunology. 2014. Vol. 5:15

            15. Maringer K, Stylianou J, Elliott G. A Network of Protein Interactions around the Herpes Simplex Virus Tegument Protein VP22. Journal of Virology. 2012. Vol. 86:12971–12982

            16. Wysocka J, Herr W. The Herpes Simplex Virus VP16-Induced Complex: The Makings of a Regulatory Switch. Trends in Biochemical Sciences. 2003. Vol. 28:294–304

            17. Sears AE, Hukkanen V, Labow MA, Levine AJ, Roizman B. Expression of the Herpes Simplex Virus 1 Alpha Transinducing Factor (VP16) Does Not Induce Reactivation of Latent Virus or Prevent the Establishment of Latency in Mice. Journal of Virology. 1991. Vol. 65:2929–2935

            18. Thompson RL, Preston CM, Sawtell NM. De Novo Synthesis of VP16 Coordinates the Exit from HSV Latency In Vivo. PLoS Pathogens. 2009. Vol. 5:e1000352

            19. Herrera FJ, Triezenberg SJ. VP16-Dependent Association of Chromatin-Modifying Coactivators and Underrepresentation of Histones at Immediate-Early Gene Promoters during Herpes Simplex Virus Infection. Journal of Virology. 2004. Vol. 78:9689–9696

            20. Suazo PA, Ibañez FJ, Retamal-Díaz AR, Paz-Fiblas MV, Bueno SM, Kalergis AM, et al.. Evasion of Early Antiviral Responses by Herpes Simplex Viruses. Mediators of Inflammation. 2015. Vol. 2015:593757

            21. Hafezi W, Lorentzen EU, Eing BR, Müller M, King NJ, Klupp B, et al.. Entry of Herpes Simplex Virus Type 1 (HSV-1) into the Distal Axons of Trigeminal Neurons Favors the Onset of Nonproductive, Silent Infection. PLoS Pathogens. 2012. Vol. 8:e1002679

            22. Thompson RL, Sawtell NM. Herpes Simplex Virus Type 1 Latency-Associated Transcript Gene Promotes Neuronal Survival. Journal of Virology. 2001. Vol. 75:6660–6675

            23. Umbach JL, Kramer MF, Jurak I, Karnowski HW, Coen DM, Cullen BR. MicroRNAs Expressed by Herpes Simplex Virus 1 during Latent Infection Regulate Viral mRNAs. Nature. 2008. Vol. 454:780–783

            24. Lee JS, Raja P, Pan D, Pesola JM, Coen DM, Knipe DM. CCCTC-Binding Factor Acts as a Heterochromatin Barrier on Herpes Simplex Viral Latent Chromatin and Contributes to Poised Latent Infection. mBio. 2018. Vol. 9:e02372–17

            25. Egan KP, Wu S, Wigdahl B, Jennings SR. Immunological Control of Herpes Simplex Virus Infections. Journal of Neurovirology. 2013. Vol. 19:328–345

            26. Nicoll MP, Hann W, Shivkumar M, Harman LE, Connor V, Coleman HM, et al.. The HSV-1 Latency-Associated Transcript Functions to Repress Latent Phase Lytic Gene Expression and Suppress Virus Reactivation from Latently Infected Neurons. PLoS Pathogens. 2016. Vol. 12:e1005539

            27. Carpenter D, Henderson G, Hsiang C, Osorio N, BenMohamed L, Jones C, et al.. Introducing Point Mutations into the ATGs of the Putative Open Reading Frames of the HSV-1 Gene Encoding the Latency Associated Transcript (LAT) Reduces its Anti-Apoptosis Activity. Microbial Pathogenesis. 2008. Vol. 44:98–102

            28. Thomas SK, Lilley CE, Latchman DS, Coffin RS. A Protein Encoded by the Herpes Simplex Virus (HSV) Type 1 2-Kilobase Latency-Associated Transcript is Phosphorylated, Localized to the Nucleus, and Overcomes the Repression of Expression from Exogenous Promoters when Inserted into the Quiescent HSV Genome. Journal of Virology. 2002. Vol. 76:4056–4067

            29. Wilson AC, Mohr I. A Cultured Affair: HSV Latency and Reactivation in Neurons. Trends in Microbiology. 2012. Vol. 20:604–611

            30. Neumann DM, Bhattacharjee PS, Hill JM. Sodium Butyrate: A Chemical Inducer of in Vivo Reactivation of Herpes Simplex Virus Type 1 in the Ocular Mouse Model. Journal of Virology. 2007. Vol. 81:6106–6110

            31. Messer HG, Jacobs D, Dhummakupt A, Bloom DC. Inhibition of H3K27me3-Specific Histone Demethylases JMJD3 and UTX Blocks Reactivation of Herpes Simplex Virus 1 in Trigeminal Ganglion Neurons. Journal of Virology. 2015. Vol. 89:3417–3420

            32. Lian S, Wang D, Xu B, Guo W, Wang L, Li W, et al.. Prenatal Cold Stress: Effect on Maternal Hippocampus and Offspring Behavior in Rats. Behavioural Brain Research. 2018. Vol. 346:1–10

            33. Huo C, Xiao C, She R, Liu T, Tian J, Dong H, et al.. Chronic Heat Stress Negatively Affects the Immune Functions of Both Spleens and Intestinal Mucosal System in Pigs through the Inhibition of Apoptosis. Microbial Pathogenesis. 2019. Vol. 136:103672

            34. Chen H, Jie C, Tang LP, Meng H, Li XB, Li YB, et al.. New Insights into the Effects and Mechanism of a Classic Traditional Chinese Medicinal Formula on Influenza Prevention. Phytomedicine. 2017. Vol. 27:52–62

            35. Wang SF, Chen XH, He B, Yin DD, Gao HJ, Zhao HQ, et al.. Acute Restraint Stress Triggers Progesterone Withdrawal and Endometrial Breakdown and Shedding Through Corticosterone Stimulation in Mouse Menstrual-Like Model. Reproduction. 2019. Vol. 157:149–161

            36. He Q, Sawada M, Yamasaki N, Akazawa S, Furuta H, Uenishi H, et al.. Neuroinflammation, Oxidative Stress, and Neurogenesis in a Mouse Model of Chronic Fatigue Syndrome, and the Treatment with Kampo Medicine. Biological & Pharmaceutical Bulletin. 2020. Vol. 43:110–115

            37. Enkel T, Spanagel R, Vollmayr B, Schneider M. Stress Triggers Anhedonia in Rats Bred for Learned Helplessness. Behavioural Brain Research. 2010. Vol. 209:183–186

            38. Xiao B, Han F, Wang HT, Shi YX. Single-Prolonged Stress Induces Increased Phosphorylation of Extracellular Signal-Regulated Kinase in a Rat Model of Post-Traumatic Stress Disorder. Molecular Medicine Reports. 2011. Vol. 4:445–449

            39. Di Marco S, Carnicelli V, Franceschini N, Di Paolo M, Piccardi M, Bisti S, et al.. Saffron: A Multitask Neuroprotective Agent for Retinal Degenerative Diseases. Antioxidants (Basel). 2019. Vol. 8:224

            40. Adamec RE, Blundell J, Burton P. Neural Circuit Changes Mediating Lasting Brain and Behavioral Response to Predator Stress. Neuroscience and Biobehavioral Reviews. 2005. Vol. 29:1225–1241

            41. Reshetnikov VV, Kovner AV, Lepeshko AA, Pavlov KS, Grinkevich LN, Bondar NP. Stress Early in Life Leads to Cognitive Impairments, Reduced Numbers of CA3 Neurons and Altered Maternal Behavior in Adult Female Mice. Genes Brain and Behavior. 2020. Vol. 19:e12541

            42. Morakinyo AO, Samuel TA, Awobajo FO, Adekunbi DA, Olatunji IO, Binibor FU, et al.. Adverse Effects of Noise Stress on Glucose Homeostasis and Insulin Resistance in Sprague-Dawley Rats. Heliyon. 2019. Vol. 5:e03004

            43. Fanous S, Hammer RP Jr, Nikulina EM. Short- and Long-Term Effects of Intermittent Social Defeat Stress on Brain-Derived Neurotrophic Factor Expression in Mesocorticolimbic Brain Regions. Neuroscience. 2010. Vol. 167:598–607

            44. Götz AA, Stefanski V. Psychosocial Maternal Stress during Pregnancy Affects Serum Corticosterone, Blood Immune Parameters and anxiety Behaviour in Adult Male Rat Offspring. Physiology & Behavior. 2007. Vol. 90:108–115

            45. Padgett DA, Sheridan JF, Dorne J, Berntson GG, Candelora J, Glaser R. Social Stress and the Reactivation of Latent Herpes Simplex Virus Type 1. Proceedings of the National Academy of Sciences of the United States of America. 1998. Vol. 95:7231–7235

            46. Pan MH, Zhu SR, Duan WJ, Ma XH, Luo X, Liu B, et al.. “Shanghuo” Increases Disease Susceptibility: Modern Significance of an Old TCM Theory. Journal of Ethnopharmacology. 2020. Vol. 250:112491

            47. Zhu SR, Luo X, Li YF, Hiroshi K, He RR. Emotional Stress-Induced Shanghuo Syndrome Increases Disease Susceptibility. Zhongguo Zhong Yao Za Zhi. 2018. Vol. 43:1529–1535

            48. Ortiz GC, Sheridan JF, Marucha PT. Stress-Induced Changes in Pathophysiology and Interferon Gene Expression during Primary HSV-1 Infection. Brain Behavior and Immunity. 2003. Vol. 17:329–338

            49. Freeman ML, Sheridan BS, Bonneau RH, Hendricks RL. Psychological Stress Compromises CD8+ T Cell Control of Latent Herpes Simplex Virus Type 1 Infections. Journal of Immunology. 2007. Vol. 179:322–328

            50. Romero ML, Butler LK. Endocrinology of Stress. International Journal of Comparative Psychology. 2007. Vol. 20:89–95

            51. Russell G, Lightman S. The Human Stress Response. Nature Reviews Endocrinology. 2019. Vol. 15:525–534

            52. Yao H, Ma R, Yang L, Hu G, Chen X, Duan M, et al.. MiR-9 Promotes Microglial Activation by Targeting MCPIP1. Nature Communications. 2014. Vol. 5:4386

            53. Shih AY, Fernandes HB, Choi FY, Kozoriz MG, Liu Y, Li P, et al.. Policing the Police: Astrocytes Modulate Microglial Activation. Journal of Neuroscience. 2006. Vol. 26:3887–3888

            54. Galliher-Beckley AJ, Cidlowski JA. Emerging Roles of Glucocorticoid Receptor Phosphorylation in Modulating Glucocorticoid Hormone Action in Health and Disease. IUBMB Life. 2009. Vol. 61:979–986

            55. Cohen S, Janicki-Deverts D, Doyle WJ, Miller GE, Frank E, Rabin BS, et al.. Chronic Stress, Glucocorticoid Receptor Resistance, Inflammation, and Disease Risk. Proceedings of the National Academy of Sciences of the United States of America. 2012. Vol. 109:5995–5999

            56. Jeanneteau F, Chao MV. Are BDNF and Glucocorticoid Activities Calibrated? Neuroscience. 2013. Vol. 239:173–195

            57. Ismaili N, Garabedian MJ. Modulation of Glucocorticoid Receptor Function via Phosphorylation. Annals of the New York Academy of Sciences. 2004. Vol. 1024:86–101

            58. Galliher-Beckley AJ, Williams JG, Cidlowski JA. Ligand-Independent Phosphorylation of the Glucocorticoid Receptor Integrates Cellular Stress Pathways with Nuclear Receptor Signaling. Journal of Molecular Cell Biology. 2011. Vol. 31:4663–4675

            59. Lambert WM, Xu CF, Neubert TA, Chao MV, Garabedian MJ, Jeanneteau FD. Brain-Derived Neurotrophic Factor Signaling Rewrites the Glucocorticoid Transcriptome via Glucocorticoid Receptor Phosphorylation. Journal of Molecular Cell Biology. 2013. Vol. 33:3700–3714

            60. Ostler JB, Harrison KS, Schroeder K, Thunuguntla P, Jones C. The Glucocorticoid Receptor (GR) Stimulates Herpes Simplex Virus 1 Productive Infection, in Part Because the Infected Cell Protein 0 (ICP0) Promoter is Cooperatively Transactivated by the GR and Krüppel-Like Transcription Factor 15. Journal of Virology. 2019. Vol. 93:e02063–18

            61. Wang S, Long J, Zheng CF. The Potential Link between PML NBs and ICP0 in Regulating Lytic and Latent Infection of HSV-1. Protein Cell. 2012. Vol. 3:372–382

            62. Everett RD, Chelbi-Alix MK. PML and PML Nuclear Bodies: Implications in Antiviral Defence. Biochimie. 2007. Vol. 89:819–830

            63. Everett RD, Rechter S, Papior P, Tavalai N, Stamminger T, Orr A. PML Contributes to a Cellular Mechanism of Repression of Herpes Simplex Virus Type 1 Infection that is Inactivated by ICP0. Journal of Virology. 2006. Vol. 80:7995–8005

            64. Cuchet D, Sykes A, Nicolas A, Orr A, Murray J, Sirma H, et al.. PML Isoforms I and II Participate in PML-Dependent Restriction of HSV-1 Replication. Journal of Cell Science. 2011. Vol. 124:280–291

            65. Li W, Luo Z, Yan CY, Wang XH, He ZJ, Ouyang SH, et al.. Autophagic Degradation of PML Promotes Susceptibility to HSV-1 by Stress-Induced Corticosterone. Theranostics. 2020. Vol. 10:9032–9049

            66. Figliozzi RW, Chen F, Balish M, Ajavon A, Hsia SV. Thyroid Hormone-Dependent Epigenetic Suppression of Herpes Simplex Virus-1 Gene Expression and Viral Replication in Differentiated Neuroendocrine Cells. Journal of the Neurological Sciences. 2014. Vol. 346:164–173

            67. Hsia SC, Bedadala GR, Balish MD. Effects of Thyroid Hormone on HSV-1 Gene Regulation: Implications in the Control of Viral Latency and Reactivation. Cell and Bioscience. 2011. Vol. 1:24

            68. Rooney JF, Bryson Y, Mannix ML, Dillon M, Wohlenberg CR, Banks S, et al.. Prevention of Ultraviolet-Light-Induced Herpes Labialis by Sunscreen. Lancet. 1991. Vol. 338:1419–1422

            69. Ludema C, Cole SR, Poole C, Smith JS, Schoenbach VJ, Wilhelmus KR. Association between Unprotected Ultraviolet Radiation Exposure and Recurrence of Ocular Herpes Simplex Virus. American Journal of Epidemiology. 2014. Vol. 179:208–215

            70. Zhao C, Hou W, Lei H, Huang L, Wang S, Cui D, et al.. Potassium 2-(l-hydroxypentyl)-Benzoate Attenuates Neuroinflammatory Responses and Upregulates Heme Oxygenase-1 in Systemic Lipopolysaccharide-Induced Inflammation in Mice. Acta Pharmaceutica Sinica B. 2017. Vol. 7:470–478

            71. Allen NJ, Lyons DA. Glia as Architects of Central Nervous System Formation and Function. Science. 2018. Vol. 362:181–185

            72. Acuña-Hinrichsen F, Muñoz M, Hott M, Martin C, Mancilla E, Salazar P, et al.. Herpes Simplex Virus Type 1 Enhances Expression of the Synaptic Protein Arc for its Own Benefit. Frontiers in Cellular Neuroscience. 2018. Vol. 12:505

            73. Rodriguez-Izquierdo I, Serramia MJ, Gomez R, De La Mata FJ, Bullido MJ, Muñoz-Fernández MA. Gold Nanoparticles Crossing Blood-Brain Barrier Prevent HSV-1 Infection and Reduce Herpes Associated Amyloid-βsecretion. Journal of Clinical Medicine. 2020. Vol. 9:155

            74. Wang Y, Jia J, Wang Y, Li F, Song X, Qin S, et al.. Roles of HSV-1 Infection-Induced Microglial Immune Responses in CNS Diseases: Friends or Foes? Critical Reviews in Microbiology. 2019. Vol. 45:581–594

            75. Frank MG, Hershman SA, Weber MD, Watkins LR, Maier SF. Chronic Exposure to Exogenous Glucocorticoids Primes Microglia to Pro-inflammatory Stimuli and Induces NLRP3 mRNA in the Hippocampus. Psychoneuroendocrinology. 2014. Vol. 40:191–200

            76. Frank MG, Thompson BM, Watkins LR, Maier SF. Glucocorticoids Mediate Stress-Induced Priming of Microglial Pro-Inflammatory Responses. Brain Behavior and Immunity. 2012. Vol. 26:337–345

            77. de Pablos RM, Villarán RF, Argüelles S, Herrera AJ, Venero JL, Ayala A, et al.. Stress Increases Vulnerability to Inflammation in the Rat Prefrontal Cortex. Journal of Neuroscience. 2006. Vol. 26:5709–5719

            78. Block ML, Zecca L, Hong JS. Microglia-Mediated Neurotoxicity: Uncovering the Molecular Mechanisms. Nature Reviews Neuroscience. 2007. Vol. 8:57–69

            79. Jurgens HA, Johnson RW. Dysregulated Neuronal-Microglial Cross-Talk during Aging, Stress and Inflammation. Experimental Neurology. 2012. Vol. 233:40–48

            80. Liddelow SA, Guttenplan KA, Clarke LE, Bennett FC, Bohlen CJ, Schirmer L, et al.. Neurotoxic Reactive Astrocytes are Induced by Activated Microglia. Nature. 2017. Vol. 541:481–487

            81. Pekny M, Pekna M, Messing A, Steinhäuser C, Lee JM, Parpura V, et al.. Astrocytes: A Central Element in Neurological Diseases. Acta Neuropathologica. 2016. Vol. 131:323–345

            82. Orihuela R, McPherson CA, Harry GJ. Microglial M1/M2 Polarization and Metabolic States. British Journal of Pharmacology. 2016. Vol. 173:649–665

            83. Subramaniam SR, Federoff HJ. Targeting Microglial Activation States as a Therapeutic Avenue in Parkinson’s Disease. Frontiers in Aging Neuroscience. 2017. Vol. 9:176

            84. Tang Y, Le W. Differential Roles of M1 and M2 Microglia in Neurodegenerative Diseases. Molecular Neurobiology. 2016. Vol. 53:1181–1194

            85. Reinert LS, Lopušná K, Winther H, Sun C, Thomsen MK, Nandakumar R, et al.. Sensing of HSV-1 by the cGAS-STING Pathway in Microglia Orchestrates Antiviral Defence in the CNS. Nature Communications. 2016. Vol. 7:13348

            86. Sato R, Kato A, Chimura T, Saitoh SI, Shibata T, Murakami Y, et al.. Combating Herpesvirus Encephalitis by Potentiating a TLR3-mTORC2 Axis. Nature Immunology. 2018. Vol. 19:1071–1082

            87. Walter S, Letiembre M, Liu Y, Heine H, Penke B, Hao W, et al.. Role of the Toll-Like Receptor 4 in Neuroinflammation in Alzheimer’s Disease. Cellular Physiology and Biochemistry. 2007. Vol. 20:947–956

            88. Go M, Kou J, Lim JE, Yang J, Fukuchi KI. Microglial Response to LPS Increases in Wild-Type Mice during Aging but Diminishes in an Alzheimer’s Mouse Model: Implication of TLR4 Signaling in Disease Progression. Biochemical and Biophysical Research Communications. 2016. Vol. 479:331–337

            89. Chen L, Hu L, Zhao J, Hong H, Feng F, Qu W, et al.. Chotosan Improves Aβ1-42-Induced Cognitive Impairment and Neuroinflammatory and Apoptotic Responses through the Inhibition of TLR-4/NF-κB Signaling in Mice. Journal of Ethnopharmacology. 2016. Vol. 191:398–407

            90. Kurt-Jones EA, Chan M, Zhou S, Wang J, Reed G, Bronson R, et al.. Herpes Simplex Virus 1 Interaction with Toll-like Receptor 2 Contributes to Lethal Encephalitis. Proceedings of the National Academy of Sciences of the United States of America. 2004. Vol. 101:1315–1320

            91. Okun E, Griffioen KJ, Mattson MP. Toll-like Receptor Signaling in Neural Plasticity and Disease. Trends in Neurosciences. 2011. Vol. 34:269–281

            92. Rogers JT, Leiter LM, McPhee J, Cahill CM, Zhan SS, Potter H, et al.. Translation of the Alzheimer Amyloid Precursor Protein mRNA is Up-regulated by Interleukin-1 through 5′-Untranslated Region Sequences. Journal of Biological Chemistry. 1999. Vol. 274:6421–6431

            93. Steinman L. Nuanced Roles of Cytokines in Three Major Human Brain Disorders. Journal of Clinical Investigation. 2008. Vol. 118:3557–3563

            94. Miller ED, Dziedzic A, Saluk-Bijak J, Bijak M. A Review of Various Antioxidant Compounds and their Potential Utility as Complementary Therapy in Multiple Sclerosis. Nutrients. 2019. Vol. 11:1528

            95. Kaur D, Sharma V, Deshmukh R. Activation of Microglia and Astrocytes: A Roadway to Neuroinflammation and Alzheimer’s Disease. Inflammopharmacology. 2019. Vol. 27:663–677

            96. Hirsch EC, Hunot S. Neuroinflammation in Parkinson’s Disease: A Target for Neuroprotection? Lancet Neurology. 2009. Vol. 8:382–397

            97. Björkqvist M, Wild EJ, Thiele J, Silvestroni A, Andre R, Lahiri N, et al.. A Novel Pathogenic Pathway of Immune Activation Detectable before Clinical Onset in Huntington’s disease. Journal of Experimental Medicine. 2008. Vol. 205:1869–1877

            98. Haidet-Phillips AM, Hester ME, Miranda CJ, Meyer K, Braun L, Frakes A, et al.. Astrocytes from Familial and Sporadic ALS Patients are Toxic to Motor Neurons. Nature Biotechnology. 2011. Vol. 29:824–828

            99. Jin M, Günther R, Akgün K, Hermann A, Ziemssen T. Peripheral Proinflammatory Th1/Th17 Immune Cell Shift is Linked to Disease Severity in Amyotrophic Lateral Sclerosis. Scientific Reports. 2020. Vol. 10:5941

            100. Coles LD, Tuite PJ, Öz G, Mishra UR, Kartha RV, Sullivan KM, et al.. Repeated-Dose Oral N-Acetylcysteine in Parkinson’s Disease: Pharmacokinetics and Effect on Brain Glutathione and Oxidative Stress. Journal of Clinical Pharmacology. 2018. Vol. 58:158–167

            101. De Chiara G, Marcocci ME, Civitelli L, Argnani R, Piacentini R, Ripoli C, et al.. APP Processing Induced by Herpes Simplex Virus Type 1 (HSV-1) Yields Several APP Fragments in Human and Rat Neuronal Cells. PLoS One. 2010. Vol. 5:e13989

            102. Valyi-Nagy T, Dermody TS. Role of Oxidative Damage in the Pathogenesis of Viral Infections of the Nervous System. Histology and Histopathology. 2005. Vol. 20:957–967

            103. Wang S, Irving G, Jiang L, Wang H, Li M, Wang X, et al.. Oxidative Stress Mediated Hippocampal Neuron Apoptosis Participated in Carbon Disulfide-Induced Rats Cognitive Dysfunction. Neurochemical Research. 2017. Vol. 42:583–594

            104. Webb SJ, Harrison DJ, Wyllie AH. Apoptosis: An Overview of the Process and its Relevance in Disease. Advances in Pharmacology. 1997. Vol. 41:1–34

            105. Aubert M, Blaho JA. Modulation of Apoptosis during Herpes Simplex Virus Infection in Human Cells. Microbes and Infection. 2001. Vol. 3:859–866

            106. Schachtele SJ, Hu S, Little MR, Lokensgard JR. Herpes Simplex Virus Induces Neural Oxidative Damage via Microglial Cell Toll-Like Receptor-2. Journal of Neuroinflammation. 2010. Vol. 7:35

            107. Zhong H, Xiao M, Zarkovic K, Zhu M, Sa R, Lu J, et al.. Mitochondrial Control of Apoptosis through Modulation of Cardiolipin Oxidation in Hepatocellular Carcinoma: A Novel Link between Oxidative Stress and Cancer. Free Radical Biology & Medicine. 2017. Vol. 102:67–76

            108. Carpenter D, Hsiang C, Jiang X, Osorio N, BenMohamed L, Jones C, et al.. The Herpes Simplex Virus Type 1 (HSV-1) Latency-Associated Transcript (LAT) Protects Cells against Cold-Shock-Induced Apoptosis by Maintaining Phosphorylation of Protein Kinase B (AKT). Journal of Neurovirology. 2015. Vol. 21:568–575

            109. Jiang X, Chentoufi AA, Hsiang C, Carpenter D, Osorio N, BenMohamed L, et al.. The Herpes Simplex Virus Type 1 Latency-Associated Transcript can Protect Neuron-Derived C1300 and Neuro2A Cells from Granzyme B-Induced Apoptosis and CD8 T-Cell Killing. Journal of Virology. 2011. Vol. 85:2325–2332

            110. Klionsky DJ, Cuervo AM, Dunn WA Jr, Levine B, van der Klei I, Seglen PO. How Shall I Eat Thee? Autophagy. 2007. Vol. 3:413–416

            111. Su LJ, Zhang JH, Gomez H, Murugan R, Hong X, Xu D, et al.. Reactive Oxygen Species-Induced Lipid Peroxidation in Apoptosis, Autophagy, and Ferroptosis. Oxidative Medicine and Cellular Longevity. 2019. Vol. 2019:5080843

            112. Santana S, Sastre I, Recuero M, Bullido MJ, Aldudo J. Oxidative Stress Enhances Neurodegeneration Markers Induced by Herpes Simplex Virus Type 1 Infection in Human Neuroblastoma Cells. PLoS One. 2013. Vol. 8:e75842

            113. Yakoub AM, Shukla D. Autophagy Stimulation Abrogates Herpes Simplex Virus-1 Infection. Scientific Reports. 2015. Vol. 5:9730

            114. Dixon SJ, Lemberg KM, Lamprecht MR, Skouta R, Zaitsev EM, Gleason CE, et al.. Ferroptosis: An Iron-Dependent form of Nonapoptotic Cell Death. Cell. 2012. Vol. 149:1060–1072

            115. Weiland A, Wang Y, Wu W, Lan X, Han X, Li Q, et al.. Ferroptosis and its Role in Diverse Brain Diseases. Molecular Neurobiology. 2019. Vol. 56:4880–4893

            116. Sun Y, Pham AN, Waite TD. The Effect of Vitamin C and Iron on Dopamine-Mediated Free Radical Generation: Implications to Parkinson’s Disease. Dalton Transactions. 2018. Vol. 47:4059–4069

            117. Santana-Codina N, Mancias JD. The Role of NCOA4-Mediated Ferritinophagy in Health and Disease. Pharmaceuticals (Basel). 2018. Vol. 11:114

            118. Valyi-Nagy T, Olson SJ, Valyi-Nagy K, Montine TJ, Dermody TS. Herpes Simplex Virus Type 1 Latency in the Murine Nervous System is Associated with Oxidative Damage to Neurons. Virology. 2000. Vol. 278:309–321

            119. Cenini G, Lloret A, Cascella R. Oxidative Stress in Neurodegenerative Diseases: From a Mitochondrial Point of View. Oxidative Medicine and Cellular Longevity. 2019. Vol. 2019:2105607

            120. Jiang T, Sun Q, Chen S. Oxidative Stress: A Major Pathogenesis and Potential Therapeutic Target of Antioxidative Agents in Parkinson’s Disease and Alzheimer’s Disease. Progress in Neurobiology. 2016. Vol. 147:1–19

            121. Jing X, Wei X, Ren M, Wang L, Zhang X, Lou H. Neuroprotective Effects of Tanshinone I Against 6-OHDA-Induced Oxidative Stress in Cellular and Mouse Model of Parkinson’s Disease through Upregulating Nrf2. Neurochemical Research. 2016. Vol. 41:779–786

            122. Wozniak MA, Mee AP, Itzhaki RF. Herpes Simplex Virus Type 1 DNA is Located within Alzheimer’s Disease Amyloid Plaques. Journal of Pathology. 2009. Vol. 217:131–138

            123. De Chiara G, Piacentini R, Fabiani M, Mastrodonato A, Marcocci ME, Limongi D, et al.. Recurrent Herpes Simplex Virus-1 Infection Induces Hallmarks of Neurodegeneration and Cognitive Deficits in Mice. PLoS Pathogens. 2019. Vol. 15:e1007617

            124. Protto V, Tramutola A, Fabiani M, Marcocci ME, Napoletani G, Iavarone F, et al.. Multiple Herpes Simplex Virus-1 (HSV-1) Reactivations Induce Protein Oxidative Damage in Mouse Brain: Novel Mechanisms for Alzheimer’s Disease Progression. Microorganisms. 2020. Vol. 8:E972

            125. Itzhaki RF. Herpes Simplex Virus Type 1 and Alzheimer’s Disease: Increasing Evidence for a Major Role of the Virus. Frontiers in Aging Neuroscience. 2014. Vol. 6:202

            126. Reich SG, Savitt JM. Parkinson’s Disease. Medical Clinics of North America. 2019. Vol. 103:337–350

            127. Sita G, Hrelia P, Tarozzi A, Morroni F. Isothiocyanates are Promising Compounds against Oxidative Stress, Neuroinflammation and Cell Death that May Benefit Neurodegeneration in Parkinson’s Disease. International Journal of Molecular Sciences. 2016. Vol. 17:1454

            128. Gaschler MM, Stockwell BR. Lipid Peroxidation in Cell Death. Biochemical and Biophysical Research Communications. 2017. Vol. 482:419–425

            129. Vonsattel JP, Myers RH, Stevens TJ, Ferrante RJ, Bird ED, Richardson EP Jr. Neuropathological Classification of Huntington’s Disease. Journal of Neuropathology & Experimental Neurology. 1985. Vol. 44:559–577

            130. Quinti L, Dayalan Naidu S, Träger U, Chen X, Kegel-Gleason K, Llères D, et al.. KEAP1-Modifying Small Molecule Reveals Muted NRF2 Signaling Responses in Neural Stem Cells from Huntington’s Disease Patients. Proceedings of the National Academy of Sciences of the United States of America. 2017. Vol. 114:E4676–E4685

            131. Silvestroni A, Faull RL, Strand AD, Möller T. Distinct Neuroinflammatory Profile in Post-Mortem Human Huntington’s Disease. Neuroreport. 2009. Vol. 20:1098–1103

            132. van Es MA, Hardiman O, Chio A, Al-Chalabi A, Pasterkamp RJ, Veldink JH, et al.. Amyotrophic Lateral Sclerosis. Lancet. 2017. Vol. 390:2084–2098

            133. Pasinelli P, Brown RH. Molecular Biology of Amyotrophic Lateral Sclerosis: Insights from Genetics. Nature Reviews Neuroscience. 2006. Vol. 7:710–723

            134. Cook C, Petrucelli L. Genetic Convergence Brings Clarity to the Enigmatic Red Line in ALS. Neuron. 2019. Vol. 101:1057–1069

            135. Won YH, Lee MY, Choi YC, Ha Y, Kim H, Kim DY, et al.. Elucidation of Relevant Neuroinflammation Mechanisms using Gene Expression Profiling in Patients with Amyotrophic Lateral Sclerosis. PLoS One. 2016. Vol. 11:e0165290

            136. Frakes AE, Ferraiuolo L, Haidet-Phillips AM, Schmelzer L, Braun L, Miranda CJ, et al.. Microglia Induce Motor Neuron Death via the Classical NF-κB Pathway in Amyotrophic Lateral Sclerosis. Neuron. 2014. Vol. 81:1009–1023

            137. Wang T, Tomas D, Perera ND, Cuic B, Luikinga S, Viden A, et al.. Ferroptosis Mediates Selective Motor Neuron Death in Amyotrophic Lateral Sclerosis. Cell Death & Differentiation. 2022. Vol. 29:1187–1198

            138. Li C, Huang J, Cheng Y-C, Zhang Y-W. Traditional Chinese medicine in depression treatment: from molecules to systems. Frontiers in Pharmacology. 2020. Vol. 11:586

            139. Li S, Zhang ZQ, Wu LJ, Zhang XG, Li YD, Wang YY. Understanding ZHENG in Traditional Chinese Medicine in the Context of Neuro-endocrine-immune Network. IET Systems Biology. 2007. Vol. 1:51–60

            140. Luo Z, Liu LF, Jiang YN, Tang LP, Li W, Ouyang SH, et al.. Novel Insights into Stress-Induced Susceptibility to Influenza: Corticosterone Impacts Interferon-β Responses by Mfn2-Mediated Ubiquitin Degradation of MAVS. Signal Transduction Target Therapy. 2020. Vol. 5:202

            141. Xu ZF, Hong SH, Wang SJ, Zhao X, Liu YY, Ding SS, et al.. Neuroendocrine-Immune Regulating Mechanisms for the Anti-Inflammatory and Analgesic Actions of Acupuncture. World Journal of Traditional Chinese Medicine. 2020. Vol. 6:384–392

            142. Han X, Gao Y, Yin X, Zhang Z, Lao L, Chen Q, et al.. The Mechanism of Electroacupuncture for Depression on Basic Research: A Systematic Review. Chinese Medicine. 2021. Vol. 16:10

            143. Tang H, Feng S, Chen J, Yang J, Yang M, Zhong Z, et al.. Effects of Electroacupuncture on Facial Nerve Function and HSV-1 DNA Quantity in HSV-1 Induced Facial Nerve Palsy Mice. Evidence Based Complementary and Alternative Medicine. 2014. Vol. 2014:693783

            144. Siddiqui S, Khan OY, Faizi S, Siddiqui BS. Studies on the Chemical Constituents of the Seeds of Peganum Harmala – Isolation and Structure of a New β-Carboline Alkaloid – Harmalicine. Heterocycles (Sendai). 1987. Vol. 26:1563–1567

            145. Frost D, Meechoovet B, Wang T, Gately S, Giorgetti M, Shcherbakova I, et al.. Beta-Carboline Compounds, Including Harmine, Inhibit DYRK1A and Tau Phosphorylation at Multiple Alzheimer’s Disease-Related Sites. PLoS One. 2011. Vol. 6:e19264

            146. Chen D, Su A, Fu Y, Wang X, Lv X, Xu W, et al.. Harmine Blocks Herpes Simplex Virus Infection through Downregulating Cellular NF-κB and MAPK Pathways Induced by Oxidative Stress. Antiviral Research. 2015. Vol. 123:27–38

            147. Chen X, Zhang N, Zou H. Protective Effect of Baicalin on Mouse with Parkinson’s Disease Induced by MPTP. Zhongguo Zhong xi yi jie he za zhi Zhongguo Zhongxiyi Jiehe Zazhi Chinese Journal of Integrated Traditional and Western Medicine. 2007. Vol. 27:1010–1012

            148. Luo Z, Kuang XP, Zhou QQ, Yan CY, Li W, Gong HB, et al.. Inhibitory Effects of Baicalein against Herpes Simplex Virus Type 1. Acta Pharmaceutica Sinica B. 2020. Vol. 10:2323–2338

            149. Huang K, Zhang P, Zhang Z, Youn JY, Wang C, Zhang H, et al.. Traditional Chinese Medicine (TCM) in the Treatment of COVID-19 and Other Viral Infections: Efficacies and Mechanisms. Pharmacology & Therapeutics. 2021. Vol. 225:107843

            150. Tong T, Wu YQ, Ni WJ, Shen AZ, Liu S. The Potential Insights of Traditional Chinese Medicine on Treatment of COVID-19. Chinese Medicine. 2020. Vol. 15:51

            151. Cui HR, Zhang JY, Cheng XH, Zheng JX, Zhang Q, Zheng R, et al.. Immunometabolism at the Service of Traditional Chinese Medicine. Pharmacological Research. 2022. Vol. 176:106081

            152. Ma HD, Deng YR, Tian Z, Lian ZX. Traditional Chinese Medicine and Immune Regulation. Clinical Reviews in Allergy & Immunology. 2013. Vol. 44:229–241

            153. Shi L, Yin F, Xin X, Mao S, Hu P, Zhao C, et al.. Astragalus Polysaccharide Protects Astrocytes from being Infected by HSV-1 through TLR3/NF-κB Signaling Pathway. Evidence-Based Complementary and Alternative Medicine. 2014. Vol. 2014:285356

            154. Wang YQ, Cai L, Zhang N, Zhang J, Wang HH, Zhu W. Protective Effect of Total Flavonoids from Ixeris Sonchifolia on Herpes Simplex Virus Keratitis in Mice. BMC Complementary Medicine Therapies. 2020. Vol. 20:113

            155. Takayama Y, Itoi M, Hamahashi T, Tsukamoto N, Mori K, Morishita D, et al.. Moxibustion Activates Host Defense against Herpes Simplex Virus Type I through Augmentation of Cytokine Production. Microbiology and Immunology. 2010. Vol. 54:551–557

            156. Yu LW, Agirman G, Hsiao EY. The Gut Microbiome as a Regulator of the Neuroimmune Landscape. Annual Review of Immunology. 2022. Vol. 40:143–167

            157. Feng W, Ao H, Peng C. Gut Microbiota, Short-Chain Fatty Acids, and Herbal Medicines. Frontiers in Pharmacology. 2018. Vol. 9:1354

            158. Silva YP, Bernardi A, Frozza RL. The Role of Short-Chain Fatty Acids from Gut Microbiota in Gut-Brain Communication. Frontiers in Endocrinology (Lausanne). 2020. Vol. 11:25

            159. Yue B, Zong G, Tao R, Wei Z, Lu Y. Crosstalk between Traditional Chinese Medicine-Derived Polysaccharides and the Gut Microbiota: A New Perspective to Understand Traditional Chinese Medicine. Phytotherapy Research. 2022. Vol. 36:4125–4138

            160. Ayton S, Lei P, Hare DJ, Duce JA, George JL, Adlard PA, et al.. Parkinson’s Disease Iron Deposition Caused by Nitric Oxide-Induced Loss of β-Amyloid Precursor Protein. Journal of Neurosciences. 2015. Vol. 35:3591–3597

            161. Muhammad F, Liu Y, Zhou Y, Yang H, Li H. Antioxidative Role of Traditional Chinese Medicine in Parkinson’s disease. Journal of Ethnopharmacology. 2022. Vol. 285:114821

            162. Chen SD, Yang JL, Lin TK, Yang DI. Emerging Roles of Sestrins in Neurodegenerative Diseases: Counteracting Oxidative Stress and Beyond. Journal of Clinical Medicine. 2019. Vol. 8:1001

            163. Kim TI, Kwon EB, Oh YC, Go Y, Choi JG. Mori ramulus and its Major Component Morusin Inhibit Herpes Simplex Virus Type 1 Replication and the Virus-Induced Reactive Oxygen Species. American Journal of Chinese Medicine. 2021. Vol. 49:163–179

            164. Li W, Wang XH, Luo Z, Liu LF, Yan C, Yan CY, et al.. Traditional Chinese Medicine as a Potential Source for HSV-1 Therapy by Acting on Virus or the Susceptibility of Host. International Journal of Molecular Science. 2018. Vol. 19:3266

            165. Fatahzadeh M, Schwartz RA. Human Herpes Simplex Virus Infections: Epidemiology, Pathogenesis, Symptomatology, Diagnosis, and Management. Journal of the American Academy of Dermatology. 2007. Vol. 57:737–763

            166. Tzeng NS, Chung CH, Lin FH, Chiang CP, Yeh CB, Huang SY, et al.. Anti-herpetic Medications and Reduced Risk of Dementia in Patients with Herpes Simplex Virus Infections-a Nationwide, Population-based Cohort Study in Taiwan. Neurotherapeutics. 2018. Vol. 15:417–429

            167. Singh A, Kukreti R, Saso L, Kukreti S. Oxidative Stress: A Key Modulator in Neurodegenerative Diseases. Molecules. 2019. Vol. 24:1583

            Author and article information

            Journal
            amm
            Acta Materia Medica
            Compuscript (Ireland )
            2737-7946
            30 December 2022
            : 1
            : 4
            : 534-551
            Affiliations
            [a ]Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou 510632, China
            [b ]The Sixth Affiliated Hospital of Jinan University, Jinan University, Dongguan 523573, China
            [c ]International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 510632, China
            [d ]Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
            [e ]The First Affiliated Hospital of Jinan University, Guangzhou 510632, China
            [f ]School of Chinese Medicine, Jinan University, Guangzhou 510632, China
            [g ]School of Chinese Materia Medica and Yunnan Key Laboratory of Southern Medicinal Utilization, Yunnan University of Chinese Medicine, Kunming 650500, China
            Author notes

            1These authors contributed equally to this work.

            Article
            10.15212/AMM-2022-0043
            f5a0dd34-61d0-4174-9c7a-f0fac11f36bc
            Copyright © 2022 The Authors.

            Creative Commons Attribution 4.0 International License

            History
            : 26 October 2022
            : 13 December 2022
            : 13 December 2022
            Page count
            Figures: 3, Tables: 1, References: 167, Pages: 18
            Categories
            Review Article

            Toxicology,Pathology,Biochemistry,Clinical chemistry,Pharmaceutical chemistry,Pharmacology & Pharmaceutical medicine
            traditional Chinese medicine,immunity,psychological stress,neuroendocrine system,herpes simplex virus type 1,neuroinflammation,latency,reactivation,neurodegenerative diseases

            Comments

            Comment on this article